Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
J Nucl Med ; 51(4): 624-31, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20351352

ABSTRACT

UNLABELLED: A significant antitumor effect was previously observed with radioimmunotherapy using anti-carcinoembryonic antigen (131)I-F6 monoclonal antibody in medullary thyroid cancer-bearing nude mice. Nevertheless, no complete response was observed. As seen with chemotherapy, drugs targeting the tumor microenvironment might improve radioimmunotherapy efficacy. This study evaluated the toxicity and efficacy of combining radioimmunotherapy with thalidomide or a cyclopeptidic vascular endothelial growth inhibitor (CBOP11) in mice grafted with the TT human medullary thyroid cancer cell line. METHODS: Six to 10 nude mice treated with 92.5 MBq of (131)I-F6 in association with 200 mg/kg/d of oral thalidomide during 20 d by force-feeding or 0.45 mg/kg/d of CBOP11 during 25 d using subcutaneous minipumps were compared with control mice receiving either treatment or naked F6 or nonspecific (131)I-734. Combined therapies included (131)I-F6 at day 0 followed by thalidomide between days 20 and 40, thalidomide between days 0 and 20 followed by (131)I-F6 at day 25, (131)I-F6 at day 0 and CBOP11 between days 0 and 25, CBOP11 between days 0 and 25 followed by (131)I-F6 at day 25, and (131)I-F6 at day 0 followed by CBOP11 between days 20 and 45. Animal weight, hematologic toxicity, tumor volume, and serum calcitonin were monitored for the following 3 mo. Improvement of (125)I-F6 tumor biodistribution by antiangiogenic drug was studied after pretreatment by thalidomide. Follow-up of the tumor after combined antiangiogenic and radioimmunotherapy therapies was performed by histology studies. RESULTS: Combined associations, as compared with radioimmunotherapy alone, increased leukopenia but not thrombocytopenia. Tumor volume-quadrupling time (TVQT) was 22.8 +/- 3.3 d in the control group, 29.9 +/- 3.6 d in the group treated with thalidomide, 34.6 +/- 4.4 d in the group treated with CBOP11, and 51.0 +/- 2.8 d after radioimmunotherapy alone. As compared with radioimmunotherapy, TVQT was significantly longer (P < 0.01) after thalidomide followed by radioimmunotherapy (69.83 +/- 3.9), CBOP11 followed by radioimmunotherapy (71.3 +/- 6.1), and CBOP11-radioimmunotherapy in concomitance (64.2 +/- 6.1). Nevertheless, TVQT was not increased after radioimmunotherapy followed by thalidomide (48.8 +/- 4) and radioimmunotherapy followed by CBOP11 (56.8 +/- 4.8). Surprisingly, pretreatment by CBOP11 or thalidomide sensitized larger tumors (>300 mm(3)) to radioimmunotherapy. Change in calcitonin levels confirmed morphologic tumor response. Tumor uptake 24 h after injection of (125)I-F6 was 4.5 +/- 0.6 percentage injected dose per gram (%ID/g) without pretreatment and 8.7 +/- 1.3 %ID/g with pretreatment by thalidomide. An increase of the antitumor effect observed using the antiangiogenic drug combined with radioimmunotherapy was correlated with a decrease of blood vessels shown by von Willebrand immunostaining. CONCLUSION: Pretreatment with antiangiogenic therapies improved radioimmunotherapy efficacy, with acceptable toxicity. Future investigations will be performed to understand how antiangiogenic agents sensitize large tumors to radioimmunotherapy.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Carcinoembryonic Antigen/metabolism , Gene Expression Regulation, Neoplastic , Radioimmunotherapy , Thyroid Neoplasms/therapy , Xenograft Model Antitumor Assays , Angiogenesis Inhibitors/immunology , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/therapeutic use , Apoptosis/drug effects , Carcinoembryonic Antigen/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Combined Modality Therapy , Endothelial Cells/drug effects , Endothelial Cells/pathology , Endothelial Growth Factors/pharmacokinetics , Endothelial Growth Factors/pharmacology , Endothelial Growth Factors/therapeutic use , Humans , Iodine Radioisotopes/chemistry , Mice , Peptides, Cyclic/pharmacokinetics , Peptides, Cyclic/pharmacology , Peptides, Cyclic/therapeutic use , Thalidomide/pharmacology , Thalidomide/therapeutic use , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Tissue Distribution , Treatment Outcome , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism
2.
J Neuroinflammation ; 5: 18, 2008 May 20.
Article in English | MEDLINE | ID: mdl-18492281

ABSTRACT

BACKGROUND: Excitotoxic brain insult is associated with extensive neuronal damage but could also cause inflammatory reactivity and vascular remodeling. The effects of the vascular endothelial growth factor (VEGF) inhibitor, Cyclo-VEGI on expression of VEGF, microgliosis and astrogliosis, blood-brain barrier (BBB) integrity and neuronal viability have been studied following intra-striatal injection of the excitotoxin, quinolinic acid (QUIN). The purpose of this study was to examine VEGF-dependent inflammatory responses in excitotoxin-injected brain and their dependence on pharmacological antagonism of VEGF receptors. METHODS: Single and double immunofluorescence staining of cellular (microglia, astrocyte, neuron) responses and dye and protein infiltration of blood-brain barrier have been applied in the absence, and presence, of pharmacological modulation using a VEGF receptor antagonist, Cyclo-VEGI. Dunn-Bonferroni statistical analysis was used to measure for significance between animal groups. RESULTS: Detailed analysis, at a single time point of 1 d post-QUIN injection, showed excitotoxin-injected striatum to exhibit marked increases in microgliosis (ED1 marker), astrogliosis (GFAP marker) and VEGF expression, compared with PBS injection. Single and double immunostaining demonstrated significant effects of Cyclo-VEGI treatment of QUIN-injected striatum to inhibit microgliosis (by 38%), ED1/VEGF (by 42%) and VEGF striatal immunoreactivity (by 43%); astrogliosis and GFAP/VEGF were not significantly altered with Cyclo-VEGI treatment. Leakiness of BBB was indicated by infiltration of Evans blue dye and plasma protein fibrinogen into QUIN-injected striatum with barrier permeability restored by 62% (Evans blue permeability) and 49% (fibrinogen permeability) with Cyclo-VEGI application. QUIN-induced toxicity was demonstrated with loss of striatal neurons (NeuN marker) and increased neuronal damage (Fluoro-Jade marker) with significant neuroprotection conferred by Cyclo-VEGI treatment (33% increase in NeuN and 38% decrease in Fluoro-Jade). CONCLUSION: An antagonist for VEGF receptor-mediated signaling, Cyclo-VEGI, has shown efficacy in a broad spectrum of activity against striatal excitotoxic insult including inhibition of microgliosis, reduction in leakiness of BBB and parenchymal infiltration of plasma fibrinogen and in conferring significant protection for striatal neurons. Antagonism of VEGF-mediated activity, possibly targeting VEGF receptors on reactive microglia, is suggested as a neuroprotective mechanism against inflammatory reactivity and a novel strategy to attenuate acute excitotoxic damage.


Subject(s)
Blood-Brain Barrier/drug effects , Corpus Striatum/drug effects , Endothelial Growth Factors/therapeutic use , Gliosis/drug therapy , Neuroprotective Agents/therapeutic use , Neurotoxins/toxicity , Peptides, Cyclic/therapeutic use , Quinolinic Acid/toxicity , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Animals , Astrocytes/drug effects , Astrocytes/pathology , Corpus Striatum/physiopathology , Drug Evaluation, Preclinical , Endothelial Growth Factors/pharmacokinetics , Endothelial Growth Factors/pharmacology , Gliosis/chemically induced , Gliosis/pathology , Gliosis/prevention & control , Injections , Injections, Intraventricular , Male , Microglia/drug effects , Microglia/pathology , Neurons/drug effects , Neurons/pathology , Neuroprotective Agents/pharmacology , Peptides, Cyclic/pharmacokinetics , Peptides, Cyclic/pharmacology , Rats , Rats, Sprague-Dawley
3.
Circulation ; 108(1): 97-103, 2003 Jul 08.
Article in English | MEDLINE | ID: mdl-12821549

ABSTRACT

BACKGROUND: A method for identifying tissue experiencing hypoxic stress due to atherosclerotic vascular disease would be clinically useful. Vascular endothelial growth factor-121 (VEGF121) is an angiogenic protein secreted in response to hypoxia that binds to VEGF receptors overexpressed by ischemic microvasculature. We tested the hypothesis that VEGF receptors could serve as markers for ischemic tissue and hence provide a target for imaging such tissue with radiolabeled human VEGF121. METHODS AND RESULTS: A rabbit model of unilateral hindlimb ischemia was created by femoral artery excision (n=14). Control rabbits (n=5) underwent identical surgery without femoral excision. On postoperative day 10, rabbits were intravenously administered 100 microCi of 111In-labeled recombinant human VEGF121, and biodistribution studies and planar imaging were conducted at 3, 24, and 48 hours. On postmortem gamma counting, there was greater accumulation of 111In-labeled VEGF121 in ischemic than in control tissue (P<0.02). Differential uptake of isotope by ischemic muscle was not seen in rabbits injected with 125I-labeled human serum albumin (n=6). Radioactivity imaged in hindlimb regions of interest was significantly higher in ischemic muscle than in sham-operated and contralateral nonoperated hindlimb at 3 hours (P<0.02). Immunohistochemical staining confirmed upregulation of VEGF receptors in ischemic skeletal muscle. CONCLUSIONS: Identification of the ischemic state via targeted radiolabeling of hypoxia-induced angiogenic receptors is possible. This approach could be useful for monitoring the efficacy of revascularization strategies such as therapeutic angiogenesis.


Subject(s)
Hindlimb/blood supply , Ischemia/diagnostic imaging , Ischemia/physiopathology , Receptors, Vascular Endothelial Growth Factor/metabolism , Animals , Binding, Competitive , Disease Models, Animal , Endothelial Growth Factors/pharmacokinetics , Femoral Artery/physiopathology , Hindlimb/diagnostic imaging , Immunohistochemistry , Indium Radioisotopes , Intercellular Signaling Peptides and Proteins/pharmacokinetics , Ischemia/pathology , Lymphokines/pharmacokinetics , Metabolic Clearance Rate , Muscle, Skeletal/blood supply , Muscle, Skeletal/diagnostic imaging , Muscle, Skeletal/physiopathology , Predictive Value of Tests , Rabbits , Radionuclide Imaging , Scintillation Counting , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
4.
Clin Pharmacol Ther ; 72(1): 20-32, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12152001

ABSTRACT

BACKGROUND: The Vascular Endothelial Growth Factor (VEGF) in Ischemia for Vascular Angiogenesis (VIVA) trial was a double-blind, placebo-controlled, phase II clinical trial designed to evaluate the safety, efficacy, and pharmacokinetics of combined intracoronary and intravenous infusions of recombinant human vascular endothelial growth factor (rhVEGF(165)) for therapeutic angiogenesis. This study describes the use of a mechanism-based model to characterize the nonlinear kinetics observed after intravenous administration of rhVEGF(165). The model predicts that rhVEGF(165) distribution occurs through both saturable binding to high-affinity receptors and reversible interactions with low-affinity binding sites. METHODS: In this trial, rhVEGF(165) was administered to patients with coronary artery disease at a dose rate of 17 or 50 ng/kg/min by means of intracoronary infusion for 20 minutes, followed by three 4-hour intravenous infusions on days 3, 6, and 9. Pharmacokinetic samples and blood pressure measurements were collected at baseline, during infusion, and for 6 hours after infusion. RESULTS: The plasma clearance, steady-state volume of distribution, and terminal half-life after a 4-hour intravenous infusion of rhVEGF(165) at the high dose were 19.1 +/- 5.7 mL/min/kg, 960 +/- 260 mL/kg, and 33.7 +/- 13 minutes, respectively. The duration of hypotension that occurred after rhVEGF(165) administration appeared to be related to the model-predicted VEGF(165) concentration associated with the high-affinity receptor compartment. CONCLUSIONS: This mechanism-based model accurately predicted VEGF concentrations and allowed for the simulation of various rhVEGF(165) dose regimens that may aid in optimization of drug delivery for future clinical trials.


Subject(s)
Endothelial Growth Factors/pharmacology , Endothelial Growth Factors/pharmacokinetics , Hemodynamics/drug effects , Lymphokines/pharmacology , Lymphokines/pharmacokinetics , Adult , Aged , Coronary Disease/physiopathology , Coronary Vessels , Female , Half-Life , Humans , Infusions, Intravenous , Injections, Intravenous , Male , Middle Aged , Models, Biological , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
5.
J Pharm Pharmacol ; 54(7): 897-905, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12162707

ABSTRACT

The objectives of this study were to characterize the pharmacokinetics of vascular endothelial growth factor (VEGF) in poly(lactic-co-glycolic) acid (PLGA) microspheres using a rat model, and to develop a pharmacokinetic model for this controlled release formulation. 14C-VEGF was encapsulated using a solid-in-oil-in-water emulsification method. The microspheres were administered subcutaneously to rats and the pharmacokinetic parameters were compared with those of protein solutions. Intravenous administration of protein solutions resulted in short half-lives and subcutaneous administration resulted in rapid clearance from the subcutaneous tissue, with high plasma concentrations as expressed by rapid absorption and elimination. The subcutaneous administration of the VEGF microspheres produced low plasma concentrations and high subcutaneous concentrations over a period of 7 weeks. The area under the curve (AUC), the time required to achieve the maximum concentration (tmax), the maximum concentration (Cmax) in blood samples and the elimination rate constant (kel) values at the subcutaneous tissue site were selected to compare the pharmacokinetic characterization of VEGF microspheres with that of protein solutions. The in-vivo release profiles of the proteins were slower than the in-vitro release profiles and they followed the same trend as the in-vitro and in-vivo PLGA degradation rates. The PLGA microsphere degradation was the determinant step for VEGF release from the microspheres and its absorption at the subcutaneous site. Microspheres appear to be an attractive system for the localized rate-controlled delivery of VEGF. 14C-Methylation via reductive alkylation of VEGF did not affect its mitogenic activity, however approximately 25% activity was lost following release from PLGA microspheres. This loss of activity may be due to degradation in an acidic environment as a result of PLGA degradation.


Subject(s)
Endothelial Growth Factors/administration & dosage , Endothelial Growth Factors/pharmacokinetics , Intercellular Signaling Peptides and Proteins/administration & dosage , Intercellular Signaling Peptides and Proteins/pharmacokinetics , Lymphokines/administration & dosage , Lymphokines/pharmacokinetics , Animals , Carbon Radioisotopes , Delayed-Action Preparations , Injections, Subcutaneous , Male , Microspheres , Polyglactin 910 , Protein Isoforms , Rats , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
6.
Am J Physiol Gastrointest Liver Physiol ; 282(6): G1088-96, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12016135

ABSTRACT

Normal human colonic microvascular endothelial cells (HUCMEC) have been isolated from surgical specimens by their adherence to Ulex europaeus agglutinin bound to magnetic dynabeads that bind alpha-L-fucosyl residues on the endothelial cell membrane. Immunocytochemistry demonstrated the presence of a range of endothelial-specific markers on HUCMEC, including the von Willebrand factor, Ulex europaeus agglutinin, and platelet endothelial cell adhesion molecule-1. The growing cells form monolayers with the characteristic cobblestone morphology of endothelial cells and eventually form tube-like structures. HUCMEC produce vascular endothelial growth factor (VEGF) and express the receptors, kinase insert domain-containing receptor (KDR) and fms-like tyrosine kinase, through which VEGF mediates its actions in the endothelium. VEGF induces the tyrosine phosphorylation of KDR and a proliferative response from HUCMEC comparable to that elicited from human umbilical vein endothelial cells (HUVEC). On binding to HUCMEC or HUVEC, (125)I-labeled VEGF internalizes or dissociates to the medium. Once internalized, (125)I-labeled VEGF is degraded and no evidence of ligand recycling was observed. However, significantly less VEGF is internalized, and more is released to the medium from HUCMEC than HUVEC. Angiogenesis results from the proliferation and migration of microvascular, not large-vessel, endothelial cells. The demonstration that microvascular endothelial cells degrade less and release more VEGF to the medium than large-vessel endothelial cells identifies a mechanism permissive of the role of microvascular cells in angiogenesis.


Subject(s)
Colon/blood supply , Endocytosis/physiology , Endothelial Growth Factors/pharmacokinetics , Endothelium, Vascular/metabolism , Lymphokines/pharmacokinetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Autocrine Communication/physiology , Cells, Cultured , Endothelial Growth Factors/genetics , Endothelium, Vascular/cytology , Gene Expression/physiology , Humans , Iodine Radioisotopes , Lymphokines/genetics , Microcirculation/physiology , Neovascularization, Physiologic/physiology , Paracrine Communication/physiology , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor , Umbilical Veins/cytology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
7.
Bioconjug Chem ; 13(3): 462-7, 2002.
Article in English | MEDLINE | ID: mdl-12009934

ABSTRACT

Targeted drug delivery by cell-specific cytokines and antibodies promises greater drug efficacy and reduced side effects. We describe a novel strategy for assembly of drug delivery vehicles that does not require chemical modification of targeting proteins. The strategy relies on a noncovalent binding of standardized "payload" modules to targeting proteins expressed with a "docking" tag. The payload modules are constructed by linking drug carriers to an adapter protein capable of binding to a docking tag. Using fragments of bovine ribonuclease A as an adapter protein and a docking tag, we have constructed vascular endothelial growth factor (VEGF) based vehicles for gene delivery and for liposome delivery. Assembled vehicles displayed remarkable selectivity in drug delivery to cells overexpressing VEGF receptors. We expect that our strategy can be employed for targeted delivery of many therapeutic or imaging agents by different recombinant targeting proteins.


Subject(s)
Drug Delivery Systems , Endothelial Growth Factors/administration & dosage , Intercellular Signaling Peptides and Proteins/administration & dosage , Lymphokines/administration & dosage , Animals , Cattle , Cells, Cultured , Endothelial Growth Factors/pharmacokinetics , Genetic Therapy , Humans , Intercellular Signaling Peptides and Proteins/pharmacokinetics , Liposomes , Lymphokines/pharmacokinetics , Peptide Fragments/metabolism , Plasmids , Polyethyleneimine/chemistry , Polyethyleneimine/metabolism , Receptors, Vascular Endothelial Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor/metabolism , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/pharmacokinetics , Ribonucleases/metabolism , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
8.
J Pharm Sci ; 91(4): 1065-74, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11948545

ABSTRACT

Semisolid, self-catalyzed poly(ortho ester)s (POEs), are investigated as potential sustained-release systems for proteins. In this study, some factors influencing protein release kinetics and protein instability were evaluated. As model proteins, lysozyme, alpha-lactalbumin, bovine serum albumin, and vascular endothelial growth factor, which were lyophilized from various buffer solutions in the absence and presence of lyoprotectants, were used. For all protein formulations, the release kinetics followed the visually observed polymer dissolution profile. In the absence of any buffers in the protein formulation, the release was continuous. Formulations containing a buffer below pH 7 accelerated POE degradation, resulting in faster protein release. In contrast, a strong buffer capacity at pH 7 reduced the POE degradation and resulted in a biphasic release pattern. Moreover, proteins with a high isoelectric point (pI > 7) appeared to catalyze the POE degradation, and the effect of the buffer strength and pH was much smaller than for proteins with low pI (< 7). In the absence of lyoprotectants, all proteins tested showed an increasing fraction of covalent protein aggregates during the release. Protein formulations containing a lyoprotectant, such as sucrose or trehalose, did not show a significantly increased aggregation, whereas there was a minor influence of the large solid loadings on the release kinetics. In conclusion, this semisolid, self-catalyzed POE showed good promise as a sustained-release matrix for bioactive proteins.


Subject(s)
Polymers/pharmacokinetics , Proteins/pharmacokinetics , Animals , Biocompatible Materials/pharmacokinetics , Delayed-Action Preparations/pharmacokinetics , Endothelial Growth Factors/pharmacokinetics , Intestinal Mucosa/metabolism , Lactalbumin/pharmacokinetics , Lymphokines/pharmacokinetics , Muramidase/pharmacokinetics , Serum Albumin, Bovine/pharmacokinetics , Swine , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
9.
Hypertension ; 39(3): 815-20, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11897770

ABSTRACT

Vascular endothelial growth factor (VEGF) induces hypotension in normotensive subjects, which is considered to be a major side effect for treatment of ischemic diseases. However, the hypotensive effect of VEGF has not been investigated in the setting of hypertension. This study determined effects of VEGF on hemodynamics, pharmacokinetics, and release of NO and prostaglandin I2 (PGI2) in vivo and on vasorelaxation of mesentery artery rings in vitro in spontaneously hypertensive rats (SHR) compared with Wistar-Kyoto rats (WKY). Intravenous infusion of VEGF for 2 hours produced a dose-related decrease in arterial pressure, which was enhanced in conscious SHR compared with WKY (P<0.01), and an increase in heart rate in WKY but not in SHR. In response to similar doses of VEGF, compared with WKY, SHR had a higher plasma VEGF level and lower VEGF clearance (P<0.01). Circulating NO and PGI2 levels after VEGF administration were not increased in SHR versus WKY, and VEGF-induced vasorelaxation was blunted in SHR versus WKY in vitro, suggesting endothelial dysfunction in SHR. One-week VEGF infusion also caused greater hypotension (P<0.05) in the absence of tachycardia in SHR compared with WKY controls. Thus, despite blunted vasorelaxation in vitro because of endothelial dysfunction, SHR exhibited exaggerated hypotension without tachycardia in response to VEGF, which was independent of NO and PGI2. The exaggerated hypotensive response to VEGF in SHR may be owing to impaired baroreflex function and reduced VEGF clearance. The data may also suggest that more caution should be taken when VEGF is administered in patients with hypertension.


Subject(s)
Antihypertensive Agents/pharmacology , Endothelial Growth Factors/pharmacology , Hypertension/physiopathology , Lymphokines/pharmacology , Animals , Antihypertensive Agents/pharmacokinetics , Blood Pressure/drug effects , Dose-Response Relationship, Drug , Endothelial Growth Factors/pharmacokinetics , Epoprostenol/blood , Heart Rate/drug effects , Hypertension/drug therapy , Hypertension/metabolism , In Vitro Techniques , Lymphokines/pharmacokinetics , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/physiology , Nitric Oxide/blood , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Vasodilation/drug effects
10.
NMR Biomed ; 15(2): 120-31, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11870908

ABSTRACT

Vascular endothelial growth factor (VEGF) is one of the key growth factors regulating tumor angiogenesis and thus it is one of the primary targets for antiangiogenic therapy. The long-term effects of VEGF include induction of proliferation and migration of endothelial cells, tube formation and maintenance of the immature capillaries. The early effects of VEGF include vasodilation and increased permeability. We hypothesize that the early responses to VEGF can serve to develop a quantitative measure of the activity of VEGF, and therefore may be applicable for monitoring the efficacy of systemic suppression of VEGF signaling during antiangiogenic therapy. For that end we tested the ability of MRI and fluorescence microscopy to detect the early response to intradermal VEGF165 in nude mice. VEGF-induced local vasodilation and increased permeability was detected by intravenous administration of macromolecular biotin-BSA-GdDTPA(23) 30 min after intradermal administration of VEGF. Contrast leak showed saturation kinetics. Delayed contrast administration (90 min after intradermal administration of VEGF) resulted in low contrast leak and demonstrated that the saturation kinetics is not due to contrast equilibration between plasma and the interstitial space, but rather is due to suppression of vascular permeability. Permeability was restored by a second bolus of VEGF, showing that the saturation kinetics is primarily due to inactivation of the growth factor. Confocal microscopy of fluorescent BSA-FITC confirmed the permeability changes monitored by MRI. Moreover, confocal microscopy showed efficient lymphatic uptake of the extravasated contrast material specifically in regions of VEGF induced hyper-permeability.


Subject(s)
Endothelial Growth Factors/therapeutic use , Lymphokines/therapeutic use , Animals , Contrast Media , Endothelial Growth Factors/pharmacokinetics , Gadolinium DTPA/pharmacokinetics , Lymphokines/pharmacokinetics , Magnetic Resonance Imaging/methods , Male , Metabolic Clearance Rate , Mice , Mice, Nude , Microscopy, Fluorescence/methods , Protein Isoforms/pharmacokinetics , Protein Isoforms/therapeutic use , Tissue Distribution , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Vasodilation
11.
J Biomed Mater Res ; 59(3): 422-8, 2002 Mar 05.
Article in English | MEDLINE | ID: mdl-11774299

ABSTRACT

The present investigation was performed to study the adsorption behavior of growth factors and their release characteristics from biodegradable implants in an in vitro study. We investigated the stability of growth factors administered on various scaffolds. We used porous tricalcium phosphate ceramics (alpha-TCP), a neutralized glass-ceramics (GB9N), a composite (polylactid/-glycolid/GB9N), and solvent dehydrated human bone as carriers. Block shaped scaffolds (sized: 7 x 7 x 10 mm) were loaded with 5 microg of either bone morphogenetic protein (rxBMP-4), basic fibroblast growth factor (rh-bFGF), or vascular endothelial growth factor (rh-VEGF) solved in 150 microL PBS. The growth factors were labeled with Iodine125 (I-125) for detecting the adsorbed and released amount of growth factors by counting the samples for total I-125 activity. We observed that the adsorption of these growth factors seems to depend on two different parameters: first on the nature of the tested material, and second on the growth factors on their own. The release kinetics of the growth factors from the biodegradable implants can be described as a two phase process-a very rapid release during the first hours by an elution of not adsorbed protein, followed by a specific release, which depends upon the chemical/physical interaction of the material and the growth factor used. Analyzing the eluted proteins on SDS-PAGEs rh-VEGF was degraded into a smaller fragment with a size of around 15 kDa, while rxBMP-4 and rh-bFGF showed a complete degradation into fragments smaller than 3 kDa after more than 3 days. Although this in vitro study suggests that biodegradable implants might be successfully used as carriers for osteogenic growth factors, the different release kinetics as well as the alteration of their molecular structure including loss of biological activity should be considered.


Subject(s)
Absorbable Implants/standards , Growth Substances/pharmacokinetics , Adsorption , Animals , Bone Morphogenetic Proteins/pharmacokinetics , Bone and Bones , Calcium Phosphates , Ceramics , Composite Resins , Drug Carriers/chemistry , Drug Carriers/standards , Drug Stability , Endothelial Growth Factors/pharmacokinetics , Fibroblast Growth Factor 2/pharmacokinetics , Humans , Intercellular Signaling Peptides and Proteins/pharmacokinetics , Kinetics , Lymphokines/pharmacokinetics , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Xenopus laevis
12.
Curr Opin Investig Drugs ; 2(6): 792-5, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11572658

ABSTRACT

GenVec, in collaboration with Pfizer (formerly Parke-Davis), is developing AdGVVEGF121.10 (BioBypass), a gene therapy involving the 121-residue isoform of vascular endothelial growth factor (VEGF), licensed from Scios, for the potential treatment of coronary artery disease (CAD) and peripheral vascular disease (PVD) [262000]. By March 2000, phase II trials in CAD had commenced [359531], [359532], [359538]. By August 2000, phase II trials were also underway for PVD [386293]. The initial phase II trial will include approximately 70 patients with severe CAD who are not candidates for bypass surgery and will assess exercise capacity and patient well-being, before and after treatment, as well as safety and drug tolerance [364137]. Scios licensed the gene transfer applications of VEGF121 to GenVec in June 1996 [263381]. In September 1997, GenVec entered into an agreement with Parke-Davis, a subsidiary of Warner-Lambert (now Pfizer), to develop the therapy [262000]. In May 1999, Warner-Lambert signed an agreement with Bioscience for a device for the administration of AdGVVEGF121.10 1325443]. In May 2000, Merrill Lynch predicted a US filing in the first half of 2003 [375962]. In January 2001, AG Edwards predicted the product would generate $70 million in revenues to Pfizer and $12 million in royalties to GenVec in 2005. In February 1999, GenVec was awarded US-05846782, covering vectors for targeting the transfer of therapeutic genes to specific tissues in the human body [316038].


Subject(s)
Angiogenic Proteins , Endothelial Growth Factors/pharmacology , Genetic Therapy , Lymphokines/pharmacology , Vascular Diseases/therapy , Animals , Clinical Trials as Topic , Endothelial Growth Factors/adverse effects , Endothelial Growth Factors/pharmacokinetics , Humans , Lymphokines/adverse effects , Lymphokines/pharmacokinetics , Structure-Activity Relationship
13.
J Control Release ; 74(1-3): 341-7, 2001 Jul 06.
Article in English | MEDLINE | ID: mdl-11489516

ABSTRACT

Selective targeting of therapeutic and diagnostic agents improves their efficacy and minimizes potentially adverse side effects. Existing methods for selective targeting are based on chemical conjugation of therapeutics and diagnostics, or their carriers, to cell-specific targeting molecules (e.g., growth factors, antibodies). These methods are limited by potential damage to targeting molecules that can be inflicted by the conjugation procedure. In addition, conjugation procedures have to be developed on a case-by-case basis. In order to avoid these problems we have developed a new approach to constructing molecular vehicles for target-mediated delivery of therapeutics and diagnostics. In this approach, the targeting molecule is expressed as a fusion protein containing a recognition tag. The recognition tag is defined as a peptide or protein that can bind non-covalently another peptide or protein (adapter). In turn, the adapter is chemically conjugated to a carrier of therapeutics or diagnostics. The assembled molecular delivery vehicle contains a carrier-adapter conjugate bound non-covalently to a recognition tag fused to the targeting protein. The advantages of this technology are: (i) no chemical modification of targeting molecules, and (ii) universal, 'off-the-shelf' carrier-adapter constructs that can be combined with different fusion targeting proteins. To obtain a proof-of-principle we have constructed VEGF fusion proteins containing a 15-aa S-peptide fragment of RNase A as a recognition tag. Using the S-protein fragment of RNase A as an adapter and polyethylenimine as a DNA carrier we have achieved selective gene delivery to cells overexpressing VEGFR-2.


Subject(s)
Drug Delivery Systems , Endothelial Growth Factors/administration & dosage , Lymphokines/administration & dosage , Neoplasms/diagnosis , Neoplasms/therapy , Animals , Endothelial Growth Factors/pharmacokinetics , Genetic Therapy , Humans , Luciferases/biosynthesis , Luciferases/genetics , Lymphokines/pharmacokinetics , Polyethyleneimine , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/genetics , Receptors, Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/pharmacokinetics , Ribonuclease, Pancreatic/metabolism , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
14.
J Intern Med ; 250(5): 373-81, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11887971

ABSTRACT

OBJECTIVE: To test the safety and bioactivity of phVEGF-A165 after intramyocardial injection during 12-month follow-up. DESIGN: Open-labelled study. SUBJECTS: Inclusion criteria were angina pectoris, Canadian Cardiovascular Society (CCS) class III-IV, unamenable to further revascularization, ejection fraction (EF) >30%, perfusion defects extending over >10% of the anterolateral left ventricle wall detectable with adenosine single photon emission computerized tomography (SPECT) and at least one patent vessel visible by coronary angiography. Seven of 39 patients referred for gene therapy were included. INTERVENTION: Via a mini-thoracotomy under general anaesthesia. phVEGF-A165 was injected directly into the myocardium at four sites in the anterolateral region of the left ventricle. RESULTS: Operative procedures were uneventful. Perioperative release of myocardial markers and electrocardiogram (ECG) changes were detected in two patients. There were no perioperative deaths but one patient died 7 months postoperatively because of myocardial infarction. Plasma vascular endothelial growth factor (VEGF)-A levels increased two to threefold peaking 6 days postoperatively (P < 0.004) and returning to baseline by day 30. A significant reduction in angina pectoris was reported. The CCS class improved from 3.3+/-0.2 to 1.9+/-0.3 (P < 0.01) and nitroglycerine intake decreased from 39+/-15 to 12+/-5 tablets week(-1) (P < 0.001) 2 months after gene transfer. Improvements remained after 12 months when nitroglycerine consumption approached zero. Improved myocardial function in the phVEGF-A165 injection region was documented in all patients (P < 0.016) by tissue velocity imaging (TVI). Reduced reversible ischaemia was detected by adenosine SPECT in four patients. Improved collateralization was detected in four patients with coronary angiography. CONCLUSION: Intramyocardial injection of phVEGF-A165 is safe and may lead to improved myocardial perfusion and function with longstanding symptomatic relief in end-stage angina pectoris. Based on these results this therapeutic potential is being tested in a double-blind placebo controlled multicentre trial.


Subject(s)
Angina Pectoris/drug therapy , Coronary Artery Disease/drug therapy , Endothelial Growth Factors/administration & dosage , Genetic Therapy , Plasmids/administration & dosage , Aged , Angina Pectoris/diagnosis , Angina Pectoris/surgery , Coronary Artery Disease/diagnosis , Coronary Artery Disease/surgery , Endothelial Growth Factors/pharmacokinetics , Endothelial Growth Factors/therapeutic use , Female , Follow-Up Studies , Gene Transfer Techniques , Humans , Injections, Intramuscular , Male , Middle Aged , Plasmids/pharmacokinetics , Plasmids/therapeutic use , Thoracotomy , Time Factors , Vascular Endothelial Growth Factor A
15.
Invest Ophthalmol Vis Sci ; 41(6): 1561-9, 2000 May.
Article in English | MEDLINE | ID: mdl-10798677

ABSTRACT

PURPOSE: To determine whether sustained elevation of vascular endothelial growth factor (VEGF) in the vitreous cavity causes retinal hyperpermeability [blood-retinal barrier (BRB) breakdown] before the development of retinal neovascularization (NV) and to document the kinetics of the integrity of BRB breakdown versus time. METHODS: Poly(L-lactide-co-glycolide)based devices loaded with VEGF were implanted intravitreally in rabbit eyes. Contrast-enhanced magnetic resonance imaging (MRI) methods were used to identify and quantitate the retinal permeability at various time points after implantation. This was done with the newly developed MR tracer AngioMARK (Epix Medical, Boston, MA). After the MRI measurements, fundus photography and fluorescein angiography (FA) also were performed on the same set of animals. RESULTS: At 3 days after implantation, the MR images showed a significant retinal leakage into the vitreous cavity (BRB breakdown) of the VEGF-implanted eyes. To quantitate this leakage, the permeability surface area product (PS) was measured. At 3 days, the mean PS product was 1.25 +/-0.25 x 10(-5) cm3/min. Based on the VEGF in vitro release study, this 3-day BRB breakdown corresponded to a total sustained release of 7.42 +/- 0.54 microg/ml of VEGF. The fundus and FA photographs of these VEGF-implanted eyes taken at 4 days after implantation also showed a considerable level of retinal vascular dilation and tortuosity. By 12 days after implantation, the mean PS product decreased to 5.83 +/- 1.38 x 10(-6) cm3/min. However, the retinal NV was observed only after the second week after implantation. By this time, a total of 10.70 +/- 0.92 microg/ml of VEGF was released in a sustained fashion. Also, after the retinal NV development, retinal detachment also was observed. The control eyes, however, which were implanted with blank devices, remained unchanged and normal during the entire course of this study (PS = 5.57 +/- 0.66 x 10(-7) cm3/min). CONCLUSIONS. The findings indicate that sustained delivery of elevated amounts of VEGF in the vitreous cavity induces a BRB breakdown even earlier than 3 days after implantation. This was achieved after a total sustained release of 7.42 +/- 0.54 microg/ml of VEGF. This retinal leakage regressed by more than half by the time the retinal NV developed. Furthermore, a retinal detachment occurred after this retinal NV. These results are similar to proliferative diabetic retinopathy (PDR). The sustained elevation of VEGF in the vitreous cavity of rabbit eyes is potentially a good model to test VEGF antagonists to treat or prevent PDR in humans. The quantifiable change of BRB breakdown by the contrast-enhanced MRI method is ideal to assess the therapeutic intervention in vivo without killing the animal and may prove to be clinically useful in humans.


Subject(s)
Blood-Retinal Barrier , Endothelial Growth Factors/pharmacokinetics , Lymphokines/pharmacokinetics , Retina/metabolism , Retinal Neovascularization/metabolism , Retinal Vessels/metabolism , Vitreous Body/metabolism , Animals , Contrast Media , Delayed-Action Preparations , Drug Carriers , Fluorescein Angiography , Fundus Oculi , Lactic Acid , Magnetic Resonance Imaging , Male , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , Polymers , Rabbits , Retina/pathology , Retinal Neovascularization/chemically induced , Retinal Neovascularization/diagnosis , Retinal Vessels/pathology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
16.
J Control Release ; 64(1-3): 91-102, 2000 Feb 14.
Article in English | MEDLINE | ID: mdl-10640648

ABSTRACT

Engineering new tissues utilizing cell transplantation on biodegradable polymer matrices is an attractive approach to treat patients suffering from the loss or dysfunction of a number of tissues and organs. The matrices must maintain structural integrity during the process of tissue formation, and promote the vascularization of the developing tissue. A number of molecules (angiogenic factors) have been identified that promote the formation of new vascular beds from endothelial cells present within tissues, and the localized, controlled delivery of these factors from a matrix may allow an enhanced vascularization of engineered tissues. We have developed a gas foaming polymer processing approach that allows the fabrication of three-dimensional porous matrices from bioabsorbable materials (e.g., copolymers of lactide and glycolide [PLG]) without the use of organic solvents or high temperatures. The effects of several processing parameters (e.g., gas type, polymer composition and molecular weight) on the process were studied. Several gases (CO(2), N(2), He) were utilized in the fabrication process, but only CO(2) resulted in the formation of highly porous, structurally intact matrices. Crystalline polymers (polylactide and polyglycolide) did not form porous matrices, while amorphous copolymers (50:50, 75:25, and 85:15 ratio of lactide:glycolide) foamed to yield matrices with porosity up to 95%. The mechanical properties of matrices were also regulated by the choice of PLG composition and molecular weight. Angiogenic factors (e.g., vascular endothelial growth factor) were subsequently incorporated into matrices during the fabrication process, and released in a controlled manner. Importantly, the released growth factor retains over 90% of its bioactivity. In summary, a promising system for the incorporation and delivery of angiogenic factors from three-dimensional, biodegradable polymer matrices has been developed, and the fabrication process allows incorporation under mild conditions.


Subject(s)
Biomedical Engineering , Drug Delivery Systems/methods , Endothelial Growth Factors/pharmacokinetics , Growth Substances/administration & dosage , Lymphokines/pharmacokinetics , Polymers/metabolism , Absorption , Carbon Dioxide/chemistry , Delayed-Action Preparations/pharmacokinetics , Drug Stability , Glycosides/chemistry , Helium/chemistry , Lactic Acid/chemistry , Nitrogen/chemistry , Porosity , Time Factors , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
17.
J Clin Invest ; 104(7): 913-23, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10510332

ABSTRACT

VEGF(165), the most abundant isoform in man, is an angiogenic cytokine that also regulates vascular permeability. Its function in the renal glomerulus, where it is expressed in visceral epithelial and mesangial cells, is unknown. To assess the role of VEGF(165) in glomerular disease, we administered a novel antagonist - a high-affinity, nuclease-resistant RNA aptamer coupled to 40-kDa polyethylene glycol (PEG) - to normal rats and to rats with mesangioproliferative nephritis, passive Heymann nephritis (PHN), or puromycin aminonucleoside nephrosis (PAN). In normal rats, antagonism of VEGF(165) for 21 days failed to induce glomerular pathology or proteinuria. In rats with mesangioproliferative nephritis, the VEGF(165) aptamer (but not a sequence-scrambled control RNA or PEG alone) led to a reduction of glomerular endothelial regeneration and an increase in endothelial cell death, provoking an 8-fold increase in the frequency of glomerular microaneurysms by day 6. In contrast, early leukocyte influx and the proliferation, activation, and matrix accumulation of mesangial cells were not affected in these rats. In rats with PHN or PAN, administration of the VEGF(165) aptamer did not influence the course of proteinuria using various dosages and administration routes. These data identify VEGF(165) as a factor of central importance for endothelial cell survival and repair in glomerular disease, and point to a potentially novel way to influence the course of glomerular diseases characterized by endothelial cell damage, such as various glomerulonephritides, thrombotic microangiopathies, or renal transplant rejection.


Subject(s)
Endothelial Growth Factors/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Glomerulonephritis, Membranoproliferative/physiopathology , Glomerulonephritis/physiopathology , Kidney Glomerulus/blood supply , Kidney Glomerulus/pathology , Lymphokines/pharmacology , Aneurysm/pathology , Animals , Cell Division/drug effects , Cornea/blood supply , Endothelial Growth Factors/pharmacokinetics , Endothelium, Vascular/drug effects , Glomerulonephritis/pathology , Glomerulonephritis, Membranoproliferative/pathology , Humans , Kidney Glomerulus/drug effects , Lymphokines/pharmacokinetics , Male , Neovascularization, Physiologic/drug effects , Polyethylene Glycols/pharmacology , Protein Isoforms/pharmacokinetics , Protein Isoforms/pharmacology , Proteinuria , Puromycin Aminonucleoside/toxicity , Rats , Rats, Sprague-Dawley , Rats, Wistar , Renal Circulation , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
19.
J Pharmacol Exp Ther ; 284(1): 103-10, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9435167

ABSTRACT

Vascular endothelial growth factor (VEGF) produces beneficial angiogenesis in animal models of coronary and peripheral ischemia. However, intravenous bolus injection of Chinese hamster ovary cell (CHO)-derived VEGF produces adverse effects on hemodynamics. The present study examined pharmacokinetic and hemodynamic responses to Escherichia coli-derived VEGF, which will be used in clinical patients, compared with responses to CHO-derived VEGF, and tested whether intravenous infusion of E. coli-derived VEGF attenuates the hemodynamic responses compared with the responses observed with intravenous bolus injection. Hemodynamic parameters were measured before and after administration of VEGF in conscious, instrumented rats. Intravenous injection of both CHO- and E. coli-derived VEGF produced a similar maximal reduction in arterial pressure, although E. coli-derived VEGF exhibited less of a depressor effect in the initial phase after injection. Either infusion or injection of E. coli-derived VEGF caused hypotension, tachycardia and reduced cardiac output and stroke volume, which were significantly attenuated when given by infusion compared with injection. The maximal hypotensive and tachycardiac responses to infusion were decreased by 50 to 60% compared with those responses observed after injection. Cardiac output was maximally reduced by 34% after injection, but only 18% after infusion. A sustained elevation in systemic vascular resistance observed after injection was avoided after infusion. Thus, the hemodynamic side effects of VEGF administration can be substantially attenuated by controlling the rate of VEGF infusion. The data indicate that infusion, instead of bolus injection, is a more appropriate regimen for VEGF administration.


Subject(s)
Endothelial Growth Factors/pharmacology , Hemodynamics/drug effects , Lymphokines/pharmacology , Animals , CHO Cells , Cricetinae , Endothelial Growth Factors/administration & dosage , Endothelial Growth Factors/pharmacokinetics , Infusions, Intravenous , Injections, Intravenous , Lymphokines/administration & dosage , Lymphokines/pharmacokinetics , Male , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
20.
Br J Rheumatol ; 36(3): 315-21, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9133962

ABSTRACT

The objective was to assess the congruity of gliostatin/platelet-derived endothelial cell growth factor (GLS PD-ECGF) with other clinical markers of rheumatoid arthritis (RA) and to define its molecular mechanism of action in the complicated cytokine network during RA pathogenesis. Immunoassay systems were used to quantify GLS or cytokine levels in laboratory and clinical samples. Expression levels of GLS were determined by reverse transcription-polymerase chain reaction methods. The GLS levels in synovial fluid were correlated with interleukin-1 (IL-1) and IL-8. The serial data of serum GLS levels reflected well changes in the disease activity during the clinical course of four representative patients with RA. In cultured fibroblast-like synoviocytes, tumour necrosis factor-alpha (TNF-alpha), IL-1, IL-6 and IL-8 induced GLS expression. In conclusion, our results suggest that the serum GLS level, mostly derived from cytokine-stimulated synoviocytes, was a useful clinical marker of RA.


Subject(s)
Arthritis, Rheumatoid/diagnosis , Biomarkers/blood , Blood Platelets/chemistry , Endothelial Growth Factors/blood , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/blood , Synovial Fluid/chemistry , Endothelial Growth Factors/pharmacokinetics , Humans , Interleukin-1/analysis , Interleukin-1/pharmacology , Interleukin-6/analysis , Interleukin-6/pharmacology , Interleukin-8/analysis , Interleukin-8/pharmacology , Nerve Tissue Proteins/immunology , Therapeutic Equivalency , Thymidine Phosphorylase , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...