Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 352
Filter
2.
Am Heart J ; 142(5): 872-80, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11685177

ABSTRACT

BACKGROUND: Patients with severe myocardial ischemia who are not candidates for percutaneous or surgical revascularization have few therapeutic options. Therapeutic angiogenesis in animal models with use of recombinant human vascular endothelial growth factor (rhVEGF) has resulted in successful revascularization of ischemic myocardium. This was a dose escalation trial designed to determine the safety and tolerability of intracoronary rhVEGF infusions. METHODS AND RESULTS: Patients were eligible if they had stable exertional angina, a significant reversible perfusion defect by stress myocardial perfusion study, and coronary anatomy that was suboptimal for percutaneous coronary intervention or coronary artery bypass grafting. rhVEGF was administered to a total of 15 patients by 2 sequential (eg, right and left) intracoronary infusions, each for 10 minutes, at rates of 0.005 (n = 4), 0.017 (n = 4), 0.050 (n = 4), and 0.167 mg/kg/min (n = 3). Pharmacokinetic sampling and hemodynamic monitoring were performed for 24 hours. Radionuclide myocardial perfusion imaging was performed before treatment and at 30 and 60 days after treatment. Follow-up angiograms were performed on selected patients at 60 days. The maximally tolerated intracardiac dose of rhVEGF was 0.050 mg/kg/min. Minimal hemodynamic changes were seen at 0.0050 mg/kg/min (2% +/- 7% [SD] mean decrease in systolic blood pressure from baseline to nadir systolic blood pressure), whereas at 0.167 mg/kg/min there was a 28% +/- 7% mean decrease from baseline to nadir (136 to 95 mm Hg systolic). Myocardial perfusion imaging was improved in 7 of 14 patients at 60 days. All 7 patients with follow-up angiograms had improvements in the collateral density score. CONCLUSION: rhVEGF appears well tolerated by coronary infusion at rates up to 0.050 mg/kg/min. This study provides the basis for future clinical trials to assess the clinical benefit of therapeutic angiogenesis with rhVEGF.


Subject(s)
Coronary Disease/drug therapy , Endothelial Growth Factors/administration & dosage , Lymphokines/administration & dosage , Protein Isoforms/administration & dosage , Coronary Circulation/drug effects , Endothelial Growth Factors/pharmacology , Endothelial Growth Factors/therapeutic use , Humans , Lymphokines/pharmacology , Lymphokines/therapeutic use , Neovascularization, Physiologic/drug effects , Protein Isoforms/pharmacology , Protein Isoforms/therapeutic use , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
3.
Rev Esp Cardiol ; 54(10): 1210-24, 2001 Oct.
Article in Spanish | MEDLINE | ID: mdl-11591302

ABSTRACT

Therapeutic angiogenesis based on the administration of growth factors with angiogenic activity allows enhancement of collateral vessels able to palliate insufficient tissue perfusion secondary to obstruction of native arteries. At present, this type of therapy is addressed to patients that fail to respond to conventional treatment (surgical or percutaneous revascularization). The most extensively investigated angiogenic growth factors are vascular endothelial growth factor (VEGF) and fibroblast growth factor (FGF). These cytokines can be administered either as recombinant proteins or as the genes encoding for these proteins. Both approaches have pros and cons that are under investigation in animal models and in clinical studies. Although clinical trials consist so far of small, often non-randomized series, preliminary results are promising. For example, administration of VEGF or FGF has been associated to objective evidence of increased tissue perfusion in patients with myocardial ischemia, and to a significant improvement of pain and ischemia in patients with peripheral arterial disease. Contrarily to expected, these interventions have been associated to scant adverse side effects, although larger clinical trials will be necessary in order to prove the safety and effectiveness of these interventions. Nevertheless, it seems clear that it is feasible to induce effective therapeutic angiogenesis in selected patients without significant associated toxicity.


Subject(s)
Endothelial Growth Factors/therapeutic use , Fibroblast Growth Factors/therapeutic use , Ischemia/therapy , Lymphokines/therapeutic use , Peripheral Vascular Diseases/therapy , Clinical Trials as Topic , Drug Evaluation , Endothelial Growth Factors/adverse effects , Endothelial Growth Factors/genetics , Fibroblast Growth Factors/adverse effects , Fibroblast Growth Factors/genetics , Genetic Therapy/methods , Humans , Ischemia/drug therapy , Leg/blood supply , Lymphokines/adverse effects , Lymphokines/genetics , Myocardial Ischemia/drug therapy , Neovascularization, Physiologic , Secondary Prevention , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
4.
Hum Gene Ther ; 12(12): 1593-4, 2001 Aug 10.
Article in English | MEDLINE | ID: mdl-11529248

ABSTRACT

Among diabetics, peripheral neuropathy is common and ultimately accounts for significant morbidity. The ultimate consequence of such sensory defects involving the lower extremities may be foot ulceration initiated by trauma that is inapparent to the patient. Such ulcerations often lead to lower extremity amputation, a complication that is 15 times higher in diabetic versus non-diabetic patients. Preliminary clinical studies have demonstrated improvement in signs and symptoms of sensory neuropathy in patients with lower extremity vascular occlusive disease following intramuscular injection of naked DNA encoding vascular endothelial growth factor (VEGF). To determine if such a strategy could be applied to diabetic patients, including those without evidence of large vessel occlusive disease, we investigated the hypothesis that experimental diabetic neuropathy results from destruction of the vasa nervorum and can be reversed by administration of an angiogenic growth factor. In two different animal models of diabetics, nerve blood flow and the number of vasa nervorum were found to be markedly attenuated resulting in severe peripheral neuropathy. In contrast, following VEGF gene transfer, vascularity and blood flow in nerves of treated animals were similar to those of non-diabetic controls; constitutive overexpression of VEGF resulted in restoration of large and small fiber peripheral nerve function. These findings implicate microvascular disruption as the basis for diabetic neuropathy and suggest that angiogenic growth factors may constitute a novel treatment strategy for this pernicious disorder. Accordingly, we now seek to address the following two objectives: 1. Objective #1: is to evaluate the safety and impact of phVEGF165 gene transfer on sensory neuropathy in patients with diabetes and associated macrovascular disease involving the lower extremities. 2. Objective #2: is to evaluate the safety and impact of phVEGF165 gene transfer on sensory neuropathy in patients with diabetes without macrovascular disease involving the lower extremities. The protocol outlined in this Investigational New Drug Application has been designed as a Phase I/II, single-site, dose escalating, double-blind, placebo controlled study to evaluate the safety and impact of phVEGF165 gene transfer on sensory neuropathy in patients with diabetes with or without macrovascular disease involving the lower extremities. Diabetic males or females > 21 years old with sensory neuropathy with or without macrovascular disease will be eligible. A total of 192 patients will be recruited into two arms of the study (each arm consisting of 96 patients) over a period of 4 years (the fifth year will be limited to follow-up examinations). The 96 patients in each of the two arms of the study will comprise 3 cohorts, each consisting of 32 patients. Within each of these cohorts, patients will be randomized to receive phVEGF165 or placebo based upon a 3:1 randomization ratio. Thus, at the completion of the study, 24 patients will have each received a given dose (1, 2, or 4 mg phVEGF165) and 24 patients will have received placebo. Doses will be employed in a serial dose-escalating fashion. The entire volume of the study drug will be divided and delivered in 8 intramuscular injections administered into the foot, calf muscle, or distal thigh muscle of the affected extremity. Following the initial set of injections, repeat treatment with an identical dose will be provided 2 and 4 weeks after initial treatment.


Subject(s)
Diabetic Neuropathies/therapy , Endothelial Growth Factors/genetics , Lymphokines/genetics , Adult , Aged , Cohort Studies , Cytomegalovirus/genetics , DNA, Complementary/metabolism , Diabetes Mellitus/genetics , Diabetes Mellitus/therapy , Diabetic Neuropathies/genetics , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Male , Middle Aged , Placebos , Plasmids/metabolism , Promoter Regions, Genetic , Protein Isoforms , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
5.
Circ Res ; 89(5): 389-400, 2001 Aug 31.
Article in English | MEDLINE | ID: mdl-11532899

ABSTRACT

Clinical trials of cardiovascular gene therapy, whether using viral (53%) or nonviral (47%) vectors, have thus far disclosed no evidence indicative of inflammatory or other complications, including death, directly attributable to the vector used. Indeed, despite the fact that initial trials of cardiovascular gene therapy targeted patients with end-stage vascular disease, including critical limb ischemia and refractory myocardial ischemia, the mortality for patients enrolled in clinical trials of cardiovascular gene therapy reported to date compares favorably with mortality for similar groups of patients in contemporary controlled studies of medical or interventional therapies. The most common morbidity reported after cardiovascular gene transfer is lower extremity edema; in contrast to data involving genetically engineered mice, however, evidence of life- or limb-threatening edema has not been described in any patients, including patients after gene transfer for myocardial ischemia. Concerns regarding the potential for angiogenic cytokines to promote the progression of atherosclerosis are not supported by angiographic follow-up of patients with coronary or peripheral vascular disease. The levels and duration of gene expression investigated for therapeutic angiogenesis transfer have been unassociated with hemangioma formation. Likewise, there is little evidence from either preclinical or clinical studies to support the notion that the administration of angiogenic growth factors, per se, is sufficient to stimulate the growth of neoplasms. Patients enrolled in clinical studies of angiogenic cytokines, including patients with diabetes and a previous history of retinopathy, have disclosed no evidence to suggest that ocular pathology is a risk of angiogenic growth factor gene transfer.


Subject(s)
Cardiovascular Diseases/therapy , Genetic Therapy , Cardiovascular Diseases/genetics , Clinical Trials as Topic/statistics & numerical data , Endothelial Growth Factors/genetics , Genetic Vectors/genetics , Humans , Lymphokines/genetics , Risk Assessment , Risk Factors , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
6.
FASEB J ; 15(11): 1877-85, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11532967

ABSTRACT

Formation of new blood vessels in the adult animal (i.e., angiogenesis) is an important event for tissue repair and for tumor growth and metastasis. Angiogenesis involves the migration and proliferation of endothelial cells. We have investigated the role of the growth suppressor p27(Kip1) (p27) on endothelial cell function in vitro and angiogenesis in vivo. We have generated Ad-TetON, a replication-deficient adenovirus that constitutively expresses the reverse tet-responsive transcriptional activator, and Ad-TRE-p27, which drives expression of p27 under the control of the tet response element. Western blot analysis demonstrated doxycycline-dependent overexpression of p27 in human umbilical vein endothelial cells (HUVECs) coinfected with Ad-TetON and Ad-TRE-p27, which resulted in a marked inhibition of DNA replication and cell migration in vitro. Inducible overexpression of p27 in cultured HUVECs inhibited the formation of tubelike structures and, when applied in a murine model of hind limb ischemia, reduced hind limb blood flow recovery and capillary density. These findings thus underscore a novel role of p27 in regulating endothelial cell migration in vitro and angiogenesis in vivo, suggesting a novel anti-angiogenic therapy based on inducible p27 overexpression.


Subject(s)
Cell Cycle Proteins/physiology , Cell Movement/physiology , Endothelium, Vascular/cytology , Neovascularization, Pathologic , Tumor Suppressor Proteins , Adenoviridae , Cell Cycle Proteins/genetics , Cell Division/physiology , Cyclin-Dependent Kinase Inhibitor p27 , Doxycycline , Extremities/blood supply , Genetic Vectors , Humans , Regional Blood Flow
7.
Hum Gene Ther ; 12(12): 1543-9, 2001 Aug 10.
Article in English | MEDLINE | ID: mdl-11506696

ABSTRACT

The current study was designed to test the hypothesis that intramuscular (i.m.) injection of naked DNA leads to distribution of the injectate remote from the site of needle placement, a finding that might be expected to facilitate i.m. gene transfer. Transcutaneous ultrasound imaging was employed to monitor online the extent to which a solution of phVEGF165 was distributed among the skeletal musculature during 288 i.m. injections in 18 consecutive patients. In 237 (82.3%) of 288 muscle sites, the injection was performed into the distal calf muscle. In 51 (17.7%) of 288 muscle sites, injection was performed into the first and/or second interosseous muscles of the dorsal foot. Unperturbed muscle was recognized by a characteristic echogenic, stippled texture that was bounded by more intensely echogenic fascia. When i.m. gene transfer was performed into the calf muscles, the injectate was distributed along a longitudinal dimension of 3.59 +/- 0.79 cm (1.39-5.87 cm); the corresponding area of injectate measured by on-line planimetry was 1.83 +/- 0.51 cm2 (0.62-3.41 cm2). When i.m. gene transfer was performed into the interosseous muscles of the foot, the longitudinal extent of injectate distribution was 2.49 +/- 0.66 cm (1.61-3.91 cm), with a corresponding injectate area of 1.71 +/- 0.54 cm2 (0.51-2.86 cm2). These findings establish that a solution of plasmid DNA administered by direct i.m. injection into the skeletal muscles of the limb is distributed well beyond the site of needle entry. Thus, the use of multiple injections performed at different sites is likely to result in broad distribution of DNA injectate, a physical factor that may act to facilitate naked DNA and/or other gene transfer strategies.


Subject(s)
DNA/administration & dosage , DNA/metabolism , Extremities/blood supply , Gene Transfer Techniques , Ischemia/therapy , Muscles/diagnostic imaging , Muscles/metabolism , Ultrasonics , Endothelial Growth Factors/genetics , Genetic Therapy/methods , Humans , Leg/diagnostic imaging , Lymphokines/genetics , Muscle, Skeletal/diagnostic imaging , Muscle, Skeletal/metabolism , Plasmids/administration & dosage , Plasmids/metabolism , Time Factors , Ultrasonography , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
8.
J Clin Invest ; 108(3): 399-405, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11489933

ABSTRACT

Endothelial progenitor cells (EPCs) have been isolated from circulating mononuclear cells in peripheral blood and shown to incorporate into foci of neovascularization, consistent with postnatal vasculogenesis. These circulating EPCs are derived from bone marrow and are mobilized endogenously in response to tissue ischemia or exogenously by cytokine stimulation. We show here, using a chemotaxis assay of bone marrow mononuclear cells in vitro and EPC culture assay of peripheral blood from simvastatin-treated animals in vivo, that the HMG-CoA reductase inhibitor, simvastatin, augments the circulating population of EPCs. Direct evidence that this increased pool of circulating EPCs originates from bone marrow and may enhance neovascularization was demonstrated in simvastatin-treated mice transplanted with bone marrow from transgenic donors expressing beta-galactosidase transcriptionally regulated by the endothelial cell-specific Tie-2 promoter. The role of Akt signaling in mediating effects of statin on EPCs is suggested by the observation that simvastatin rapidly activates Akt protein kinase in EPCs, enhancing proliferative and migratory activities and cell survival. Furthermore, dominant negative Akt overexpression leads to functional blocking of EPC bioactivity. These findings establish that augmented mobilization of bone marrow-derived EPCs through stimulation of the Akt signaling pathway constitutes a novel function for HMG-CoA reductase inhibitors.


Subject(s)
Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Protein Serine-Threonine Kinases , Animals , Cell Survival/drug effects , Cells, Cultured , Chemotaxis/drug effects , Cornea/blood supply , Cornea/drug effects , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/metabolism , Hematopoietic Stem Cells/metabolism , Humans , In Vitro Techniques , Lymphokines/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neovascularization, Physiologic/drug effects , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Signal Transduction/drug effects , Simvastatin/pharmacology , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
10.
Nat Med ; 7(6): 706-11, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11385508

ABSTRACT

Sonic hedgehog (Shh) is a prototypical morphogen known to regulate epithelial/mesenchymal interactions during embryonic development. We found that the hedgehog-signaling pathway is present in adult cardiovascular tissues and can be activated in vivo. Shh was able to induce robust angiogenesis, characterized by distinct large-diameter vessels. Shh also augmented blood-flow recovery and limb salvage following operatively induced hind-limb ischemia in aged mice. In vitro, Shh had no effect on endothelial-cell migration or proliferation; instead, it induced expression of two families of angiogenic cytokines, including all three vascular endothelial growth factor-1 isoforms and angiopoietins-1 and -2 from interstitial mesenchymal cells. These findings reveal a novel role for Shh as an indirect angiogenic factor regulating expression of multiple angiogenic cytokines and indicate that Shh might have potential therapeutic use for ischemic disorders.


Subject(s)
Endothelial Growth Factors/metabolism , Lymphokines/metabolism , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Neovascularization, Physiologic/physiology , Proteins/metabolism , Signal Transduction , Trans-Activators , Angiopoietin-1 , Angiopoietin-2 , Animals , Blood Vessels/anatomy & histology , Blood Vessels/physiology , Corneal Neovascularization , Endothelial Growth Factors/genetics , Female , Genes, Reporter , Hedgehog Proteins , Hindlimb/blood supply , Humans , Immunohistochemistry , Ischemia/therapy , Lymphokines/genetics , Male , Membrane Glycoproteins/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Myocardium/metabolism , Patched Receptors , Proteins/genetics , Receptors, Cell Surface , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/metabolism , Regional Blood Flow/physiology , Up-Regulation , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
12.
Arch Neurol ; 58(5): 761-8, 2001 May.
Article in English | MEDLINE | ID: mdl-11346371

ABSTRACT

OBJECTIVE: To investigate the effects of vascular endothelial growth factor gene therapy on ischemic neuropathy in patients with critical limb ischemia. DESIGN: An open-label, dose-escalating trial. Patients with angiographically proven critical leg ischemia received injections of phVEGF(165) human plasmid in the muscles of the ischemic limb. Testing before treatment and at 3 and 6 months included (1) symptom severity score, (2) clinical examination score, and (3) electrophysiologic studies. Clinical and electrophysiologic examiners were masked to each other's findings. SETTING: A tertiary care referral hospital and a major teaching affiliate of Tufts University School of Medicine, Boston, Mass. RESULTS: Of 29 consecutive patients enrolled, 17 (19 limbs) completed the 6 months of study. Six patients had diabetes. Compared with baseline studies, treated patients had significant clinical improvements in the symptom score (P<.01), sensory examination score (P<.01), total examination score (P =.01), peroneal motor amplitude (P =.03), and quantitative vibration threshold (P =.04). Improvement in the vascular ankle-brachial index in treated legs (P<.01) corresponded to improvement in neuropathy in the same limb. Neurologic improvement was seen in 4 of 6 patients with diabetes who completed the study. No clinical, electrophysiologic, or vascular improvements were observed in untreated legs. CONCLUSIONS: Ischemic neuropathy might be a reversible condition, and therapeutic angiogenesis might be an effective treatment. The presence of diabetes does not preclude a response to this therapy.


Subject(s)
Critical Illness/therapy , Endothelial Growth Factors/genetics , Genetic Therapy , Ischemia/therapy , Leg/innervation , Lymphokines/genetics , Peripheral Nerves/blood supply , Adult , Aged , Chronic Disease , Cohort Studies , Electrophysiology , Female , Gene Dosage , Gene Expression , Humans , Ischemia/physiopathology , Male , Middle Aged , Neovascularization, Physiologic , Peripheral Nerves/physiopathology , Prospective Studies , Sensation , Severity of Illness Index , Transgenes/genetics , Treatment Outcome , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
13.
J Clin Invest ; 107(9): 1083-92, 2001 May.
Article in English | MEDLINE | ID: mdl-11342572

ABSTRACT

The pathogenetic basis for diabetic neuropathy has been enigmatic. Using two different animal models of diabetes, we have investigated the hypothesis that experimental diabetic neuropathy results from destruction of the vasa nervorum and can be reversed by administration of an angiogenic growth factor. Nerve blood flow, as measured by laser Doppler imaging or direct detection of a locally administered fluorescent lectin analogue, was markedly attenuated in rats with streptozotocin-induced diabetes, consistent with a profound reduction in the number of vessels observed. A severe peripheral neuropathy developed in parallel, characterized by significant slowing of motor and sensory nerve conduction velocities, compared with nondiabetic control animals. In contrast, 4 weeks after intramuscular gene transfer of plasmid DNA encoding VEGF-1 or VEGF-2, vascularity and blood flow in the nerves of treated animals were similar to those of nondiabetic control rats; constitutive overexpression of both transgenes resulted in restoration of large and small fiber peripheral nerve function. Similar experiments performed in a rabbit model of alloxan-induced diabetes produced comparable results. These findings support the notion that diabetic neuropathy results from microvascular ischemia involving the vasa nervorum and suggest the feasibility of a novel treatment strategy for patients in whom peripheral neuropathy constitutes a secondary complication of diabetes.


Subject(s)
Diabetes Mellitus, Experimental/complications , Diabetic Nephropathies/etiology , Diabetic Nephropathies/therapy , Endothelial Growth Factors/genetics , Genetic Therapy/methods , Lymphokines/genetics , Animals , Endothelial Growth Factors/therapeutic use , Female , Gene Transfer Techniques , Laser-Doppler Flowmetry , Lymphokines/therapeutic use , Peripheral Nervous System Diseases/etiology , Peripheral Nervous System Diseases/therapy , Rabbits , Rats , Rats, Sprague-Dawley , Sciatic Nerve/blood supply , Streptozocin , Tibial Nerve/blood supply , Vasa Nervorum/pathology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
14.
Circulation ; 103(17): 2138-43, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11331253

ABSTRACT

BACKGROUND: Catheter-based myocardial gene transfer (GTx) has not been previously tested in human subjects. Accordingly, we performed a pilot study to investigate the feasibility and safety of catheter-based myocardial GTx of naked plasmid DNA encoding vascular endothelial growth factor-2 (phVEGF-2) in patients with chronic myocardial ischemia. METHODS AND RESULTS: A steerable, deflectable 8F catheter incorporating a 27-guage needle was advanced percutaneously to the left ventricular myocardium of 6 patients with chronic myocardial ischemia. Patients were randomized (1:1) to receive phVEGF-2 (total dose, 200 microgram), which was administered as 6 injections into ischemic myocardium (total, 6.0 mL), or placebo (mock procedure). Injections were guided by NOGA left ventricular electromechanical mapping. Patients initially randomized to placebo became eligible for phVEGF-2 GTx if they had no clinical improvement 90 days after their initial procedure. Catheter injections (n=36) caused no changes in heart rate or blood pressure. No sustained ventricular arrhythmias, ECG evidence of infarction, or ventricular perforations were observed. phVEGF-2-transfected patients experienced reduced angina (before versus after GTx, 36.2+/-2.3 versus 3.5+/-1.2 episodes/week) and reduced nitroglycerin consumption (33.8+/-2.3 versus 4.1+/-1.5 tablets/week) for up to 360 days after GTx; reduced ischemia by electromechanical mapping (mean area of ischemia, 10.2+/-3.5 versus 2.8+/-1.6 cm(2), P=0.04); and improved myocardial perfusion by SPECT-sestamibi scanning for up to 90 days after GTx when compared with images obtained after control procedure. Conclusions-This randomized trial of catheter-based phVEGF-2 myocardial GTx provides preliminary indications regarding the feasibility, safety, and potential efficacy of percutaneous myocardial GTx to human left ventricular myocardium.


Subject(s)
Cardiac Catheterization , DNA, Recombinant/administration & dosage , Myocardial Ischemia/therapy , Neovascularization, Physiologic/genetics , Transfection , Vascular Endothelial Growth Factors/therapeutic use , Ventricular Function, Left , Aged , DNA, Recombinant/genetics , DNA, Recombinant/therapeutic use , Feasibility Studies , Female , Heart Ventricles/physiopathology , Humans , Male , Myocardial Ischemia/diagnostic imaging , Myocardial Ischemia/physiopathology , Pilot Projects , Safety , Tomography, Emission-Computed, Single-Photon , Treatment Outcome , Vascular Endothelial Growth Factors/genetics
15.
Annu Rev Physiol ; 63: 427-50, 2001.
Article in English | MEDLINE | ID: mdl-11181962

ABSTRACT

This review surveys a range of approaches using plasmid DNA encoding the 165-amino-acid isoform of vascular endothelial growth factor (phVEGF165) to therapeutically modulate micro- or macrovascular endothelial cells, focusing on strategies to augment postnatal collateral circulation in arterial insufficiency or to accelerate re-endothelialization after balloon angioplasty to prevent restenosis. We focus on intra-arterial and intramuscular/intramyocardial gene transfer of the VEGF165 gene, the options that have been most thoroughly studied to date in patients. We review developmental and postnatal significance of the endothelial-cell-specific mitogen VEGF that has stimulated these studies and present limitations of current knowledge as well as challenges for the future.


Subject(s)
Coronary Disease/therapy , Endothelial Growth Factors/genetics , Genetic Therapy , Lymphokines/genetics , Neovascularization, Physiologic/genetics , Animals , Humans , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
16.
J Mol Cell Cardiol ; 33(3): 379-93, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11181008

ABSTRACT

In animal models of ischemia, a large body of evidence indicates that administration of angiogenic growth factors, either as recombinant protein or by gene transfer, can augment nutrient perfusion through neovascularization. While many cytokines have angiogenic activity, the best studied both in animal models and clinical trials are vascular endothelial growth factor (VEGF) and fibroblast growth factor (FGF). Clinical trials of therapeutic angiogenesis in patients with end-stage coronary artery disease have shown large increases in exercise time and marked reductions in symptoms of angina, as well as objective evidence of improved perfusion and left ventricular function. Larger scale placebo-controlled trials have been limited to intracoronary and intravenous administration of recombinant protein, and have not yet shown significant improvement in either exercise time or angina when compared to placebo. Larger scale placebo-controlled studies of gene transfer are in progress. Future clinical studies will be required to determine the optimal dose, formulation, route of administration and combinations of growth factors, as well as the requirement for endothelial progenitor cell or stem cell supplementation, to provide effective and safe therapeutic myocardial angiogenesis.


Subject(s)
Endothelial Growth Factors/therapeutic use , Fibroblast Growth Factors/therapeutic use , Lymphokines/therapeutic use , Myocardial Ischemia/therapy , Neovascularization, Pathologic/therapy , Animals , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/therapy , Cerebrovascular Disorders/drug therapy , Cerebrovascular Disorders/therapy , Coronary Disease/drug therapy , Coronary Disease/therapy , Cytokines/metabolism , Endothelial Growth Factors/genetics , Endothelial Growth Factors/metabolism , Fibroblast Growth Factors/metabolism , Forecasting , Gene Transfer Techniques , Genetic Therapy , Humans , Hypoxia , Lymphokines/genetics , Lymphokines/metabolism , Myocardial Ischemia/drug therapy , Neovascularization, Pathologic/drug therapy , Neovascularization, Physiologic , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor , Recombinant Proteins/therapeutic use , Risk Factors , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
17.
Circulation ; 103(5): 634-7, 2001 Feb 06.
Article in English | MEDLINE | ID: mdl-11156872

ABSTRACT

BACKGROUND: We investigated the therapeutic potential of ex vivo expanded endothelial progenitor cells (EPCs) for myocardial neovascularization. METHODS AND RESULTS: Peripheral blood mononuclear cells obtained from healthy human adults were cultured in EPC medium and harvested 7 days later. Myocardial ischemia was induced by ligating the left anterior descending coronary artery in male Hsd:RH-rnu (athymic nude) rats. A total of 10(6) EPCs labeled with 1,1'-dioctadecyl-1 to 3,3,3',3'-tetramethylindocarbocyanine perchlorate were injected intravenously 3 hours after the induction of myocardial ischemia. Seven days later, fluorescence-conjugated Bandeiraea simplicifolia lectin I was administered intravenously, and the rats were immediately killed. Fluorescence microscopy revealed that transplanted EPCs accumulated in the ischemic area and incorporated into foci of myocardial neovascularization. To determine the impact on left ventricular function, 5 rats (EPC group) were injected intravenously with 10(6) EPCs 3 hours after ischemia; 5 other rats (control group) received culture media. Echocardiography, performed just before and 28 days after ischemia, disclosed ventricular dimensions that were significantly smaller and fractional shortening that was significantly greater in the EPC group than in the control group by day 28. Regional wall motion was better preserved in the EPC group. After euthanization on day 28, necropsy examination disclosed that capillary density was significantly greater in the EPC group than in the control group. Moreover, the extent of left ventricular scarring was significantly less in rats receiving EPCs than in controls. Immunohistochemistry revealed capillaries that were positive for human-specific endothelial cells. CONCLUSIONS: Ex vivo expanded EPCs incorporate into foci of myocardial neovascularization and have a favorable impact on the preservation of left ventricular function.


Subject(s)
Endothelium, Vascular/cytology , Hematopoietic Stem Cells/physiology , Leukocytes, Mononuclear/transplantation , Myocardial Ischemia/therapy , Neovascularization, Pathologic/therapy , Animals , Cell Differentiation , Cell Transplantation , Cells, Cultured , Disease Models, Animal , Humans , Leukocytes, Mononuclear/cytology , Male , Rats , Rats, Nude , Treatment Outcome , Ventricular Function, Left
18.
Anesth Analg ; 92(1): 19-25, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11133594

ABSTRACT

UNLABELLED: Gene transfer for therapeutic angiogenesis represents a novel treatment for medically intractable angina in patients judged not amenable to further conventional revascularization. We describe the anesthetic management of 30 patients with class 3 or 4 angina, enrolled in a Phase 1 clinical trial to assess the safety and bioactivity of direct myocardial gene transfer of naked DNA-encoding vascular endothelial growth factor (phVEGF(165)), as sole therapy for refractory angina. The phVEGF(165) was injected directly into the myocardium through a mini-thoracotomy. All patients had major clinical predictors for adverse perioperative cardiac complications. Fast-track anesthetic management with remifentanil and desflurane, multimodal analgesia, and aggressive hemodynamic control with nitroglycerin and esmolol were used. All patients tolerated anesthesia and surgery without problems. No perioperative myocardial infarction, hemodynamic instability, or ventricular failure occurred. VEGF injections caused no clinically significant changes in cardiovascular function. Mean hospital stay was 3.8 days. There was one late death (5 months postoperative). Twenty-nine of 30 patients experienced reduced angina (56.2 +/- 4.1 episodes/week preoperatively versus 3.8 +/- 1.6 postoperatively, P < 0.0001) and reduced sublingual nitroglycerin consumption (60.1 +/- 4.4 tablets/week preoperatively versus 2.9 +/- 1.1 postoperatively, P < 0.0001). IMPLICATIONS: Previously revascularized patients now judged "inoperable," continue to present with chronic, recurrent angina. Our study describes the anesthetic considerations and management of such patients treated with a novel approach by using gene therapy to stimulate angiogenesis and improve perfusion to ischemic myocardium.


Subject(s)
Anesthesia, General/methods , Angina Pectoris/therapy , DNA/administration & dosage , Endothelial Growth Factors/genetics , Genetic Therapy/methods , Lymphokines/genetics , Myocardial Revascularization/methods , DNA/genetics , Drug Administration Routes , Female , Hemodynamics , Humans , Male , Middle Aged , Myocardium , Neovascularization, Physiologic/genetics , Nitroglycerin/administration & dosage , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Vasodilator Agents/administration & dosage
19.
Arterioscler Thromb Vasc Biol ; 21(1): 6-12, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11145928

ABSTRACT

Multiple lines of evidence suggest that estrogen directly modulates angiogenesis via effects on endothelial cells. Under physiological conditions, angiogenesis is routinely observed in the uterus in association with fluctuations in the levels of circulating estradiol and other sex steroids. In pathological circumstances, such as breast cancer, a clear association between estrogen, estrogen receptor expression by endothelial cells, angiogenic activity, and/or tumor invasiveness has been made. Studies performed in our laboratory have revealed that estradiol accelerates functional endothelial recovery after arterial injury. Despite these consistent observations, the mechanisms by which estrogen regulates angiogenesis under physiological and pathological circumstances have not been defined.


Subject(s)
Estrogens/adverse effects , Estrogens/physiology , Neovascularization, Pathologic/etiology , Neovascularization, Physiologic , Animals , Endothelium, Vascular/cytology , Endothelium, Vascular/pathology , Endothelium, Vascular/physiology , Humans
20.
Vasc Med ; 6(4): 235-9, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11958389

ABSTRACT

Intermittent claudication is the most common symptom of peripheral arterial disease (PAD), in part due to an inadequate rise in limb blood flow with exercise. Claudication causes a severe impairment in functional capacity and quality of life in over 3 million Americans. Basic fibroblast growth factor (bFGF) stimulates angiogenesis in vivo and improves limb blood flow in several animal models of hindlimb ischemia. However, the relative safety and efficacy of angiogenic molecules in the treatment of claudication has not been fully evaluated in prospective, blinded clinical trials. In this study, a randomized, double-blind, placebo-controlled, phase II trial of recombinant human bFGF for the treatment of intermittent claudication was performed. bFGF was administered weekly by intravenous infusions of 2 microg/kg for 6 sequential weeks (total dose 12 microg/kg). The primary efficacy endpoint was change in peak walking time (PWT) on a graded exercise treadmill protocol. Secondary efficacy endpoints included changes in functional status as measured by validated questionnaires. The study was stopped prematurely after treatment of the first 24 subjects due to proteinuria in five of the 16 subjects who received systemic bFGF, which exceeded 1000 mg/24 h in four of these five subjects. The small sample size limited evaluation of the predefined efficacy endpoints; however, there was no significant difference between the treatment and control groups for any of the measures of efficacy. In conclusion, intravenous administration of bFGF delivered at low doses weekly for 6 weeks was associated with a high rate of severe proteinuria. It is speculated that bFGF-related proteinuria in this study was primarily related to the systemic route of administration and the frequent dosing schedule. Future clinical trials of bFGF protein should carefully monitor renal function and consider alternative dosing schedules and drug administration routes.


Subject(s)
Fibroblast Growth Factor 2/therapeutic use , Intermittent Claudication/complications , Intermittent Claudication/drug therapy , Proteinuria/chemically induced , Aged , Circadian Rhythm , Double-Blind Method , Endpoint Determination , Exercise Test , Female , Follow-Up Studies , Humans , Male , Middle Aged , Risk Factors , Surveys and Questionnaires , Time Factors , Treatment Outcome , United States/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL
...