Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 24
1.
Expert Opin Biol Ther ; 16(3): 303-15, 2016.
Article En | MEDLINE | ID: mdl-26581236

INTRODUCTION: Stimulation of coronary collateral vessel growth by therapeutic angiogenesis (TA) offers an alternative treatment option for patients with refractory angina. Several TA modalities, including delivery to the heart of angiogenic growth factors (proteins or genes) and cells have been tested in clinical trials in the past two decades, but so far none of them resulted in significant therapeutic efficacy in large scale studies. This review attempts to identify the main obstacles hindering clinical success and recommends measures to overcome them in the future. AREAS COVERED: After stating the medical need and rational for TA, and listing and briefly discussing past and current TA clinical trials, three main areas of obstacles are described: conceptual questions, technical limitations and clinical design uncertainties. Based on scientific and technical advances and lessons learned in past clinical trials, potential solutions to overcome some of these obstacles are proposed. EXPERT OPINION: Several success criteria are identified, which apply to any TA approach of choice. It is emphasized, that each of these criteria needs to be met in future clinical trials to have a chance of therapeutic success.


Angina Pectoris/therapy , Collateral Circulation/genetics , Genetic Therapy/methods , Neovascularization, Physiologic/genetics , Adenoviridae , Cell- and Tissue-Based Therapy , Fibroblast Growth Factors/genetics , Humans , Stem Cell Transplantation , Vascular Endothelial Growth Factor A/genetics
2.
J Cardiovasc Pharmacol ; 64(2): 109-19, 2014 Aug.
Article En | MEDLINE | ID: mdl-24736636

Well-developed coronary collateral vessels in patients with symptomatic coronary artery disease were shown to be associated with reduced future cardiovascular morbidity and mortality. However, the majority of patients with coronary artery disease lack adequate functional coronary collateral circulation. Stimulation of collateral vessel development by angiogenic growth factor therapy (therapeutic angiogenesis) has been tested in many clinical trials in the past, but the potential of this new treatment paradigm has not been realized yet in late stage clinical trials. Mechanistic insights into collateral vessel development and the collective clinical experience in the past decade identified specific obstacles that might have impeded the progress in the field. This review identifies some of the key conceptual, technical, and clinical hurdles and recommends strategies to overcome them in future clinical trials.


Genetic Therapy/methods , Myocardial Ischemia/therapy , Neovascularization, Physiologic/genetics , Adenoviridae/genetics , Clinical Trials as Topic , Collateral Circulation/genetics , Fibroblast Growth Factors/genetics , Gene Transfer Techniques , Genetic Vectors , Humans , Myocardial Ischemia/genetics , Myocardial Ischemia/physiopathology , Vascular Endothelial Growth Factor A/genetics
3.
Mol Ther ; 21(4): 725-38, 2013 Apr.
Article En | MEDLINE | ID: mdl-23403495

Stimulation of collateral vessel development in the heart by angiogenic growth factor therapy has been tested in animals and humans for almost two decades. Discordance between the outcome of preclinical studies and clinical trials pointed to the difficulties of translation from animal models to patients. Lessons learned in this process identified specific mechanistic, technical, and clinical hurdles, which need to be overcome. This review summarizes current understanding of the mechanisms leading to the establishment of a functional coronary collateral network and the biological processes growth factor therapies should stimulate even under conditions of impaired natural adaptive vascular response. Vector delivery methods are recommended to maximize angiogenic gene therapy efficiency and reduce side effects. Optimization of clinical trial design should include the choice of clinical end points which provide mechanistic proof-of-concept and also reflect clinical benefits (e.g., surrogates to assess increased collateral flow reserve, such as myocardial perfusion imaging). Guidelines are proposed to select patients who may respond to the therapy with high(er) probability. Both short and longer term strategies are outlined which may help to make therapeutic angiogenesis (TA) work in the future.


Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/therapy , Animals , Collateral Circulation/drug effects , Humans , Neovascularization, Physiologic/drug effects
4.
Hum Gene Ther Methods ; 23(3): 204-12, 2012 Jun.
Article En | MEDLINE | ID: mdl-22816318

Efficiency of intracoronary (IC) adenoviral vector transfection is impaired by the vascular endothelium. Ischemia and substances that increase vascular permeability (sodium nitroprusside, nitroglycerin) may augment adenoviral vector transfection efficiency (TE). We tested whether TE of adenoviral vector following IC infusion is improved by nitrates or by ischemia. Fluoroscopically guided angioplasty balloon catheters occluded the coronary artery in Yorkshire pigs and delivered adenoviral type 5 vector encoding the luciferase gene (Ad5Luc, 10(11) viral particles). TE (luciferase activity) was minimal and was not augmented by IC co-administration of 50 µg/min sodium nitroprusside to nonischemic myocardium. Two (but not one) 3-min episodes of occlusion tended to increase luciferase activity (p=0.06), and luciferase activity was further increased by IC co-administration of nitroglycerin (p<0.001). After 75 min of coronary artery occlusion, luciferase activity was greater than with shorter periods of ischemia, and was significantly greater in the ischemia-reperfused zone compared to the border zone 3 and 14 days after infusion; there was no transfection in nonischemic myocardium. IC delivery of Ad5Luc into post-ischemic myocardium caused no local inflammation or hemodynamic instability. We conclude that the uptake of IC Ad5 to ischemic reperfused myocardium validates use of IC Ad5 delivery protocols in future human gene therapy trials in patients following myocardial ischemia.


Adenoviridae/metabolism , Genetic Therapy/methods , Genetic Vectors/metabolism , Ischemia/metabolism , Myocardium/metabolism , Transfection/methods , Adenoviridae/genetics , Analysis of Variance , Animals , Balloon Occlusion , Cardiac Catheterization , Coronary Vessels/metabolism , DNA Primers/genetics , Genetic Vectors/genetics , Luciferases/genetics , Luciferases/metabolism , Nitroglycerin , Nitroprusside , Polymerase Chain Reaction , Sus scrofa , Transfection/standards
5.
Pharmacol Res ; 63(6): 448-54, 2011 Jun.
Article En | MEDLINE | ID: mdl-20813186

Significant discoveries in biology and medicine are rare. The progress in these fields is predominantly incremental, but sometimes new observations revolutionize the field by opening new directions in research for decades to come. Two cornerstone observations in the late 1970s and early 1980s are examples of such "revolutionary" events. The first, by Furchgott and Zawadzki, was the discovery of the "obligatory role of the endothelium in vasorelaxation by acetylcholine". The other, by Hickey and colleagues, was the first description and characterization of a vasoconstrictor polypeptide produced by endothelial cells in culture. Both of these observations were achieved by the application of bioassay and serendipity played an important role in each of them. They both represent starting points for rapid growth in research activity world-wide leading to the identification of EDRF as nitric oxide, and the polypeptide EDCF as endothelin a few years later. These early observations also raised interest and initiated intensive R&D activity in the pharma industry culminating in the regulatory approval and marketing of novel medicines treating human diseases. This review describes the events leading to the discovery and early characterization of the peptidergic endothelium-derived constrictor factor, and its purification, sequencing and naming it endothelin.


Biological Assay/methods , Endothelins/metabolism , Endothelium/metabolism , Vasoconstrictor Agents/metabolism , Vasodilator Agents/metabolism , Animals , Cells, Cultured , Drug Discovery , Endothelins/chemistry , Endothelins/pharmacology , Humans , Peptides/chemistry , Peptides/metabolism , Vasoconstrictor Agents/chemistry , Vasoconstrictor Agents/pharmacology , Vasodilation/drug effects , Vasodilation/physiology , Vasodilator Agents/chemistry , Vasodilator Agents/pharmacology
6.
Eur J Pharmacol ; 654(1): 68-74, 2011 Mar 01.
Article En | MEDLINE | ID: mdl-21187082

Endothelial dysfunction is a hallmark of, and plays a pivotal role in the pathogenesis of cardiometabolic diseases, including type II diabetes, obesity, and hypertension. It has been well established that epoxyeicosatrienoic acids (EETs) act as an endothelial derived hyperpolarization factor (EDHF). Soluble epoxide hydrolase (s-EH) rapidly hydrolyses certain epoxylipids (e.g. EETs) to less bioactive diols (DHETs), thereby attenuating the evoked vasodilator effects. The aim of the present study was to examine if inhibition of s-EH can restore impaired endothelial function in three animal models of cardiometabolic diseases. Isolated vessel rings of the aorta and/or mesenteric artery from mice or rats were pre-contracted using phenylephrine or U46619. Endothelium-dependent and independent vasorelaxation to acetylcholine and sodium nitroprusside (SNP) were measured using wire myography in vessels isolated from db/db or diet-induced obesity (DIO) mice, and angiotensin II-induced hypertensive rats treated chronically with s-EH inhibitors AR9281 or AR9276 or with vehicle. Vasorelaxation to acetylcholine, but not to SNP was severely impaired in all three animal models. Oral administration of AR9281 or AR9276 abolished whole blood s-EH activity, elevated epoxy/diol lipid ratio, and abrogated endothelial dysfunction in all three models. Incubating the mesenteric artery of db/db mice with L-NAME and indomethacin to block nitric oxide (NO) and prostacyclin formation did not affect AR9821-induced improvement of endothelial function. These data indicate that inhibition of s-EH ameliorates endothelial dysfunction and that effects in the db/db model are independent of the presence of NO and cyclooxygenase derived prostanoids. Thus, preserving vasodilator EETs by inhibition of s-EH may be of therapeutic benefit by improving endothelial function in cardiometabolic diseases.


Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Epoxide Hydrolases/antagonists & inhibitors , Obesity/drug therapy , Adamantane/administration & dosage , Adamantane/analogs & derivatives , Adamantane/pharmacology , Administration, Oral , Animals , Aorta/drug effects , Aorta/metabolism , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 2/physiopathology , Disease Models, Animal , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Epoxide Hydrolases/metabolism , Hypertension/drug therapy , Hypertension/physiopathology , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/metabolism , Mice , Mice, Inbred C57BL , Niacinamide/administration & dosage , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Obesity/physiopathology , Phenylurea Compounds/administration & dosage , Phenylurea Compounds/pharmacology , Rats , Rats, Sprague-Dawley , Urea/administration & dosage , Urea/analogs & derivatives , Urea/pharmacology , Vasodilation/drug effects
7.
PLoS One ; 3(6): e2409, 2008 Jun 18.
Article En | MEDLINE | ID: mdl-18560559

BACKGROUND: Viral-mediated oncolysis is a novel cancer therapeutic approach with the potential to be more effective and less toxic than current therapies due to the agents selective growth and amplification in tumor cells. To date, these agents have been highly safe in patients but have generally fallen short of their expected therapeutic value as monotherapies. Consequently, new approaches to generating highly potent oncolytic viruses are needed. To address this need, we developed a new method that we term "Directed Evolution" for creating highly potent oncolytic viruses. METHODOLOGY/PRINCIPAL FINDINGS: Taking the "Directed Evolution" approach, viral diversity was increased by pooling an array of serotypes, then passaging the pools under conditions that invite recombination between serotypes. These highly diverse viral pools were then placed under stringent directed selection to generate and identify highly potent agents. ColoAd1, a complex Ad3/Ad11p chimeric virus, was the initial oncolytic virus derived by this novel methodology. ColoAd1, the first described non-Ad5-based oncolytic Ad, is 2-3 logs more potent and selective than the parent serotypes or the most clinically advanced oncolytic Ad, ONYX-015, in vitro. ColoAd1's efficacy was further tested in vivo in a colon cancer liver metastasis xenograft model following intravenous injection and its ex vivo selectivity was demonstrated on surgically-derived human colorectal tumor tissues. Lastly, we demonstrated the ability to arm ColoAd1 with an exogenous gene establishing the potential to impact the treatment of cancer on multiple levels from a single agent. CONCLUSIONS/SIGNIFICANCE: Using the "Directed Evolution" methodology, we have generated ColoAd1, a novel chimeric oncolytic virus. In vitro, this virus demonstrated a >2 log increase in both potency and selectivity when compared to ONYX-015 on colon cancer cells. These results were further supported by in vivo and ex vivo studies. Furthermore, these results have validated this methodology as a new general approach for deriving clinically-relevant, highly potent anti-cancer virotherapies.


Colonic Neoplasms/therapy , Directed Molecular Evolution , Oncolytic Virotherapy , Oncolytic Viruses/physiology , Adenoviridae/physiology , Animals , Cell Line, Tumor , Humans , Liver Neoplasms/secondary , Mice , Neoplasm Transplantation , Oncolytic Viruses/genetics
8.
Mol Ther ; 15(7): 1340-7, 2007 Jul.
Article En | MEDLINE | ID: mdl-17505483

We have developed a one-plasmid regulated gene expression system, pBRES, based on a mifepristone (MFP)-inducible two-plasmid system. The various expression elements of the pBRES system (promoters, 5' and 3' untranslated regions (UTRs), introns, target gene, and polyA sequences) are bounded by restriction enzyme sites so that each module can be conveniently replaced by alternate DNA elements in order to tailor the system for particular tissues, organs, or conditions. There are four possible orientations of the two expression units relative to each other, and insertion of a variety of expression elements and target genes into the four different orientations revealed orientation- and gene-dependent effects on induced and uninduced levels of gene expression. Induced target gene expression from the pBRES system was shown to be comparable to the two-plasmid system and higher than the expression from the cytomegalovirus (CMV) promoter in vivo, while maintaining low uninduced levels of expression. Finally, a pBRES expression cassette was transferred to an adeno-associated virus (AAV) vector and shown to be capable of regulated gene expression in vivo for nearly 1 year.


Gene Expression Regulation/genetics , Plasmids/genetics , Animals , Dependovirus/genetics , Genetic Vectors/genetics , Humans , Interferon-beta/genetics , Interferon-beta/metabolism , Mice , Mice, Inbred C57BL , Transgenes/genetics
9.
J Interferon Cytokine Res ; 26(10): 699-705, 2006 Oct.
Article En | MEDLINE | ID: mdl-17032164

Recombinant interferon-beta (IFN-beta) protein is used successfully for the treatment of multiple sclerosis (MS). Gene therapy might be an alternative approach to overcome drawbacks occurring with IFN-beta protein therapy. A critical issue in developing a new approach is detection of biologically active IFN-beta in preclinical models. The goal of the present study was to determine if Mx1 and IP-10, which are known to be activated after IFN-beta treatment in humans, can be used as biomarkers in mice. In three in vivo experiments, the correlation between different methods of murine IFN-beta (MuIFN-beta) delivery and biomarker induction was studied: (1) bolus protein delivery by intravenous (i.v.) or intramuscular (i.m.) injection, (2) gene-based delivery of IFN- beta by i.m. injection of plasmid DNA, followed by electroporation, and (3) gene-based delivery of IFN-beta by i.m. injection of adenovirus-associated type 1 (AAV1). Short-term induction of Mx1 mRNA and IP-10 was observed after treatment with bolus MuIFN-beta protein. Long-term induction of both biomarkers was observed after IFN-beta plasmid DNA delivery or when AAV1 was used as the vector. The experiments demonstrate that gene-based delivery provides sustained levels of IFN-beta compared with bolus protein injection and that Mx1 RNA and IP-10 can be used to monitor biologically active circulating plasma MuIFN-beta protein in mice.


Chemokines, CXC/biosynthesis , GTP-Binding Proteins/biosynthesis , Gene Transfer Techniques , Interferon-beta/genetics , Animals , Biomarkers/metabolism , Chemokine CXCL10 , Chemokines, CXC/genetics , Dependovirus/genetics , Electroporation , GTP-Binding Proteins/genetics , Genetic Vectors , Injections , Interferon-beta/administration & dosage , Interferon-beta/pharmacology , Kinetics , Male , Mice , Mice, Inbred C57BL , Myxovirus Resistance Proteins , Plasmids/genetics , RNA, Messenger/metabolism
10.
J Interferon Cytokine Res ; 26(7): 449-54, 2006 Jul.
Article En | MEDLINE | ID: mdl-16800783

Experimental allergic encephalomyelitis (EAE) is a model of central nervous system (CNS) inflammation that follows immunization with certain CNS antigens. The course and clinical manifestations of EAE are similar to those of multiple sclerosis (MS) in humans; therefore, EAE has become an accepted animal model to study MS. The purpose of this study was to demonstrate that systemic expression of murine interferon-beta (IFN-beta) (MuIFN-beta), following intramuscular (i.m.) delivery of plasmid DNA encoding MuIFN-beta to the hind limb of mice, is effective in reducing the clinical manifestations of disease in a model of EAE. The results of the study demonstrate that gene-based delivery of MuIFN-beta caused significantly decreased clinical scores compared with delivery of the null vector. A single injection of the MuIFN-beta plasmid was as effective in reducing the severity of the disease as an every other day injection of MuIFN-beta protein.


Encephalomyelitis, Autoimmune, Experimental/therapy , Gene Targeting , Genetic Therapy , Interferon-beta/biosynthesis , Animals , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Interferon-beta/genetics , Mice , Multiple Sclerosis/metabolism , Multiple Sclerosis/therapy , Plasmids/genetics , Plasmids/pharmacology
11.
J Cardiovasc Pharmacol ; 47(4): 587-93, 2006 Apr.
Article En | MEDLINE | ID: mdl-16680073

Morbidity and mortality of peripheral arterial occlusive disease significantly increases with age, often exhibiting more severe disease pathology and decreased treatment effectiveness. Therapeutic angiogenesis with angiogenic growth factors may represent a valuable treatment option for the severely ill, older adult patient population. Aging is considered an independent cardiovascular risk factor, but pathomechanistically it is not well understood. Diminished endothelial nitric oxide (EDNO) production has been considered as a major contributor to the aging process. To investigate the effect of age on postischemic revascularization independent of changes in EDNO, we used endothelial nitric oxide synthase-deficient (ecNOS-KO) mice. We found an age-dependent acceleration in ischemic injury following unilateral femoral artery ligation in these animals compared to C57BL/J6 mice. Postischemic revascularization, quantified by measuring von Willebrand factor expression, was significantly impaired, suggesting that factors other than progressive EDNO deterioration are also involved in the age-dependent severe disease phenotype. Ischemia led to an increase in the expression of vascular endothelial growth factor receptor-2, KDR, in younger ecNOS-KO; however, this increase in KDR expression was absent in the older animals. Lack of increased KDR expression may provide a mechanistic explanation for the severe ischemic injury and perhaps can be used as a clinical marker to identify severe, vascular endothelial growth factor refractory patient population.


Myocardial Ischemia/physiopathology , Nitric Oxide Synthase Type III/deficiency , Vascular Endothelial Growth Factor Receptor-2/biosynthesis , Vascular Endothelial Growth Factor Receptor-2/physiology , Aging , Animals , Endothelium, Vascular/metabolism , Femoral Artery/physiology , Gene Expression/physiology , Hedgehog Proteins , Ischemia/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/physiology , Necrosis , Nitric Oxide/metabolism , Nitric Oxide/physiology , Regional Blood Flow/physiology , Trans-Activators/genetics , Up-Regulation
12.
Am J Physiol Heart Circ Physiol ; 290(3): H1190-8, 2006 Mar.
Article En | MEDLINE | ID: mdl-16272204

Although they are implicated on their own as risk factors for cardiovascular disease, the potential link between nitric oxide (NO) deficiency, ANG II, and vascular stiffening has not been tested before. We evaluated the role of chronic ANG II treatment and NO deficiency, alone and in combination, on aortic stiffness in mice and tested parameters contributing to increases in active or passive components of vascular stiffness, including blood pressure, vascular smooth muscle contractility, and extracellular matrix components. Untreated (control) mice and mice treated with a NO synthase (NOS) inhibitor [N(omega)-nitro-L-arginine methyl ester (L-NAME), 0.5 g/l] were implanted with osmotic minipumps delivering ANG II (500 ng.kg(-1).min(-1)) for 28 days. Aortic stiffness was then measured in vivo by pulse wave velocity (PWV) and ex vivo by load-strain analysis to obtain values of maximal passive stiffness (MPS). Blood pressure and aortic contractility ex vivo were measured. ANG II treatment or NOS inhibition with L-NAME did not independently increase vascular stiffness; however, the combined treatments worked synergistically to increase PWV and MPS. The combined treatments of ANG II + L-NAME also significantly increased aortic wall collagen content while decreasing elastin. These novel results suggest that NO deficiency and ANG II act synergistically to increase aortic stiffness in mice predominantly via changes in aortic wall collagen/elastin ratio.


Angiotensin II/administration & dosage , Aorta/physiology , Muscle, Smooth, Vascular/physiology , NG-Nitroarginine Methyl Ester/administration & dosage , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide/metabolism , Animals , Aorta/drug effects , Drug Combinations , Drug Synergism , Elasticity , Male , Mice , Mice, Inbred C57BL , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle, Smooth, Vascular/drug effects , Stress, Mechanical , Vascular Resistance/drug effects , Vascular Resistance/physiology
13.
Proc Natl Acad Sci U S A ; 102(31): 10999-1004, 2005 Aug 02.
Article En | MEDLINE | ID: mdl-16043715

The genetic loss of endothelial-derived nitric oxide synthase (eNOS) in mice impairs vascular endothelial growth factor (VEGF) and ischemia-initiated blood flow recovery resulting in critical limb ischemia. This result may occur through impaired arteriogenesis, angiogenesis, or mobilization of stem and progenitor cells. Here, we show that after ischemic challenge, eNOS knockout mice [eNOS (-/-)] have defects in arteriogenesis and functional blood flow reserve after muscle stimulation and pericyte recruitment, but no impairment in endothelial progenitor cell recruitment. More importantly, the defects in blood flow recovery, clinical manifestations of ischemia, ischemic reserve capacity, and pericyte recruitment into the growing neovasculature can be rescued by local intramuscular delivery of an adenovirus encoding a constitutively active allele of eNOS, eNOS S1179D, but not a control virus. Collectively, our data suggest that endogenous eNOS-derived NO exerts direct effects in preserving blood flow, thereby promoting arteriogenesis, angiogenesis, and mural cell recruitment to immature angiogenic sprouts.


Ischemia/enzymology , Nitric Oxide Synthase/physiology , Animals , Extremities/blood supply , Gene Expression , Gene Transfer Techniques , Ischemia/pathology , Ischemia/physiopathology , Male , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/blood supply , Neovascularization, Pathologic , Nitric Oxide Synthase/deficiency , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Pericytes/pathology , Regional Blood Flow
15.
BMC Cancer ; 4: 43, 2004 Aug 04.
Article En | MEDLINE | ID: mdl-15294024

BACKGROUND: The Hedgehog (Hh) signaling pathway regulates a variety of developmental processes, including vasculogenesis, and can also induce the expression of pro-angiogenic factors in fibroblasts postnatally. Misregulation of the Hh pathway has been implicated in a variety of different types of cancer, including pancreatic and small-cell lung cancer. Recently a putative antagonist of the pathway, Hedgehog-interacting protein (HIP), was identified as a Hh binding protein that is also a target of Hh signaling. We sought to clarify possible roles for HIP in angiogenesis and cancer. METHODS: Inhibition of Hh signaling by HIP was assayed by measuring the induction of Ptc-1 mRNA in TM3 cells treated with conditioned medium containing Sonic hedgehog (Shh). Angiogenesis was assayed in vitro by EC tube formation on Matrigel. Expression of HIP mRNA was assayed in cells and tissues by Q-RT-PCR and Western blot. HIP expression in human tumors or mouse xenograft tumors compared to normal tissues was assayed by Q-RT-PCR or hybridization of RNA probes to a cancer profiling array. RESULTS: We show that Hedgehog-interacting protein (HIP) is abundantly expressed in vascular endothelial cells (EC) but at low or undetectable levels in other cell types. Expression of HIP in mouse epithelial cells attenuated their response to Shh, demonstrating that HIP can antagonize Hh signaling when expressed in the responding cell, and supporting the hypothesis that HIP blocks Hh signaling in EC. HIP expression was significantly reduced in tissues undergoing angiogenesis, including PC3 human prostate cancer and A549 human lung cancer xenograft tumors, as well as in EC undergoing tube formation on Matrigel. HIP expression was also decreased in several human tumors of the liver, lung, stomach, colon and rectum when compared to the corresponding normal tissue. CONCLUSION: These results suggest that reduced expression of HIP, a naturally occurring Hh pathway antagonist, in tumor neo-vasculature may contribute to increased Hh signaling within the tumor and possibly promote angiogenesis.


Carrier Proteins/metabolism , Endothelium, Vascular/metabolism , Membrane Glycoproteins/metabolism , Neoplasms/genetics , Neovascularization, Pathologic/genetics , Animals , Base Sequence , Carrier Proteins/antagonists & inhibitors , Cell Division/genetics , Down-Regulation , Hedgehog Proteins , Humans , Intracellular Signaling Peptides and Proteins , Leydig Cells/metabolism , Male , Membrane Glycoproteins/antagonists & inhibitors , Membrane Proteins , Mice , Mice, Nude , Molecular Sequence Data , Neoplasms/metabolism , Neovascularization, Physiologic/genetics , Patched Receptors , Patched-1 Receptor , Proteins/genetics , Proteins/metabolism , RNA, Messenger/metabolism , Receptors, Cell Surface , Trans-Activators/metabolism , Transplantation, Heterologous , Tumor Cells, Cultured
16.
Endothelium ; 11(1): 45-57, 2004.
Article En | MEDLINE | ID: mdl-15203878

Gene expression profiling has revealed that cultured vascular endothelial cells (EC) respond to fluid mechanical forces by modulating the mRNA level of a large number of genes. However, differences between the gene arrays and the experimental conditions employed by different researchers make comparison between data sets difficult, and limit the interpretation of the results. Despite these problems, analysis of recent data indicates that the transcriptional response of cultured EC to low-shear disturbed flow conditions similar to those at atherosclerosis-prone areas is distinct from that elicited by atheroprotective high shear laminar flow, providing a molecular basis for the focal nature of atherosclerosis. Many of the genes altered by disturbed flow are involved in key biological processes relevant to atherosclerosis such as inflammation, cell cycle control, apoptosis, thrombosis and oxidative stress. Overall, the gene expression profiling data are consistent with the hypothesis of the hemodynamic etiology of atherosclerotic predilection, viz that at predilected areas in vivo the presence of low shear, non-laminar flow is sufficient to induce a gene expression profile that pre-disposes the endothelium to the initiation and development of atherosclerotic lesions.


Arteriosclerosis/genetics , Endothelial Cells/physiology , Gene Expression Profiling/trends , Gene Expression Regulation/genetics , Regional Blood Flow/physiology , Animals , Arteriosclerosis/physiopathology , Arteriosclerosis/prevention & control , Endothelial Cells/cytology , Humans , Inflammation/genetics , Inflammation/physiopathology , Inflammation/prevention & control , Stress, Mechanical , Thrombosis/genetics , Thrombosis/physiopathology , Thrombosis/prevention & control
17.
Hum Gene Ther ; 15(6): 574-87, 2004 Jun.
Article En | MEDLINE | ID: mdl-15212716

This paper reports the preclinical data that were used to support clinical trials of intracoronary delivery of a replication-incompetent human adenovirus-5 vector encoding human fibroblast growth factor 4 (Ad5FGF4). Using stress-induced myocardial ischemia in pigs, intracoronary injection of Ad5FGF4 resulted in mRNA and protein expression of the transferred gene. Two weeks after gene transfer, regional stress-induced dysfunction and perfusion were ameliorated and improved function persisted for at least 12 weeks. Transgene protein was present in hearts of all animals that received gene transfer but was not found in extracardiac sites. FGF4 was undetectable in samples of plasma obtained at multiple time points after intracoronary delivery of Ad5FGF4. Adenovirus vector DNA was detected in some extracardiac tissues by polymerase chain reaction (PCR) and was dose dependent, occurring primarily after the highest dose delivered (10(12) virus particles [vp]) with much less incidence at 10(11) vp. Histologic evaluation indicated that intracoronary administration of Ad5FGF4 was not associated with abnormal findings in any organ examined. These data provide a rationale for intracoronary delivery of Ad5FGF4 to increase regional cardiac perfusion and function in patients with myocardial ischemia. Based on these preclinical studies, the method does not appear to be associated with major toxic effects.


Adenoviruses, Human/genetics , Coronary Vessels/drug effects , Fibroblast Growth Factors/genetics , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors , Myocardial Ischemia/therapy , Proto-Oncogene Proteins/genetics , Animals , DNA, Viral/analysis , Defective Viruses , Disease Models, Animal , Fibroblast Growth Factor 4 , Fibroblast Growth Factors/metabolism , Mice , Mice, SCID , Myocardial Ischemia/etiology , Neovascularization, Physiologic/drug effects , Polymerase Chain Reaction , Proto-Oncogene Proteins/metabolism , RNA, Messenger/metabolism , Regional Blood Flow/drug effects , Stress, Physiological , Sus scrofa , Transgenes/physiology
18.
J Gene Med ; 6(4): 395-404, 2004 Apr.
Article En | MEDLINE | ID: mdl-15079814

BACKGROUND: Although the transient nature of transgene expression using first-generation adenovirus (Ad) vectors is well known, the exact mechanisms responsible for this phenomenon are uncertain. METHODS: Rats were given intramuscular (i.m.) injections of a first-generation Ad containing the human fibroblast growth factor 4 (hFGF-4) gene driven by the cytomegalovirus (CMV) promoter and enhancer (CMV-PE). The copy number of hFGF-4 mRNA and viral DNA was measured in the same muscles by quantitative RT-PCR and quantitative PCR at times between 1 h and 84 days after virus injection. Quantitative Southern blot analysis for the intact hFGF-4 transcription unit DNA was also performed, and the methylation status of the CMV-PE DNA in the muscle was determined using bisulfite sequencing. RESULTS: The copy number of hFGF-4 mRNA peaked at 6 h then decreased 56-fold by 24 h, and a further 240-fold between days 3 and 28. Although the viral DNA copy number also decreased 23-fold between 6 h and 28 days, the ratio of copies of hFGF-4 mRNA per copy of viral DNA decreased 385-fold over this period. Methylation of the CMV-PE DNA in the muscle at both CpG and non-CpG sites was observed 24 h after virus administration and had increased at day 7. CONCLUSIONS: Decreased transcription associated with extensive methylation of the CMV-PE was the major mechanism responsible for the decrease in transgene mRNA levels. Strategies for preventing transcriptional silencing will be valuable for improving the duration of transgene expression from adenoviral vectors.


Cytomegalovirus/genetics , Gene Silencing , Gene Transfer Techniques , Muscle, Skeletal/physiology , Promoter Regions, Genetic/genetics , Animals , CpG Islands , DNA Methylation , Enhancer Elements, Genetic , Fibroblast Growth Factor 4 , Fibroblast Growth Factors/genetics , Gene Expression Regulation , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Humans , Injections, Intramuscular , Male , Proto-Oncogene Proteins/genetics , Rats , Rats, Sprague-Dawley , Transcription, Genetic
19.
Am J Cardiol ; 92(9B): 24N-31N, 2003 Nov 07.
Article En | MEDLINE | ID: mdl-14615023

Angiogenic gene therapy for stable angina is aimed at promoting new blood vessel formation in the heart, thus providing enhanced cardiac perfusion, symptom relief, increased exercise capacity, improved quality of life, and reduced risk of coronary events. Ad5FGF-4 is a replication-deficient serotype 5 adenovirus encoding the gene for fibroblast growth factor-4 (FGF-4) driven by a cytomegalovirus promoter. In preclinical studies using a pig model of myocardial ischemia, a single intracoronary infusion of Ad5FGF-4 improved cardiac contractile function and regional blood flow in the ischemic region during stress. These effects were apparent after 2 weeks and maintained for > or =12 weeks. Histologic evidence of capillary angiogenesis was observed. FGF-4 gene expression was detected in the heart but not at extracardiac sites. Placebo-controlled trials in humans with chronic stable angina indicate that Ad5FGF-4 increases treadmill exercise duration and improves stress-related ischemia measured by perfusion sestamibi single-photon emission computed tomography. More patients receiving Ad5FGF-4 than placebo reported complete resolution of their angina and no nitroglycerin use. Ad5FGF-4 gene therapy was well tolerated. The administration procedure did not cause any adverse events, although mild, transient fever, a transient modest fall in platelet count, and a transient mild elevation in hepatic enzymes and uric acid levels occurred in a few patients. This adverse event profile concurs with other adenoviral gene trials. There was no evidence of myocarditis, retinal neovascularization, or angioma formation. FGF-4 was not detected in venous blood. Larger clinical trials will assess Ad5FGF-4 with regard to cardiovascular prognosis, symptom relief, and safety profile in patients with chronic stable angina.


Adenoviridae/genetics , Coronary Disease/therapy , Fibroblast Growth Factors/therapeutic use , Genetic Therapy/methods , Animals , Coronary Circulation/physiology , Exercise Test , Fibroblast Growth Factors/administration & dosage , Genetic Vectors , Humans , Neovascularization, Physiologic , Ventricular Function, Left
20.
FASEB J ; 17(1): 100-2, 2003 Jan.
Article En | MEDLINE | ID: mdl-12475908

Previous studies have shown that fibroblast growth factor (FGF)-1, FGF-2, and FGF-5 induce therapeutic angiogenesis. Here, we investigated the potential of FGF-4 for therapeutic neovascularization in comparison to vascular endothelial growth factor (VEGF), using adenoviral gene transfer in a novel rabbit hind limb ischemia model, with ischemia restricted to the calf. Magnetic resonance imaging and a modified Miles assay showed that both AdFGF-4 and AdVEGF given intramuscularly (i.m.) resulted in increases in vascular permeability and edema in transduced muscles 6 days after the gene transfer. In contrast, recombinant FGF-4 protein injected in the rabbit skin did not induce acute vascular permeability. Injections (i.m.) of AdFGF-4 and AdVEGF, but not intra-arterially administered AdVEGF, increased collateral growth, popliteal blood flow, and muscle perfusion compared with controls. The angiogenesis response consisted mainly of the enlargement of pre-existing vessels rather than an increase in capillary density. Adenoviral FGF-4 overexpression up-regulated endogenous VEGF, which may explain many of the effects thought to be specific for VEGF such as the increase in vascular permeability. This study demonstrates for the first time that FGF-4 induces vascular permeability, therapeutic angiogenesis, and arteriogenesis comparable to that of VEGF and could be useful for the treatment of peripheral vascular disease.


Arteries/growth & development , Capillary Permeability , Fibroblast Growth Factors/genetics , Ischemia/therapy , Neovascularization, Physiologic , Proto-Oncogene Proteins/genetics , Adenoviridae/genetics , Animals , Capillaries/cytology , Capillaries/growth & development , Cell Division/drug effects , Cells, Cultured , Collateral Circulation , Edema/etiology , Edema/pathology , Endothelial Growth Factors/genetics , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Fibroblast Growth Factor 4 , Fibroblast Growth Factors/pharmacology , Genetic Vectors , Hindlimb/blood supply , Intercellular Signaling Peptides and Proteins/genetics , Ischemia/pathology , Lymphokines/genetics , Muscle, Skeletal/blood supply , Proto-Oncogene Proteins/pharmacology , Rabbits , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
...