Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Prev Med ; 171: 107489, 2023 06.
Article in English | MEDLINE | ID: mdl-37031910

ABSTRACT

The diagnosis of peripheral arterial disease (PAD) is not always evident as symptoms and signs may show great variation. As all grades of PAD are linked to both an increased risk for cardiovascular complications and adverse limb events, awareness of the condition and knowledge about diagnostic measures, prevention and treatment is crucial. This article presents in a condensed form information on PAD and its management.


Subject(s)
Atherosclerosis , Peripheral Arterial Disease , Humans , Peripheral Arterial Disease/diagnosis , Atherosclerosis/diagnosis , Risk Factors
4.
Gene Ther ; 19(3): 264-70, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21716303

ABSTRACT

Peripheral artery disease is a progressive disease. Primary ischemic leg symptoms are muscle fatigue, discomfort or pain during ambulation, known as intermittent claudication. The most severe manifestation of peripheral artery disease is critical limb ischemia (CLI). The long-term safety of gene therapy in peripheral artery disease remains unclear. This four center peripheral artery disease registry was designed to evaluate the long-term safety of the intramuscular non-viral fibroblast growth factor-1 (NV1FGF), a plasmid-based angiogenic gene for local expression of fibroblast growth factor-1 versus placebo in patients with peripheral artery disease who had been included in five different phase I and II trials. Here we report a 3-year follow-up in patients suffering from CLI or intermittent claudication. There were 93 evaluable patients, 72 of them in Fontaine stage IV (47 NV1FGF versus 25 placebo) and 21 patients in Fontaine stage IIb peripheral artery disease (15 NV1FGF versus 6 placebo). Safety parameters included rates of non-fatal myocardial infarction (MI), stroke, death, cancer, retinopathy and renal dysfunction. At 3 years, in 93 patients included this registry, there was no increase in retinopathy or renal dysfunction associated with delivery of this angiogenic factor. There was also no difference in the number of strokes, MI or deaths, respectively, for NV1FGF versus placebo. In the CLI group, new cancer occurred in two patients in the NV1FGF group. Conclusions that can be drawn from this relatively small patient group are limited because of the number of patients followed and can only be restricted to safety. Yet, data presented may be valuable concerning rates in cancer, retinopathy, MI or strokes following angiogenesis gene therapy in the absence of any long-term data in angiogenesis gene therapy. It may take several years until data from larger patient populations will become available.


Subject(s)
Fibroblast Growth Factor 1/genetics , Genetic Vectors/administration & dosage , Peripheral Arterial Disease/genetics , Peripheral Arterial Disease/therapy , Aged , Aged, 80 and over , Amputation, Surgical , Female , Fibroblast Growth Factor 1/metabolism , Follow-Up Studies , Genetic Therapy , Genetic Vectors/adverse effects , Humans , Injections, Intramuscular , Male , Middle Aged , Myocardial Infarction/complications , Neoplasms/complications , Peripheral Arterial Disease/complications , Peripheral Arterial Disease/mortality , Stroke/complications , Survival Analysis
5.
Eur J Vasc Endovasc Surg ; 42(3): 365-73, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21696982

ABSTRACT

BACKGROUND: Patients with critical limb ischaemia (CLI) unsuitable for revascularisation have a high rate of amputation and mortality (30% and 25% at 1 year, respectively). Localised gene therapy using plasmid DNA encoding acidic fibroblast growth factor (NV1FGF, riferminogene pecaplasmid) has showed an increased amputation-free survival in a phase II trial. This article provides the rationale, design and baseline characteristics of CLI patients enrolled in the pivotal phase III trial (EFC6145/TAMARIS). METHODS: An international, double-blind, placebo-controlled, randomised study composed of 525 CLI patients recruited from 170 sites worldwide who were unsuitable for revascularisation and had non-healing skin lesions was carried out to evaluate the potential benefit of repeated intramuscular administration of NV1FGF. Randomisation was stratified by country and by diabetic status. RESULTS: The mean age of the study cohort was 70 ± 10 years, and included 70% males and 53% diabetic patients. Fifty-four percent of the patients had previous lower-extremity revascularisation and 22% had previous minor amputation of the index leg. In 94% of the patients, the index leg had distal occlusive disease affecting arteries below the knee. Statins were prescribed for 54% of the patients, and anti-platelet drugs for 80%. Variation in region of origin resulted in only minor demographic imbalance. Similarly, while diabetic status was associated with a frequent history of coronary artery disease, it had little impact on limb haemodynamics and vascular lesions. CONCLUSIONS: Clinical characteristics and vascular anatomy of CLI patients with ischaemic skin lesions who were unsuitable for revascularisation therapy show little variations by region of origin and diabetic status. The findings from this large CLI cohort will contribute to our understanding of this disease process. This study is registered with ClinicalTrials.gov, number NCT00566657.


Subject(s)
Angiogenesis Inducing Agents/therapeutic use , Arterial Occlusive Diseases/drug therapy , Diabetic Angiopathies/drug therapy , Fibroblast Growth Factor 1/therapeutic use , Ischemia/etiology , Lower Extremity/blood supply , Aged , Aged, 80 and over , Arterial Occlusive Diseases/complications , Diabetic Angiopathies/complications , Female , Humans , Internationality , Male , Middle Aged , Research Design
6.
Dtsch Med Wochenschr ; 136(14): 672-4, 2011 Apr.
Article in German | MEDLINE | ID: mdl-21448824

ABSTRACT

The world-wide largest angiogenesis gene therapy trial in 525 patients suffering from critical limb ischemia and treated with the non-viral gene construct for fibroblast growth factor (NV1FGF) or placebo was negative for all endpoints. Also, stem cell or progenitor cell therapy did no reveal clinical benefit in patients with peripheral artery disease. Data from large randomized placebo-controlled are still not available.


Subject(s)
Arterial Occlusive Diseases/therapy , Fibroblast Growth Factor 1/genetics , Genetic Therapy/methods , Neovascularization, Physiologic/genetics , Stem Cell Transplantation/methods , Amputation, Surgical , Angiography , Arterial Occlusive Diseases/genetics , Arterial Occlusive Diseases/mortality , Controlled Clinical Trials as Topic , Gene Transfer Techniques , Humans , Intermittent Claudication/genetics , Intermittent Claudication/mortality , Intermittent Claudication/therapy , Ischemia/genetics , Ischemia/mortality , Ischemia/therapy , Leg/blood supply , Pilot Projects , Randomized Controlled Trials as Topic , Stem Cells/cytology , Treatment Failure
7.
Vasa ; 36(3): 165-73, 2007 Aug.
Article in English | MEDLINE | ID: mdl-18019272

ABSTRACT

Peripheral artery disease is a highly prevalent disease which is characterised by a high unmet medical need particularly in the more advanced stages of disease. Recent advances in the knowledge of the complex regulation of angiogenesis and arteriogenesis and ways to its induction offer hope for a novel strategy that is based on the generation of such new vessels. This strategy termed "therapeutic angiogenesis" is a concept based on the use of angiogenic factors or stem cells or their combination to promote neovascularisation for the treatment of ischaemic tissues. This article reviews both regulation of angiogenesis and the development of therapeutic strategies based on this knowledge using gene therapy. This includes knowledge from animal experiments as well as from phase I and phase II clinical trials. This information may be particularly important at a time when angiogenesis gene therapy enters the stage of phase III clinical testing hopefully leading to the first time approval of this completely new class of drug in the near future. Following articles of this series will review therapeutic angiogenesis approaches based on cytokine therapy and stem cell therapy.


Subject(s)
Angiogenic Proteins/metabolism , Extremities/blood supply , Genetic Therapy/methods , Ischemia/therapy , Neovascularization, Physiologic/genetics , Peripheral Vascular Diseases/therapy , Angiogenic Proteins/genetics , Animals , Endothelial Cells/metabolism , Gene Transfer Techniques , Genetic Therapy/adverse effects , Humans , Ischemia/etiology , Ischemia/genetics , Ischemia/metabolism , Ischemia/physiopathology , Myocytes, Smooth Muscle/metabolism , Peripheral Vascular Diseases/complications , Peripheral Vascular Diseases/genetics , Peripheral Vascular Diseases/metabolism , Peripheral Vascular Diseases/physiopathology , Stem Cells/metabolism , Treatment Outcome
8.
Vasa ; 36(4): 241-51, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18357916

ABSTRACT

Peripheral arterial disease (PAD) is a common manifestation of systemic atherosclerosis that is associated with a significant limitation in limb function due to ischaemia and high risk of cardiovascular mortality. The lower limb manifestations of PAD principally fall into the categories of chronic stable claudication, critical leg ischaemia, and, rarely, acute limb ischaemia. Lower limb ischaemia induced by PAD is a major health problem. In the absence of effective pharmacological, interventional or surgical treatment, amputation is undertaken at the end-stage as a solution to unbearable symptoms. The concept of "therapeutic angiogenesis" has become widely accepted during the past few years. Bone marrow consists of multiple cell populations, including endothelial progenitor cells, which have been shown to differentiate into endothelial cells and release several angiogenic factors and thereby enhance neovascularisation in animal models of hind limb ischaemia. The promising results from various preclinical studies provide the basis for clinical trials using bone marrow-derived cells or non-bone marrow cells, like cells from the peripheral blood or other tissues. However, the mechanisms by which these cells exert their positive effects are poorly understood until now. This review summarises the data from experimental and clinical studies related to peripheral arterial disease and cellular therapy.


Subject(s)
Angiogenesis Inducing Agents/metabolism , Arterial Occlusive Diseases/therapy , Bone Marrow Transplantation , Ischemia/therapy , Leg/blood supply , Animals , Arterial Occlusive Diseases/physiopathology , Cell Differentiation , Clinical Trials as Topic , Disease Models, Animal , Endothelial Cells/cytology , Hindlimb/blood supply , Humans , Ischemia/physiopathology , Mice , Neovascularization, Physiologic/physiology , Rabbits , Rats
9.
Vasa ; 36(4): 253-60, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18357917

ABSTRACT

In recent years the importance of circulating bone marrow-derived cells in angiogenesis and collateral growth has been demonstrated in peripheral artery disease (PAD) and other ischaemic diseases. Although the mechanisms by which these cells exert their angiogenetic/arteriogenetic effects are not completely understood, improving the accumulation of bone marrow-derived cells at the site of vascular growth using cytokines has become one aim in some of the regenerative therapies. Interestingly recent data indicate that in addition to effects attributed to such accumulated cells there are also direct effects of cytokines used via their receptors. Several investigations in animal hind limb models of ischaemia have demonstrated the beneficial effect of bone marrow mobilisation using colony-stimulating factors (CSF) on collateral growth and perfusion recovery. Clinical studies in PAD patients, however are still rare and led to inconsistent data, in part due to different application protocols, choice of cytokine and low patient numbers with strong placebo effects. Moreover; the aetiology of the disease in humans differs markedly from the artificial occlusion of the femoral artery in a mostly healthy animal in the preclinical setting. Another approach to enhance arteriogenesis, which has been successful in animal models of hind limb ischaemia, is the local injection of the monocyte chemoattractant protein 1 (MCP-1). This treatment stimulated the invasion of monocytes leading to improved collateral growth and restoration of limb perfusion. Recent reports from animal experiments, in which both treatment strategies were combined (i.e. bone marrow mobilisation and enhancement of cell migration to the site of vascular growth), have shown strong synergistic effects, pointing at the importance to orchestrate the different processes involved in vascular repair in order to achieve maximal therapeutic effects.


Subject(s)
Arterial Occlusive Diseases/therapy , Bone Marrow Cells , Cytokines/therapeutic use , Ischemia/therapy , Leg/blood supply , Neovascularization, Physiologic/drug effects , Animals , Arterial Occlusive Diseases/physiopathology , Bone Marrow Cells/drug effects , Chemokine CCL2 , Disease Models, Animal , Granulocyte Colony-Stimulating Factor/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Hindlimb/blood supply , Humans , Ischemia/physiopathology
10.
Acta Physiol Scand ; 176(2): 151-9, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12354175

ABSTRACT

Clinical trials have demonstrated therapeutic benefit in inducing angiogenesis in chronic occlusive arterial disease. The route of application mostly used was the intramuscular injection of high dosages of plasmid. Therefore, a local perivascular application of low amounts of vascular endothelial growth factor (VEGF) plasmid was used in an interventional occlusion model, and the effect of VEGF on coronary and peripheral occlusions compared in the same animal model. Coronary and peripheral arteries were chronically occluded in Pietrain pigs using a non-surgical, interventional approach. Adventitial delivery of the DNA for VEGF was performed with a needle injection catheter. The DNA was applied as lipoplexes using the novel cationic liposomes DOCSPER. Optimized transfer conditions were used. Angiography, polymerase chain reaction (PCR), reverse transcriptase-polymerase chain reaction (RT-PCR) and immunohistochemistry were undertaken within a follow-up period of 6 months. Expression of the transfected VEGF gene was observed at 1 and 3 weeks following application. The DNA was detected up to 5 months following application. Around occluded coronary arteries, there was formation of new collaterals and arterial prolongation, whereas surrounding occluded peripheral arteries there was no collateralization but development of new arterial branches was seen. Results demonstrate that the response to VEGF is also sufficient, when minimal amounts of plasmid encoding for VEGF are applied locally into the perivasculature allowing for more safety of this therapy. Comparison of treatment of chronic coronary and peripheral arterial disease revealed differences in angiogenesis following VEGF application during a total follow-up period of almost 6 months which may be related to their different developmental origins. This may have important implications for developing future therapeutic strategies using VEGF in different vessels.


Subject(s)
Arterial Occlusive Diseases/therapy , Coronary Stenosis/therapy , Endothelial Growth Factors/administration & dosage , Intercellular Signaling Peptides and Proteins/administration & dosage , Lymphokines/administration & dosage , Neovascularization, Physiologic/drug effects , Animals , Arterial Occlusive Diseases/diagnostic imaging , DNA/administration & dosage , DNA/analysis , Endothelial Growth Factors/genetics , Genetic Therapy/methods , Genetic Vectors , Iliac Artery , Intercellular Signaling Peptides and Proteins/genetics , Lymphokines/genetics , Models, Biological , Neovascularization, Physiologic/genetics , Plasmids/administration & dosage , Plasmids/genetics , Radiography , Swine , Transfection , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
11.
Z Kardiol ; 91(7): 523-35, 2002 Jul.
Article in German | MEDLINE | ID: mdl-12242949

ABSTRACT

Young women are at a 3-fold higher risk of experiencing venous thromboembolism compared to young men. This risk is further increased by oral contraceptives, smoking, hereditary thrombophilia and/or low socioeconomic status. There are also hints for gender-specific differences in the arterial system. Coronary heart disease which is particularly important, develops in women 10-15 years later than in men. However, the clinical course in women is often more complicated. In women, thrombophilic predispositions play a more important role before menopause and metabolic risk factors after menopause compared to men. Thrombus formation needs to be initiated by disturbances of at least one element of the Virchow trias. There is principally no difference between men and women. However, all three elements are influenced by estrogen, resulting in gender-specific differences. There is a favorable influence on blood flow by the inhibition of atherogenesis and on the endothelial function by increased NO release, and the unfavorable influence on hemostasis resulting in hypercoagibility. Thus, particularly non-selective hormone replacement therapy for the inhibition of atherogenesis may be questionable due to increased thrombogenicity. The solution may be more selective estrogen receptor modulators. Clinical trials are still needed for both hormone replacement therapy and the therapy of venous and arterial thromboembolism, which have been studied in predominantly male patient cohorts so far.


Subject(s)
Arteriosclerosis/epidemiology , Myocardial Infarction/epidemiology , Thromboembolism/epidemiology , Adult , Age Factors , Aged , Arteriosclerosis/etiology , Arteriosclerosis/prevention & control , Female , Hormone Replacement Therapy/adverse effects , Humans , Middle Aged , Myocardial Infarction/etiology , Myocardial Infarction/prevention & control , Risk , Thromboembolism/etiology , Thromboembolism/prevention & control
12.
Cell Prolif ; 34(5): 305-20, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11591178

ABSTRACT

Smooth muscle cells and endothelial cells play an important role in cardiovascular diseases and may therefore be a potential target for gene therapy. Most in vitro experiments are performed using proliferating cell cultures. Nevertheless, non-dividing cells would represent more realistic in vivo conditions for gene therapy. Therefore, a simple method to achieve physiologically quiescence in cell cultures is needed for experiments. Growth to confluence is sufficient for endothelial cells to reach quiescence, in contrast to smooth muscle cells. Alternative techniques were investigated to achieve quiescence for smooth muscle cells. N-acetyl-cysteine, heparin, aphidicolin and serum-free medium are known inhibitors of smooth muscle cell proliferation and were tested for cell viability, necrosis and apoptosis. The inhibition status was evaluated counting cells in a cell counter. Toxicity, necrosis and apoptosis were determined using FACS analysis. Then, smooth muscle cells and endothelial cells were transfected with plasmid containing the beta-galactosidase gene using liposomes. Analysis of gene expression in transfected cells included a quantitative beta-galactosidase assay and X-gal staining. Growth inhibition was achieved with all agents tested. Using N-acetyl-cysteine, only slightly reduced growth rates were observed. Aphidicolin stopped cell growth almost immediately, but demonstrated enhanced toxicity. The amount of apoptotic and necrotic cells was lowest using heparin in the presence of foetal calf serum. Transfection experiments using stationary cultures of smooth muscle cells using heparin or aphidicolin demonstrated 5-10-fold lower transfection rates compared to transfected proliferating cell cultures serving as controls. Transfection experiments using stationary cultures of endothelial cells using growth inhibition through confluence demonstrated 40-fold lower transfection rates than transfected proliferating cell cultures. Transfer efficiency was much lower in endothelial cells compared to smooth muscle cells. In conclusion, quiescent cells simulate more realistically the in vivo situation and may therefore represent a better model for future in vivo experiments based on in vitro findings.


Subject(s)
Cell Survival/drug effects , Endothelium, Vascular/cytology , Growth Inhibitors/pharmacology , Muscle, Smooth, Vascular/cytology , Animals , Aorta , Aphidicolin/pharmacology , Apoptosis , Cell Division/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Flow Cytometry , Heparin/pharmacology , Kinetics , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Necrosis , Recombinant Proteins/biosynthesis , Swine , Transfection , beta-Galactosidase/genetics
13.
J Endovasc Ther ; 8(4): 401-7, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11552732

ABSTRACT

PURPOSE: To determine the optimal endovascular approach to achieve long-term occlusion of large arteries, while preserving the integrity of periarterial tissue, in an animal model of ischemia. METHODS: Femoral artery occlusions were created in 16 pigs using detachable balloons, coils, or blinded stent-grafts. Feasibility, safety, primary and long-term success, and the degree of neovascularization were determined over a 6-month period by serial angiography and histological analyses. Four animals served as untreated controls. RESULTS: Overall primary success for all occlusion devices was 100%. The 6-month occlusion rate using detachable balloons or coils was 33% and 0%, respectively; however, all arteries occluded with blinded stent-grafts remained obstructed to the end of the study. There was no significant difference in capillary densities and collateralization of periarterial areas when occluded arteries were compared with nonoccluded controls in the same animal. No increase in collateralization was observed following endovascular arterial occlusion. CONCLUSIONS: Percutaneous insertion of blinded stent-grafts easily, safely, and reliably creates long-term arterial occlusion in pigs, which may make this a more appropriate model for studying the effects of angiogenic factors in vivo.


Subject(s)
Arterial Occlusive Diseases/surgery , Femoral Artery/surgery , Vascular Surgical Procedures , Anatomy, Cross-Sectional , Animals , Catheterization , Disease Models, Animal , Extremities/blood supply , Femoral Artery/anatomy & histology , Femoral Artery/diagnostic imaging , Follow-Up Studies , Radiography , Stents , Swine , Time , Time Factors , Vascular Surgical Procedures/instrumentation
15.
J Microencapsul ; 18(4): 491-506, 2001.
Article in English | MEDLINE | ID: mdl-11428678

ABSTRACT

Long-term benefits of coronary angioplasty remain limited by the treatment-induced renarrowing of arteries, termed restenosis. One of the mechanisms leading to restenosis is the proliferation of smooth muscle cells. Therefore, proliferating cells of the injured arterial wall, which can be selectively transduced by retroviruses, are potential targets for gene therapy strategies. A direct single-dose therapeutic application of retroviral vectors for inhibition of cell proliferation is normally limited by too low transduction efficiencies. Encapsulated retrovirus-producing cells release viral vectors from microcapsules, and may enhance the transduction efficiency by prolonged infection. Primary and immortal murine and porcine cells and murine retrovirus-producing cells were encapsulated in cellulose sulphate. Cell viability was monitored by analysing cell metabolism. Safety, stability, transfer efficiency and extent of restenosis using capsules were determined in a porcine restenosis model for local gene therapy using morphometry, histology, in situ beta-galactosidase assay and PCR. Encapsulation of cells did not impair cell viability. Capsules containing retrovirus-producing cells expressing the beta-galactosidase reporter gene were implanted into periarterial tissue or a pig model of restenosis. Three weeks following implantation, beta-galactosidase activity was detected in the pericapsular tissue with a transduction efficiency of approximately 1 in 500 cells. Adventitial implantation of vector-producing encapsulated cells for gene therapy may, therefore, facilitate successful targeting of proliferating vascular smooth muscle cells, and allow stable integration of therapeutic genes into surrounding cells. The encapsulation of vector-producing cells could represent a novel and feasible way to optimize local retroviral gene therapy.


Subject(s)
Cell Transplantation/methods , Cellulose/analogs & derivatives , Drug Compounding/methods , Gene Transfer Techniques , Retroviridae/genetics , 3T3 Cells , Animals , Base Sequence , Cell Division , Cells, Cultured , Coronary Restenosis/pathology , Coronary Restenosis/therapy , DNA Primers/genetics , Genes, Reporter , Genetic Vectors , Mice , Muscle, Smooth, Vascular/pathology , Swine , beta-Galactosidase/genetics
16.
J Invasive Cardiol ; 13(4): 333-8, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11287726

ABSTRACT

No systemic pharmacological treatment has been shown to convincingly reduce the incidence of restenosis after angioplasty or increase the formation of collaterals in ischemic tissue in patients. The lack of success of many pharmaceutical agents in reducing restenosis rates or in inducing angiogenesis post-angioplasty and following stent implantation has encouraged the development of new technological treatment approaches. Gene therapy is a novel strategy with the potential to prevent some of the sequelae after arterial injury, particularly cell proliferation, and to induce growth of new vessels or remodeling of pre-existing vessel branches, which may help patients with critical ischemia. Gene therapy strategies have the advantage of minimizing systemic side effects and may have a long-term effect as the encoded protein is released. Most clinical trials investigating gene therapy for vascular disease have been uncontrolled phase I and IIa trials. Gene therapy into vessels with the genes for growth factors has been demonstrated to be feasible and efficient. Local drug delivery devices have been used in combination with gene therapy in several trials to maximize safety and efficiency. Data from experimental animal work indicates that gene therapy may modify intimal hyperplasia after arterial injury, but there are few clinical trials on restenosis in patients. Preliminary clinical results show only limited success in altering restenosis rates. In vitro and experimental in vivo investigations into gene therapy for angiogenesis demonstrate increased formation of collaterals and functional improvement of limb ischemia. There is some evidence of increased collateral formation and clinical improvement in patients with critical limb ischemia. Results of placebo-controlled and double-blind trials of gene therapy for vascular disease are awaited.


Subject(s)
Genetic Therapy , Vascular Diseases/therapy , Clinical Trials as Topic , Collateral Circulation , Constriction, Pathologic/prevention & control , Endothelial Growth Factors/genetics , Gene Transfer Techniques , Humans , Ischemia/therapy , Lymphokines/genetics , Protein Isoforms , Recurrence , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
17.
Herz ; 25(6): 600-10, 2000 Sep.
Article in German | MEDLINE | ID: mdl-11076318

ABSTRACT

The identification of angiogenic growth factors was the basis for the development of novel strategies for the treatment of occlusive vascular diseases. Therapeutic angiogenesis resulting in capillary sprouting (angiogenesis) and collateral vessel development (arteriogenesis) may be a potential alternative for patients suffering from critical limb ischemia. Extensive investigations performed in vitro and in vivo demonstrated therapeutic efficacy through the enhancement of collateral growth resulting in augmented blood flow in ischemic tissues following administration of both, recombinant growth factors and genes encoding for such factors. Advantages of gene therapy strategies over the use of proteins include minimal systemic adverse effects and a slow and continuous release of the encoded angiogenic factor which may lead to a long-lasting therapeutic effect. Several uncontrolled phase-I and -IIa trials demonstrated the feasibility and safety of the use of angiogenic growth factors. However, the results of placebo-controlled and double-blind trials are needed to prove their therapeutic potential.


Subject(s)
Angiogenesis Inducing Agents/therapeutic use , Arterial Occlusive Diseases/drug therapy , Endothelial Growth Factors/therapeutic use , Fibroblast Growth Factors/therapeutic use , Lymphokines/therapeutic use , Neovascularization, Physiologic/drug effects , Angiogenesis Inducing Agents/genetics , Animals , Arterial Occlusive Diseases/physiopathology , Clinical Trials as Topic , Endothelial Growth Factors/genetics , Fibroblast Growth Factors/genetics , Genetic Therapy , Humans , Lymphokines/genetics , Neovascularization, Physiologic/physiology , Recombinant Proteins/therapeutic use , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
18.
Mol Ther ; 1(4): 366-75, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10933955

ABSTRACT

Gene therapy strategies for the prevention of restenosis postangioplasty are promising. Nonviral gene transfer to the arterial wall in vivo has so far been limited by poor efficiency. This study aimed to optimize transfection of primary vascular smooth muscle cells using cationic nonviral formulations based on cholesterol derivates (DC-, DAC-, DCQ-, and Sp-Chol), double-chained amphiphils (LipofectAMINE, DOTMA, DOSGA, DOSPER, and DOCSPER), or heterogeneous reagents (Superfect, Effectene, and Tfx-50). Estimation of transfection efficiencies was performed using galactosidase assays at different ratios of transfection reagent to plasmid DNA with reporter gene. Toxicity was monitored by analyzing cell metabolism. Transfer efficiency and safety were determined in a porcine restenosis model for local gene therapy using morphometry, histology, galactosidase assays, and reverse-transcriptase polymerase chain reaction. The highest in vitro transfection efficiency was achieved using the recently developed DOCSPER liposomes, with transfer rates of at least 20% in vascular smooth muscle cells. Transfer efficiency was further enhanced up to 20% by complexing with poly-L-lysine. Transfection efficiency in vivo in a porcine restenosis model was up to 15% of adventitial cells using DOCSPER versus 0.1% using LipofectAMINE. Toxicity in vivo and in vitro was lowest using DOCSPER. Increased biological effects were demonstrated following optimization of transfer conditions.


Subject(s)
Gene Transfer Techniques , Muscle, Smooth, Vascular/metabolism , Angioplasty , Animals , Base Sequence , DNA Primers/genetics , Gene Expression , Genes, Reporter , Genetic Therapy , Humans , In Vitro Techniques , Liposomes , Muscle, Smooth, Vascular/cytology , Plasmids/administration & dosage , Plasmids/genetics , Swine , Transfection , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
19.
Int J Angiol ; 9(2): 87-94, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10758203

ABSTRACT

Up to 30% of patients undergoing coronary angioplasty develop a renarrowing of treated vessels following percutaneous transluminal coronary angioplasty with or without stent implantation, called restenosis. Smooth muscle cell proliferation, among other mechanisms, is an important factor in restenosis leading to neointima formation and consequent arterial lumen narrowing. Cecropins are antimicrobial peptides with antiproliferative properties in mammalian cells which have been shown to suppress neointimal formation. In this investigation, a plasmid carrying the gene for pre-pro-cecropin A, complexed with new generation liposomes optimized for transfer conditions for vascular cells was delivered to the adventitia of arteries in a porcine arterial injury model using a needle injection catheter. Retention of the plasmid in treated arteries was demonstrated for at least 21 days following delivery. Whereas previous experiments using first generation liposomes demonstrated significant but not complete neointima inhibition, the use of new liposomes under optimized conditions resulted in almost total suppression of neointimal proliferation. Thus, in vivo gene transfer of cecropins may be therapeutically applicable in restenosis prevention.

20.
J Vasc Res ; 37(6): 585-93, 2000.
Article in English | MEDLINE | ID: mdl-11146413

ABSTRACT

The reporter gene for beta-galactosidase is frequently used to determine the efficiency of gene transfer in arteries. However, blood is often present in arterial explants and may compromise the results by the presence of hemoglobin. The light absorption of hemoglobin is similar to the absorption of several colorimetric products of the commonly used beta-galactosidase substrates, including o-nitrophenyl-beta-D-galactopyranoside (ONPG) and chlorophenol red galactopyranoside (CPRG). This may result in false-positive measurements of beta-galactosidase enzyme activity. The aim of this investigation was to determine the most appropriate method for quantification of beta-galactosidase activity in the presence of blood. Colorimetric substrates (ONPG, CPRG) or the chemiluminescent Galacton-Plus substrate were used, and light absorption was measured at different concentrations of erythrocyte extract. Among the beta-galactosidase substrates tested, CPRG was the most appropriate, allowing detection of enzyme activity at concentrations as low as 0.05 mU, independent of blood contamination. Addition of reducer stabilized enzyme activity for at least 5 h. Endogenous beta-galactosidase activity was evaluated and used to correct results. CPRG substrate, in combination with the reducer agent mercaptoethanol, was found to be the optimal reagent for quantifying beta-galactosidase activity in the presence of blood after nonviral in vivo reporter gene transfection, even with a relatively low transfer efficiency.


Subject(s)
Artifacts , Blood , Colorimetry/methods , Lac Operon , Luminescent Measurements , beta-Galactosidase/analysis , Animals , Arteries/cytology , Cattle , Cell Extracts , Cells, Cultured , Chlorophenols/analysis , Erythrocytes/chemistry , Galactosides/analysis , Genes, Reporter , Hemoglobins/chemistry , Humans , Mercaptoethanol/pharmacology , Muscle, Smooth, Vascular/cytology , Nitrophenylgalactosides/analysis , Photometry , RNA, Messenger/biosynthesis , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/metabolism , Reducing Agents/pharmacology , Serum Albumin, Bovine/pharmacology , Swine , Transfection , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...