Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
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
2.
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
3.
Cardiology ; 95(1): 40-7, 2001.
Article in English | MEDLINE | ID: mdl-11385191

ABSTRACT

We evaluated intracardiac masses in vivo, in situ and histologically to determine tissue properties revealed by magnetic resonance (MR) imaging. In 15 consecutive patients scheduled for cardiotomy, the cardiac chambers were studied preoperatively with MR imaging and echocardiography. Visual examination of one or more chambers was performed during cardiotomy for mitral valve replacement, aneurysmectomy, atrial septal repair and atriotomy. Six thrombi (1 atrial appendage, 5 ventricular) and 2 atrial myxomas were removed and subjected to histological analysis. All masses were detected preoperatively by MR imaging. The smallest was a subacute 3-mm mural clot in the left ventricle and was undetected by transesophageal and transthoracic echocardiography. The 3 subacute clots had homogeneously low MR signals, did not enhance with gadolinium and exhibited magnetic susceptibility effects; histopathology confirmed these clots to be avascular and laden with dense iron deposition related to hemoglobin breakdown products. The 3 organized clots had intermediate and heterogeneous MR signals and multiple areas of gadolinium enhancement. The 2 myxomas had low MR signals and gadolinium enhancement in the core and septal attachment; these areas had dense neovascular channels. Subacute thrombi appear to have MR features that are distinct from organized thrombi and myxomas, and MR images of subacute thrombi contrast sharply with normal cardiac structures, enabling detection of thin mural clots that may be echographically occult. These findings may be of value, because a subacute clot may be more likely than an organized thrombus to give rise to an embolus.


Subject(s)
Heart Diseases/diagnosis , Magnetic Resonance Imaging , Myxoma/diagnosis , Thrombosis/diagnosis , Aged , Aged, 80 and over , Echocardiography, Transesophageal , Female , Heart Neoplasms/diagnosis , Humans , Male , Middle Aged
4.
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
5.
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
6.
Am J Cardiovasc Drugs ; 1(3): 159-66, 2001.
Article in English | MEDLINE | ID: mdl-14728030

ABSTRACT

Gene therapy is evolving as an alternative mode to pharmacological intervention in the treatment of cardiovascular diseases. Experimental observations indicating that introduction of genes encoding for angiogenic peptide growth factors could result in improvement in perfusion to ischemic myocardium have led to the initiation of a number of preliminary clinical trials to evaluate this therapeutic modality. Sustained expression of the growth factor product from somatic cells transfected with the DNA for that protein has proven to be one of the major advantages of a gene therapy based approach over administration of the recombinant protein. A number of gene therapy vectors have been developed, prominent among these being adenoviral vectors and naked plasmid DNA. Whereas plasmid DNA results in less efficient transfection, its tolerability profile may be superior to adenoviral vectors. Plasmid DNA is particularly suitable when the gene product to be produced is capable of being secreted by the cell which is producing it. Vascular endothelial growth factor (VEGF) is not only essential to the process of angiogenesis, but, because it can be secreted from intact cells, appears to be ideal for gene transfer therapy aimed at improving perfusion to ischemic myocardium. The DNA can be delivered to the myocardium by intra-arterial or intramuscular injection. At present, direct injection into the muscle either via a small thoracotomy incision or by use of a recently developed percutaneous catheter technique appears to be superior to arterial administration. Several clinical trials based on intramyocardial injection of VEGF DNA in patients with otherwise inoperable coronary artery disease and intractable angina pectoris have recently been completed. These phase I trials have documented the tolerability of gene transfer using plasmid DNA and show promise of being able to improve myocardial perfusion and reduce anginal symptoms in the majority of patients treated thus far. While the trials involving gene transfer via a thoracotomy did not allow for randomization to a placebo group, the recent advent of a percutaneous delivery modality has allowed for randomization which should enhance our ability to determine whether angiogenic gene therapy will prove to be as effective as initial results suggest. In the future, results from such randomized placebo-controlled trials, improvement in vectors utilized for gene transfer and innovative new delivery techniques will undoubtedly enhance the potential of this novel approach to myocardial revascularization.


Subject(s)
Genetic Therapy , Myocardial Ischemia/therapy , Clinical Trials as Topic , Gene Transfer Techniques , Genetic Vectors , Humans , Myocardial Ischemia/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/therapy , Vascular Endothelial Growth Factors/genetics
7.
Ann Thorac Surg ; 70(3): 829-34, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11016318

ABSTRACT

BACKGROUND: Direct transfection of ischemic myocardium with naked plasmid DNA encoding for vascular endothelial growth factor-165 (VEGF165) has been shown to mobilize endothelial progenitor cells (EPCs). This study examined the kinetics of circulating EPCs isolated from peripheral blood mononuclear cells after gene transfer, and their role in neovascularization of ischemic myocardium. METHODS: The mononuclear cell population was isolated from peripheral venous blood samples of patients with functional class III or IV angina receiving intramyocardial VEGF165 gene transfer. Peripheral blood mononuclear cells were examined by an in vitro EPC culture assay and fluorescent-activated cell sorting. The data were compared with a control group consisting of patients who had undergone off-pump coronary artery bypass grafting without receiving gene transfer. RESULTS: Coinciding with a rise in VEGF levels, mobilization of EPCs increased significantly over base line for 9 weeks after the treatment (121+/-14 cells/mm2 versus 36.8+/-8 cells/mm2, p < 0.0005), followed by a subsequent decrease. Fluorescent-activated cell sorting analysis confirmed culture assay data, with a statistically significant rise in cells expressing vascular endothelial-cadherin, CD51/61 [alphavbeta3], CD62E [E-selectin], CD34, and KDR. The control group failed to show significant mobilization of EPCs. CONCLUSIONS: Mobilization of EPCs with resultant postnatal vasculogenesis, may play a role in revascularizing ischemic myocardium following human gene transfer with VEGF165.


Subject(s)
Coronary Disease/therapy , Endothelial Growth Factors/genetics , Endothelium, Vascular/cytology , Gene Transfer Techniques , Lymphokines/genetics , Cell Separation , Coronary Artery Bypass , Flow Cytometry , Humans , Transfection , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
8.
Circulation ; 102(9): 965-74, 2000 Aug 29.
Article in English | MEDLINE | ID: mdl-10961959

ABSTRACT

BACKGROUND: NOGA left ventricular (LV) electromechanical mapping (EMM) can be used to distinguish among infarcted, ischemic, and normal myocardium. We investigated the use of percutaneous LV EMM to assess the efficacy of myocardial gene transfer (GTx) of naked plasmid DNA encoding for vascular endothelial growth factor (phVEGF(165)), administered during surgery by direct myocardial injection in patients with chronic myocardial ischemia. METHODS AND RESULTS: A total of 13 consecutive patients (8 men, mean age 60.1+/-2. 3 years) with chronic stable angina due to angiographically documented coronary artery disease, all of whom had failed conventional therapy (drugs, PTCA, and/or CABG), were treated with direct myocardial injection of phVEGF(165) via a minithoracotomy. Foci of ischemic myocardium were identified on LV EMM by preserved viability associated with an impairment in linear local shortening. Myocardial viability, defined by mean unipolar and bipolar voltage recordings >/=5 and >/=2 mV, respectively, did not change significantly after GTx. Analysis of linear local shortening in areas of myocardial ischemia, however, disclosed significant improvement after (15.26+/-0.98%) versus before (9.94+/-1.53%, P:=0. 004) phVEGF(165) GTx. The area of ischemic myocardium was consequently reduced from 6.45+/-1.37 cm(2) before GTx to 0.95+/-0. 41 cm(2) after GTx (P:=0.001). These findings corresponded to improved perfusion scores calculated from single-photon emission CT-sestamibi myocardial perfusion scans recorded at rest (7.4+/-2.1 before GTx versus 4.5+/-1.4 after GTx, P:=0.009) and after pharmacological stress (12.8+/-2.7 before GTx versus 8.5+/-1.7 after GTx, P:=0.047). CONCLUSIONS: The results of EMM constitute objective evidence that phVEGF(165) GTx augments perfusion of ischemic myocardium. These findings, together with reduction in the size of the defects documented at rest by serial single-photon emission CT-sestamibi imaging, suggest that phVEGF(165) GTx may successfully rescue foci of hibernating myocardium.


Subject(s)
Endothelial Growth Factors/genetics , Genetic Therapy/methods , Lymphokines/genetics , Myocardial Ischemia/therapy , Neovascularization, Physiologic , Angiogenesis Inducing Agents/therapeutic use , Coronary Circulation/drug effects , Electrocardiography/methods , Endothelial Growth Factors/therapeutic use , Gene Transfer Techniques , Heart/diagnostic imaging , Heart/physiopathology , Humans , Lymphokines/therapeutic use , Male , Microinjections , Middle Aged , Myocardial Ischemia/diagnostic imaging , Myocardial Ischemia/physiopathology , Plasmids/therapeutic use , Radionuclide Imaging , Thoracotomy , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Ventricular Function, Left
9.
J Card Surg ; 15(4): 283-90, 2000.
Article in English | MEDLINE | ID: mdl-11758065

ABSTRACT

BACKGROUND: Therapeutic angiogenesis, which involves the administration of angiogenic cytokines to stimulate collateral formation and improve myocardial perfusion, is being tested as an alternative treatment for patients with medically intractable angina who are no longer candidates for conventional revascularization techniques. METHODS: Administration of recombinant protein formulations for these growth factors and transfection of the DNA itself have proven effective in animal models. At present, transfection of the DNA (gene therapy) appears preferable in that a single administration achieves a prolonged but transient, localized exposure of the ischemic tissues to the angiogen. The important technical aspects of surgical gene therapy via a mini thoracotomy incision are described. RESULTS: Four Phase I clinical trials of angiogenic therapy recently have been reported, two involving administration of the recombinant protein combined with coronary bypass and two involving gene therapy alone. These studies have shown that this approach is relatively safe; the early evidence of efficacy is encouraging. The largest study reported six-month follow-up on 30 patients who received plasmid DNA for vascular endothelial growth factor-165 (VEGF165) and documented improvement in angina, treadmill exercise tolerance, and myocardial perfusion in the majority of patients studied. CONCLUSIONS: These preliminary results will need to be supported by randomized clinical trials in which angiogenic therapy alone is compared to standard medical therapy or a placebo. Catheter-based intramyocardial gene transfer and combined coronary arterial bypass grafting or transmyocardial laser revascularization plus gene therapy trials are also underway. If proven effective, angiogenic therapy may provide a relatively noninvasive modality for revascularization of ischemic myocardium not amenable to current techniques.


Subject(s)
Angiogenesis Inducing Agents , Endothelial Growth Factors/therapeutic use , Genetic Therapy , Lymphokines/therapeutic use , Myocardial Revascularization/methods , Animals , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Coronary Artery Bypass , Humans , Myocardial Reperfusion/methods , Recombinant Proteins/therapeutic use , Transfection , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
10.
Hum Gene Ther ; 10(18): 2953-60, 1999 Dec 10.
Article in English | MEDLINE | ID: mdl-10609656

ABSTRACT

Both VEGF protein and VEGF DNA in combination with an adenoviral vector have been shown to enhance collateral formation in a porcine model of chronic myocardial ischemia. We sought to determine whether direct intramyocardial injection of naked DNA encoding for VEGF could similarly improve myocardial perfusion. Initially, 23 nonischemic pigs received either 200 microg of plasmid DNA encoding beta-galactosidase (pCMVbeta, n = 11) or 500 microg of phVEGF165 (n = 12) into four separate sites in the myocardium via a small anterolateral thoracotomy incision in the fourth intercostal space. Two additional groups of pigs received an intramyocardial injection of either phVEGF165 (n = 6) or pCMVbeta (n = 7) 3 to 4 weeks after implantation of an ameroid constrictor around the left circumflex coronary artery. The injections caused no change in heart rate or blood pressure, and no ventricular arrhythmias or histologic evidence of inflammation. VEGF protein was detected by Western blot in VEGF-treated animals, with the strongest bands closest to the injection site. Plasma VEGF concentration (ELISA) increased from 3+/-2 to 27+/-13 pg/ml (p = 0.035) by day 4 after treatment. No increase in VEGF protein was noted in pCMVbeta-treated animals whereas these did stain positive for beta-Gal. Resting myocardial blood flow (colored microspheres) was significantly reduced in the ischemic versus nonischemic territory in control animals (1.07+/-0.05 versus 1.32+/-0.05; p < 0.05) but not VEGF-treated pigs (1.32+/-0.24 versus 1.13+/-0.12; p = NS). Maximal vasodilatation with adenosine significantly increased flow to the ischemic region in VEGF-treated pigs (2.16+/-0.57 versus 1.32+/-0.24; p < 0.05) but not controls (1.31+/-0.05 versus 1.17+/-0.06;p = NS). Collateral filling of the occluded circumflex artery improved in five of six VEGF-treated pigs (mean change in Rentrop score, +1.5). We conclude that direct intramyocardial transfection phVEGF165 is safe and capable of producing sufficient VEGF protein to enhance collateral formation and myocardial perfusion. This approach may offer an alternative therapy for patients with intractable myocardial ischemia not amenable to PTCA or CABG.


Subject(s)
Collateral Circulation , Coronary Vessels/physiopathology , DNA/administration & dosage , Endothelial Growth Factors/genetics , Genetic Therapy , Lymphokines/genetics , Myocardial Ischemia/therapy , Myocardium/metabolism , Animals , Disease Models, Animal , Gene Expression , Myocardial Ischemia/physiopathology , Swine , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , beta-Galactosidase/genetics
11.
Ann Thorac Surg ; 68(3): 830-6; discussion 836-7, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10509970

ABSTRACT

BACKGROUND: Patients presenting with medically intractable angina who have undergone previous coronary bypass (CABG) and/or percutaneous revascularization procedures are frequently deemed "inoperable" based on angiographic findings of diffuse distal disease or a lack of available conduits. We initiated a phase I clinical trial to assess the safety and bioactivity of intramyocardial transfection of plasmid DNA encoding for the angiogenic mitogen vascular endothelial growth factor (ph-VEGF165) in such patients. METHODS: phVEGF165 (125 microg, n = 10; 250 microg, n = 10) was injected directly into the myocardium through a mini left anterior thoracotomy as sole therapy in 20 patients (15 male, 5 female, age 48 to 74 years) with class III or IV angina, reversible ischemia on stress sestamibi scans, and "inoperable" coronary artery disease. RESULTS: All patients tolerated surgery uneventfully and were extubated on the table. No perioperative myocardial infarction, hemodynamic instability, or change in ventricular function occurred. Mean hospital stay was 3.9 days. There was one late death (4 months). Plasma VEGF protein level increased from 30.6+/-4.1 pg/mL pretreatment to 73.7+/-10.1 pg/mL 14 days posttreatment (p = 0.0002) and returned to baseline by day 90. All 16 patients followed to day 90 reported a reduction in angina (nitroglycerin use/week = 60.2+/-4.9 preop vs 3.5+/-1.6 at 90 days; p<0.0001). Seventy percent (7 of 10) patients were completely angina free at 6 months. A reduction in ischemic defects on single photon emission computerized tomography sestamibi scans was observed in 13 of 17 patients at 60 days (7 of 8 in the 250-microg group). Stress perfusion score decreased from 19.4+/-3.7 at baseline to 15.9+/-3.4 at 60 days (p = 0.025). Angiographic evidence of improved collateral filling of at least one occluded vessel was observed in all patients evaluated at day 60. CONCLUSIONS: Direct myocardial gene transfer with phVEGF165 via a mini-thoracotomy can be performed safely and may result in significant symptomatic improvement in patients with "inoperable" coronary artery disease.


Subject(s)
Coronary Disease/therapy , Endothelial Growth Factors/genetics , Genetic Therapy , Lymphokines/genetics , Aged , Coronary Angiography , Coronary Circulation , Coronary Disease/diagnostic imaging , Endothelial Growth Factors/blood , Escherichia coli , Female , Genetic Vectors , Humans , Lymphokines/blood , Male , Middle Aged , Technetium Tc 99m Sestamibi , Tomography, Emission-Computed, Single-Photon , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
12.
Hum Gene Ther ; 10(14): 2307-14, 1999 Sep 20.
Article in English | MEDLINE | ID: mdl-10515450

ABSTRACT

Gene transfer for therapeutic angiogenesis represents a novel treatment for patients with chronic angina refractory to standard medical therapy and not amenable to conventional revascularization. We sought to assess the role of intraoperative multiplane transesophageal echocardiography (MPTEE) in guiding injection of naked DNA encoding vascular endothelial growth factor (VEGF) into the left ventricular (LV) myocardium of patients with refractory angina. After exposing the LV myocardium via a limited lateral thoracotomy, each of 17 patients in this series received 4 separate injections of VEGF DNA into different myocardial sites. Initial injections in the first patient produced intracavitary microbubbles, indicating injection of DNA into the LV chamber. Subsequently, each injection was preceded by a test injection of agitated saline. The absence of microbubbles while visualizing the LV cavity during the test injection verified that the ensuing injection of DNA would not be inadvertently squandered in the LV chamber itself. Intracavitary LV microbubbles were observed by MPTEE in 13 of 64 (20.3%) saline test injections and in 8 of 16 (50.0%) patients in which saline test injection was used, leading to adjustments in needle position. MPTEE imaging detected a previously unknown large, apical left ventricular thrombus in one patient, thereby preventing inadvertent injection of VEGF DNA through the myocardium into the thrombus. Imaging during and after injection verified no deleterious impact on LV function. We conclude that MPTEE is a useful tool for ensuring that myocardial gene therapy performed by direct needle injection results in gene transfer to the LV myocardium.


Subject(s)
Angina Pectoris/therapy , Endothelial Growth Factors/genetics , Genetic Therapy , Lymphokines/genetics , Myocardium/metabolism , Aged , Angina Pectoris/diagnostic imaging , Angina Pectoris/physiopathology , Cardiac Surgical Procedures , Echocardiography, Transesophageal , Endothelial Growth Factors/metabolism , Female , Gene Transfer Techniques , Humans , Intraoperative Period , Lymphokines/metabolism , Male , Middle Aged , Plasmids/administration & dosage , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Ventricular Function, Left
13.
Curr Cardiol Rep ; 1(4): 335-43, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10980864

ABSTRACT

Despite the proven effectiveness of coronary bypass surgery and percutaneous angioplasty techniques, an increasing number of patients are presenting with severe, medically intractable angina who are not candidates for either procedure. Two alternative strategies, transmyocardial laser revascularization and exogeneous administration of angiogenic growth factors (therapeutic angiogenesis) are currently being evaluated in such patients. This review focuses on the current status of these two procedures, emphasizing their similarities and differences in order to provide insight into what role each may ultimately play in the management of patients with otherwise unrevascularizable myocardial ischemia.


Subject(s)
Coronary Disease/therapy , Endothelial Growth Factors/therapeutic use , Genetic Therapy , Laser Therapy/methods , Lymphokines/therapeutic use , Myocardial Revascularization/methods , Administration, Cutaneous , Animals , Combined Modality Therapy , Coronary Disease/surgery , Cost-Benefit Analysis , Fibroblast Growth Factors/therapeutic use , Humans , Pericardium/surgery , Transfection , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
14.
Circulation ; 98(25): 2800-4, 1998.
Article in English | MEDLINE | ID: mdl-9860779

ABSTRACT

BACKGROUND: We initiated a phase 1 clinical study to determine the safety and bioactivity of direct myocardial gene transfer of vascular endothelial growth factor (VEGF) as sole therapy for patients with symptomatic myocardial ischemia. METHODS AND RESULTS: VEGF gene transfer (GTx) was performed in 5 patients (all male, ages 53 to 71) who had failed conventional therapy; these men had angina (determined by angiographically documented coronary artery disease). Naked plasmid DNA encoding VEGF (phVEGF165) was injected directly into the ischemic myocardium via a mini left anterior thoracotomy. Injections caused no changes in heart rate (pre-GTx=75+/-15/min versus post-GTx=80+/-16/min, P=NS), systolic BP (114+/-7 versus 118+/-7 mm Hg, P=NS), or diastolic BP (57+/-2 versus 59+/-2 mm Hg, P=NS). Ventricular arrhythmias were limited to single unifocal premature beats at the moment of injection. Serial ECGs showed no evidence of new myocardial infarction in any patient. Intraoperative blood loss was 0 to 50 cm3, and total chest tube drainage was 110 to 395 cm3. Postoperative cardiac output fell transiently but increased within 24 hours (preanesthesia=4.8+/-0.4 versus postanesthesia=4.1+/-0.3 versus 24 hours postoperative=6. 3+/-0.8, P=0.02). Time to extubation after closure was 18.4+/-1.4 minutes; average postoperative hospital stay was 3.8 days. All patients had significant reduction in angina (nitroglycerin [NTG] use=53.9+/-10.0/wk pre-GTx versus 9.8+/-6.9/wk post-GTx, P<0.03). Postoperative left ventricular ejection fraction (LVEF) was either unchanged (n=3) or improved (n=2, mean increase in LVEF=5%). Objective evidence of reduced ischemia was documented using dobutamine single photon emission computed tomography (SPECT)-sestamibi imaging in all patients. Coronary angiography showed improved Rentrop score in 5 of 5 patients. CONCLUSIONS: This initial experience with naked gene transfer as sole therapy for myocardial ischemia suggests that direct myocardial injection of naked plasmid DNA, via a minimally invasive chest wall incision, is safe and may lead to reduced symptoms and improved myocardial perfusion in selected patients with chronic myocardial ischemia.


Subject(s)
Endothelial Growth Factors/genetics , Genetic Therapy/methods , Lymphokines/genetics , Myocardial Ischemia/therapy , Aged , Coronary Angiography , Humans , Injections, Intralesional , Male , Middle Aged , Minimally Invasive Surgical Procedures , Myocardial Ischemia/diagnostic imaging , Neovascularization, Physiologic , Plasmids/genetics , Thoracotomy/methods , Tomography, Emission-Computed, Single-Photon , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
15.
J Vasc Surg ; 28(6): 964-73; discussion 73-5, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9845647

ABSTRACT

PURPOSE: Thromboangiitis obliterans (TAO), or Buerger's disease, a distinct form of vascular occlusive disease that afflicts the peripheral arteries of young smokers, is often characterized by an inexorable downhill course even in patients who discontinue smoking once a stage of critical limb ischemia associated with ulceration or gangrene is reached. As part of a phase I clinical trial to document the safety and efficacy of intramuscular gene transfer of naked plasmid DNA-encoding vascular endothelial growth factor (phVEGF165) in the treatment of critical limb ischemia, we treated TAO in 6 patients. METHODS: Seven limbs in 6 patients (3 men, 3 women; mean age, 33 years; range, 33 to 51 years) who satisfied the criteria for TAO and had signs or symptoms of critical limb ischemia were treated twice, 4 weeks apart, with 2 or 4 mg of phVEGF165, which was administered by direct intramuscular injection at 4 arbitrarily selected sites in the ischemic limb. The gene expression was documented by enzyme-linked immunosorbent assay that was performed on peripheral blood samples. RESULTS: The ulcers that were nonhealing for more than 1 month healed completely in 3 of 5 limbs after the intramuscular phVEGF165 gene therapy. Nocturnal rest pain was relieved in the remaining 2 patients, although both continue to have claudication. The evidence of the improved perfusion to the distal ischemic limb included an increase of more than 0.1 in the ankle brachial index in 3 limbs, an improved flow shown with magnetic resonance imaging in 7 of the 7 limbs, and newly visible collateral vessels shown with serial contrast angiography in 7 of the 7 limbs. The adverse consequences of the phVEGF165 gene transfer were limited to transient ankle or calf edema in 3 of the 7 limbs. Two patients with advanced distal forefoot gangrene ultimately required below-knee amputation despite the evidence of improved perfusion. A histologic section disclosed the classic pathologic findings of TAO. CONCLUSION: Therapeutic angiogenesis with phVEGF165 gene transfer, if instituted before the development of forefoot gangrene, may provide a novel therapy for patients with advanced Buerger's disease that is unresponsive to standard medical or surgical treatment methods.


Subject(s)
Endothelial Growth Factors/genetics , Gene Transfer Techniques , Genetic Therapy , Lymphokines/genetics , Thromboangiitis Obliterans/therapy , Adult , Angiography, Digital Subtraction , Endothelial Growth Factors/blood , Enzyme-Linked Immunosorbent Assay , Female , Gene Transfer Techniques/adverse effects , Genetic Therapy/adverse effects , Hemodynamics , Humans , Injections, Intramuscular , Ischemia/diagnosis , Ischemia/etiology , Leg/blood supply , Lymphokines/blood , Magnetic Resonance Angiography , Male , Middle Aged , Neovascularization, Physiologic , Thromboangiitis Obliterans/complications , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
16.
J Clin Invest ; 101(11): 2567-78, 1998 Jun 01.
Article in English | MEDLINE | ID: mdl-9616228

ABSTRACT

We tested the hypothesis that endothelial nitric oxide synthase (eNOS) modulates angiogenesis in two animal models in which therapeutic angiogenesis has been characterized as a compensatory response to tissue ischemia. We first administered L-arginine, previously shown to augment endogenous production of NO, to normal rabbits with operatively induced hindlimb ischemia. Angiogenesis in the ischemic hindlimb was significantly improved by dietary supplementation with L-arginine, compared to placebo-treated controls; angiographically evident vascularity in the ischemic limb, hemodynamic indices of limb perfusion, capillary density, and vasomotor reactivity in the collateral vessel-dependent ischemic limb were all improved by oral L-arginine supplementation. A murine model of operatively induced hindlimb ischemia was used to investigate the impact of targeted disruption of the gene encoding for ENOS on angiogenesis. Angiogenesis in the ischemic hindlimb was significantly impaired in eNOS-/- mice versus wild-type controls evaluated by either laser Doppler flow analysis or capillary density measurement. Impaired angiogenesis in eNOS-/- mice was not improved by administration of vascular endothelial growth factor (VEGF), suggesting that eNOS acts downstream from VEGF. Thus, (a) eNOS is a downstream mediator for in vivo angiogenesis, and (b) promoting eNOS activity by L-arginine supplementation accelerates in vivo angiogenesis. These findings suggest that defective endothelial NO synthesis may limit angiogenesis in patients with endothelial dysfunction related to atherosclerosis, and that oral L-arginine supplementation constitutes a potential therapeutic strategy for accelerating angiogenesis in patients with advanced vascular obstruction.


Subject(s)
Ischemia/physiopathology , Neovascularization, Physiologic , Nitric Oxide Synthase/physiology , Animals , Arginine/pharmacology , Cyclic GMP/analysis , Endothelial Growth Factors/genetics , Endothelial Growth Factors/pharmacology , Hemodynamics/drug effects , Iliac Artery/physiology , Lymphokines/genetics , Lymphokines/pharmacology , Male , Mice , Mice, Inbred C57BL , Rabbits , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
17.
J Vasc Surg ; 27(1): 167-73, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9474095

ABSTRACT

PURPOSE: Recent evidence suggests that vascular endothelial growth factor (VEGF), in addition to stimulating angiogenesis, also serves a repair/maintenance or survival function, modulating various aspects of endothelial cell function. This study was designed to examine the effect of VEGF pretreatment in a model of vein graft intimal hyperplasia. METHODS: Reversed jugular vein-to-common carotid artery interposition grafts were constructed in New Zealand White rabbits. White rabbits. Vein conduits were immersed in solution containing 500 micrograms rhVEGF165 or saline solution for 20 minutes before implantation. Twenty-eight days later the vein grafts and contralateral control jugular veins were harvested for either histologic or isometric tension studies. RESULTS: VEGF-treated vein grafts showed a 23% reduction in intimal area (0.76 +/- 0.07 mm2 vs 0.98 +/- 0.06 mm2; p = 0.028) and a 30% reduction in intimal thickness (62 +/- 6 microns vs 89 +/- 5 microns; p = 0.001) when compared with control grafts. After precontraction with norepinephrine, the maximal relaxation to acetylcholine (endothelium-dependent, receptor-mediated agonist) for control vein grafts was 0%, whereas for VEGF-treated vein grafts it was 25% +/- 9% (p < 0.05 vs control grafts). The maximal relaxation to the calcium ionophore A23187 (endothelium-dependent, receptor-independent agonist) was also greater in VEGF-treated grafts than in control grafts (172.3% +/- 19.4% vs 122.5% +/- 13.7%; p < 0.05). There was no difference in the response to sodium nitroprusside (endothelium-independent agonist) between the two groups. CONCLUSIONS: A single topical application of VEGF before implantation reduces intimal hyperplasia and improves endothelial function in a rabbit vein graft model. Further evaluation of this simple strategy to improve vein graft patency appears warranted.


Subject(s)
Endothelial Growth Factors/pharmacology , Endothelium, Vascular/physiology , Lymphokines/pharmacology , Tunica Intima/pathology , Vasodilation , Veins/pathology , Veins/transplantation , Acetylcholine/pharmacology , Animals , Calcimycin/pharmacology , Endothelial Growth Factors/physiology , Hyperplasia , Lymphokines/physiology , Male , Nitroprusside/pharmacology , Rabbits , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Vasodilation/drug effects
18.
Arterioscler Thromb Vasc Biol ; 17(11): 2793-800, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9409257

ABSTRACT

In vitro studies suggest that vascular endothelial growth factor/vascular permeability factor (VEGF/VPF) may stimulate release of nitric oxide (NO) from endothelial cells. To investigate the hemodynamic consequences of recombinant VEGF/VPF administered in vivo, recombinant human VEGF/VPF was administered as a bolus dose of 500 micrograms to anesthetized (n = 6) or conscious (n = 5) New Zealand White rabbits, as well as anesthetized rabbits with diet-induced hypercholesterolemia (HC; n = 7). Anesthetized Yorkshire farm pigs (no specific dietary pretreatment) were studied before and after receiving 500 micrograms intravenous (IV; n = 5) or intracoronary (IC; n = 5) VEGF/VPF. In anesthetized, normal rabbits, mean arterial pressure (MAP) fell by 20.5 +/- 1.4% (P < .05 versus baseline) within 3 minutes after IV VEGF/VPF. Pretreatment with N omega-nitro-L-arginine caused a significant inhibition of VEGF/VPF-induced hypotension. In conscious, normal rabbits, VEGF/VPF produced a consistent though lesser reduction in MAP. The fall in MAP induced by VEGF/VPF in anesthetized, HC rabbits (21.5 +/- 2.5% from baseline) was no different from that observed in normal anesthetized rabbits. In pigs, both IV and IC administration of VEGF/VPF produced a prompt reduction in MAP. Heart rate increased, while cardiac output, stroke volume, left atrial pressure, and total peripheral resistance all declined to a similar, statistically significant degree in both IV and IC groups. Epicardial echocardiography disclosed neither global nor segmental wall motion abnormalities in response to VEGF/VPF. We conclude that (1) VEGF/VPF-stimulated release of NO, previously suggested in vitro, occurs in vivo; (2) this finding suggests that functional VEGF/VPF receptors are present on quiescent adult endothelium, consistent with a maintenance function for VEGF/VPF, which may include regulation of NO; and (3) the preserved response of HC rabbits suggests that endothelial cell receptors for VEGF/VPF are spared in the setting of hypercholesterolemia.


Subject(s)
Endothelial Growth Factors/toxicity , Endothelium, Vascular/metabolism , Hypotension/chemically induced , Lymphokines/toxicity , Nitric Oxide/physiology , Animals , Aorta/drug effects , Cholesterol, Dietary/toxicity , Diet, Atherogenic , Echocardiography , Endothelial Growth Factors/pharmacology , Enzyme Inhibitors/pharmacology , Hemodynamics/drug effects , Humans , Hypercholesterolemia/etiology , Hypercholesterolemia/physiopathology , Hypotension/physiopathology , Lymphokines/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Nitroarginine/pharmacology , Rabbits , Receptor Protein-Tyrosine Kinases/drug effects , Receptor Protein-Tyrosine Kinases/physiology , Receptors, Growth Factor/drug effects , Receptors, Growth Factor/physiology , Receptors, Vascular Endothelial Growth Factor , Recombinant Proteins/pharmacology , Recombinant Proteins/toxicity , Secretory Rate/drug effects , Swine , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Vasodilation/drug effects , omega-N-Methylarginine/pharmacology
19.
Circulation ; 96(9 Suppl): II-382-8, 1997 Nov 04.
Article in English | MEDLINE | ID: mdl-9386128

ABSTRACT

BACKGROUND: Ischemic skeletal muscle has been shown to be advantageous for taking up and expressing genes transferred in the form of naked plasmid DNA. Therefore, acutely ischemic skeletal muscle may represent a potential target for IM gene therapy with naked DNA. Accordingly, we investigated the impact of IM injection of plasmid DNA encoding the secreted angiogenic growth factor, vascular endothelial growth factor (VEGF), on collateral vessel development in an animal model of acute hindlimb ischemia. METHODS AND RESULTS: After ligation of distal external iliac artery in New Zealand White rabbits, we directly injected 500 microg of phVEGF165 into the ischemic thigh muscles. At 30 days posttransfection, VEGF-transfected animals had more angiographically recognizable collateral vessels (angiographic score=0.72+/-0.06 versus 0.48+/-0.10; P<.01) as well as histologically assessed capillaries (248+/-37 versus 180+/-32/mm2, P<.01) compared to controls. Hemodynamic deficit was less severe in VEGF-transfected animals by calf systolic blood pressure ratio (0.80+/-0.09 versus 0.56+/-0.10, P<.01) and by flow to the ischemic limb measured with Doppler guidewire (resting flow=22+/-5 versus 14+/-4; P<.01; hyperemic flow=59+/-17 versus 39+/-12 mL/min; P<.05). Human VEGF mRNA was expressed in the transfected ischemic muscles as long as 14 days after gene transfer. Based on reporter plasmid expression, transfection efficiency was sixfold higher in ischemic muscles than in nonischemic control muscles. CONCLUSIONS: These results suggest the feasibility of employing direct IM transfer of naked VEGF plasmid DNA to optimize treatment of acute limb ischemia.


Subject(s)
Endothelial Growth Factors/genetics , Extremities/blood supply , Genetic Therapy , Ischemia/therapy , Lymphokines/genetics , Acute Disease , Animals , Cytokines/pharmacology , Gene Transfer Techniques , Humans , Injections, Intramuscular , Muscle, Skeletal/metabolism , Neovascularization, Physiologic , Rabbits , Transfection , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
20.
Ann Thorac Surg ; 64(4): 993-8, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9354516

ABSTRACT

BACKGROUND: Hypoxic endothelial cell activation plays a key role in the myocardial dysfunction resulting from ischemia-reperfusion injury. Recent evidence suggests that vascular endothelial growth factor (VEGF) may, in addition to promoting angiogenesis, modulate various aspects of endothelial function and repair. We examined whether administration of VEGF in the cardioplegic solution might have a beneficial effect on myocardial ischemia-reperfusion injury in an isolated rat heart model. METHODS: Hearts from Sprague-Dawley rats were perfused with Krebs-Henseleit solution in a modified Langendorff apparatus. Percent recovery of cardiac output, coronary flow, stroke work, and percent increase in coronary vascular resistance were measured after 2 hours of global ischemia and 40 minutes of reperfusion. Coronary effluent was collected after ischemia and reperfusion for measurement of creatine kinase. RESULTS: Hearts receiving cardioplegia solution containing 125 microg VEGF showed significantly improved recovery of cardiac output, coronary flow, and stroke work, and significantly reduced coronary vascular resistance compared with hearts receiving hyperkalemic cardioplegia only (p < 0.05). Coadministration of a nitric oxide synthase inhibitor attenuated the VEGF-induced cardiprotective effects. Hearts treated with VEGF released significantly less creatine kinase compared with control hearts. CONCLUSIONS: Addition of VEGF to hyperkalemic cardioplegia protects against myocardial ischemia-reperfusion injury in the isolated rat heart.


Subject(s)
Endothelial Growth Factors/therapeutic use , Lymphokines/therapeutic use , Myocardial Reperfusion Injury/prevention & control , Animals , Cardiac Output/drug effects , Cardioplegic Solutions , Creatine Kinase/metabolism , Endothelial Growth Factors/pharmacology , Glucose , Heart/drug effects , Heart/physiology , Lymphokines/pharmacology , Male , Myocardium/enzymology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , Tromethamine , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Vascular Resistance/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...