1
|
Stewart DJ, Gianchetti A, Byrnes D, Dittrich HC, Thorne B, Manza LL, Reinhardt RR. Safety and biodistribution of XC001 (encoberminogene rezmadenovec) gene therapy in rats: a potential therapy for cardiovascular diseases. Gene Ther 2024; 31:45-55. [PMID: 37592080 DOI: 10.1038/s41434-023-00416-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 07/19/2023] [Accepted: 07/27/2023] [Indexed: 08/19/2023]
Abstract
Adenovirus-mediated gene therapy holds promise for the treatment of cardiovascular diseases such as refractory angina. However, potential concerns around immunogenicity and vector dissemination from the target injected tissue require evaluation. This study was undertaken to evaluate the safety and biodistribution of XC001, a replication-deficient adenovirus serotype 5 vector expressing multiple isoforms of human vascular endothelial growth factor (VEGF), following direct administration into normal rat myocardium. Animals received the buffer formulation or increasing doses of XC001 (1 × 107, 2.5 × 108 or 2.5 × 109 viral particles). Based on in-life parameters (general health, body weights, clinical pathology, serum cardiac troponin I, plasma VEGF, and gross necropsy), there were no findings of clinical concern. On Day 8, intramyocardial administration of XC001 was associated with dose-related, left ventricular myocardial inflammation at injection sites, resolving by Day 30. XC001 DNA was not detected in blood at any time but was present at Day 8 around the site of injection and to a much lesser extent in the spleen, liver, and lungs, persisting at low levels in the heart and spleen until at least Day 91. These findings demonstrate that intramyocardial injection of XC001 is supported for use in human studies.
Collapse
Affiliation(s)
- Duncan J Stewart
- Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute and the Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
| | | | | | | | | | - Linda L Manza
- Pharmaron (San Diego) Lab Services LLC, San Diego, CA, USA
| | | |
Collapse
|
2
|
Damle SR, Krzyzanowska AK, Korsun MK, Morse KW, Gilbert S, Kim HJ, Boachie-Adjei O, Rawlins BA, van der Meulen MCH, Greenblatt MB, Hidaka C, Cunningham ME. Inducing Angiogenesis in the Nucleus Pulposus. Cells 2023; 12:2488. [PMID: 37887332 PMCID: PMC10605635 DOI: 10.3390/cells12202488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 10/28/2023] Open
Abstract
Bone morphogenetic protein (BMP) gene delivery to Lewis rat lumbar intervertebral discs (IVDs) drives bone formation anterior and external to the IVD, suggesting the IVD is inhospitable to osteogenesis. This study was designed to determine if IVD destruction with a proteoglycanase, and/or generating an IVD blood supply by gene delivery of an angiogenic growth factor, could render the IVD permissive to intra-discal BMP-driven osteogenesis and fusion. Surgical intra-discal delivery of naïve or gene-programmed cells (BMP2/BMP7 co-expressing or VEGF165 expressing) +/- purified chondroitinase-ABC (chABC) in all permutations was performed between lumbar 4/5 and L5/6 vertebrae, and radiographic, histology, and biomechanics endpoints were collected. Follow-up anti-sFlt Western blotting was performed. BMP and VEGF/BMP treatments had the highest stiffness, bone production and fusion. Bone was induced anterior to the IVD, and was not intra-discal from any treatment. chABC impaired BMP-driven osteogenesis, decreased histological staining for IVD proteoglycans, and made the IVD permissive to angiogenesis. A soluble fragment of VEGF Receptor-1 (sFlt) was liberated from the IVD matrix by incubation with chABC, suggesting dysregulation of the sFlt matrix attachment is a possible mechanism for the chABC-mediated IVD angiogenesis we observed. Based on these results, the IVD can be manipulated to foster vascular invasion, and by extension, possibly osteogenesis.
Collapse
Affiliation(s)
- Sheela R. Damle
- HSS Research Institute, Hospital for Special Surgery, 515 E 71st Street, New York, NY 10021, USA
| | - Agata K. Krzyzanowska
- HSS Research Institute, Hospital for Special Surgery, 515 E 71st Street, New York, NY 10021, USA
| | - Maximilian K. Korsun
- HSS Research Institute, Hospital for Special Surgery, 515 E 71st Street, New York, NY 10021, USA
| | - Kyle W. Morse
- HSS Research Institute, Hospital for Special Surgery, 515 E 71st Street, New York, NY 10021, USA
| | - Susannah Gilbert
- HSS Research Institute, Hospital for Special Surgery, 515 E 71st Street, New York, NY 10021, USA
| | - Han Jo Kim
- HSS Research Institute, Hospital for Special Surgery, 515 E 71st Street, New York, NY 10021, USA
- Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Oheneba Boachie-Adjei
- HSS Research Institute, Hospital for Special Surgery, 515 E 71st Street, New York, NY 10021, USA
- Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Bernard A. Rawlins
- HSS Research Institute, Hospital for Special Surgery, 515 E 71st Street, New York, NY 10021, USA
- Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Marjolein C. H. van der Meulen
- HSS Research Institute, Hospital for Special Surgery, 515 E 71st Street, New York, NY 10021, USA
- Meinig School of Biomedical Engineering and Sibley School of Mechanical & Aerospace Engineering, Cornell University, Ithaca, NY 14853, USA
| | | | - Chisa Hidaka
- HSS Research Institute, Hospital for Special Surgery, 515 E 71st Street, New York, NY 10021, USA
- Department of Genetic Medicine and Belfer Gene Therapy Core Facility, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Matthew E. Cunningham
- HSS Research Institute, Hospital for Special Surgery, 515 E 71st Street, New York, NY 10021, USA
- Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| |
Collapse
|
3
|
Crystal RG. My Pathway to Gene Therapy. Hum Gene Ther 2020; 31:273-282. [DOI: 10.1089/hum.2020.29112.rgc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Ronald G. Crystal
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| |
Collapse
|
4
|
Guo GL, Sun LQ, Sun MH, Xu HM. LncRNA SLC8A1-AS1 protects against myocardial damage through activation of cGMP-PKG signaling pathway by inhibiting SLC8A1 in mice models of myocardial infarction. J Cell Physiol 2018; 234:9019-9032. [PMID: 30378115 DOI: 10.1002/jcp.27574] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 09/17/2018] [Indexed: 12/13/2022]
Abstract
Extensive investigations into long noncoding RNAs (lncRNAs) in various diseases and cancers, including acute myocardial infarction (AMI) have been conducted. The current study aimed to investigate the role of lncRNA solute carrier family 8 member A1 antisense RNA 1 (SLC8A1-AS1) in myocardial damage by targeting solute carrier family 8 member A1 (SLC8A1) via cyclic guanosine 3',5'-monophosphate-protein kinase G (cGMP-PKG) signaling pathway in AMI mouse models. Differentially expressed lncRNA in AMI were initially screened and target relationship between lncRNA SLC8A1-AS1 and SLC8A1 was then verified. Infarct size, levels of inflammatory factors, biochemical indicators, and the positive expression of the SLC8A1 protein in AMI were subsequently determined. The expression of SLC8A1-AS1, SLC8A1, PKG1, PKG2, atrial natriuretic peptide, and brain natriuretic peptide was detected to assess the effect of SLC8A1-AS1 on SLC8A1 and cGMP-PKG. The respective contents of superoxide dismutase, lactate dehydrogenase (LDH), and malondialdehyde (MDA) were detected accordingly. Microarray data GSE66360 provided evidence indicating that SLC8A1-AS1 was poorly expressed in AMI. SLC8A1 was verified to be a target gene of lncRNA SLC8A1-AS1. SLC8A1-AS1 upregulation decreased levels of left ventricular end-systolic diameter, -dp/ dt max , interleukin 1β (IL-1β), IL-6, transforming growth factor α, nitric oxide, inducible nitric-oxide synthase, endothelial nitric-oxide synthase, infarct size, LDH activity and MDA content, and increased IL-10, left ventricular end-diastolic pressure and + dp/ dt max . Furthermore, the overexpression of SLC8A1-AS1 was noted to elicit an inhibitory effect on the cGMP-PKG signaling pathway via SLC8A1. In conclusion, lncRNA SLC8A1-AS1, by downregulating SLC8A1 and activating the cGMP-PKG signaling pathway, was observed to alleviate myocardial damage, inhibit the release of proinflammatory factors and reduce infarct size, ultimately protecting against myocardial damage.
Collapse
Affiliation(s)
- Gong-Liang Guo
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Li-Qun Sun
- Department of Pediatrics, The First Hospital of Jilin University, Changchun, China
| | - Mei-Hua Sun
- Department of Pediatrics, The First Hospital of Jilin University, Changchun, China
| | - Hai-Ming Xu
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
5
|
Maslov M, Foianini S, Lovich M. Delivery of drugs, growth factors, genes and stem cells via intrapericardial, epicardial and intramyocardial routes for sustained local targeted therapy of myocardial disease. Expert Opin Drug Deliv 2017; 14:1227-1239. [PMID: 28276968 DOI: 10.1080/17425247.2017.1292249] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Local myocardial delivery (LMD) of therapeutic agents is a promising strategy that aims to treat various myocardial pathologies. It is designed to deliver agents directly to the myocardium and minimize their extracardiac concentrations and side effects. LMD aims to enhance outcomes of existing therapies by broadening their therapeutic window and to utilize new agents that could not be otherwise be implemented systemically. Areas covered: This article provides a historical overview of six decades LMD evolution in terms of the approaches, including intrapericardial, epicardial, and intramyocardial delivery, and the wide array of classes of agents used to treat myocardial pathologies. We examines delivery of pharmaceutical compounds, targeted gene transfection and cell implantation techniques to produce therapeutic effects locally. We outline therapeutic indications, successes and failures as well as technical approaches for LMD. Expert opinion: While LMD is more complicated than conventional oral or intravenous administration, given recent advances in interventional cardiology, it is safe and may provide better therapeutic outcomes. LMD is complex as many factors impact pharmacokinetics and biologic result. The choice between routes of LMD is largely driven not only by the myocardial pathology but also by the nature and physicochemical properties of the therapeutic agents.
Collapse
Affiliation(s)
- Mikhail Maslov
- a Department of Anesthesiology, Pain Medicine and Critical Care , Steward St. Elizabeth's Medical Center/Tufts University School of Medicine , Boston , MA , USA
| | - Stephan Foianini
- a Department of Anesthesiology, Pain Medicine and Critical Care , Steward St. Elizabeth's Medical Center/Tufts University School of Medicine , Boston , MA , USA
| | - Mark Lovich
- a Department of Anesthesiology, Pain Medicine and Critical Care , Steward St. Elizabeth's Medical Center/Tufts University School of Medicine , Boston , MA , USA
| |
Collapse
|
6
|
Hadjizadeh A, Ghasemkhah F, Ghasemzaie N. Polymeric Scaffold Based Gene Delivery Strategies to Improve Angiogenesis in Tissue Engineering: A Review. POLYM REV 2017. [DOI: 10.1080/15583724.2017.1292402] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Afra Hadjizadeh
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Farzaneh Ghasemkhah
- Institute of Nanotechnology, Amirkabir University of Technology, Tehran, Iran
| | - Niloofar Ghasemzaie
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| |
Collapse
|
7
|
Crystal RG. Adenovirus: the first effective in vivo gene delivery vector. Hum Gene Ther 2014; 25:3-11. [PMID: 24444179 DOI: 10.1089/hum.2013.2527] [Citation(s) in RCA: 251] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Ronald G Crystal
- Department of Genetic Medicine, Weill Cornell Medical College , New York, NY 10065
| |
Collapse
|
8
|
Kaminsky SM, Rosengart TK, Rosenberg J, Chiuchiolo MJ, Van de Graaf B, Sondhi D, Crystal RG. Gene therapy to stimulate angiogenesis to treat diffuse coronary artery disease. Hum Gene Ther 2014; 24:948-63. [PMID: 24164242 DOI: 10.1089/hum.2013.2516] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Cardiac gene therapy offers a strategy to treat diffuse coronary artery disease (CAD), a disorder with no therapeutic options. The use of genes to revascularize the ischemic myocardium has been the focus of two decades of preclinical research with a variety of angiogenic mediators, including vascular endothelial growth factor, fibroblast growth factor, hepatocyte growth factor, and others encoded by DNA plasmids or adenovirus vectors. The multifaceted challenge for developing efficient induction of collateral vessels in the ischemic heart requires a choice for route of delivery, dosing level, a relevant animal model, duration of treatment, and assessment of phenotype for efficacy. Overall, studies of gene therapy for ischemia in experimental models are very encouraging, with clear evidence of safety and efficacy, strongly supporting the concept that gene therapy to induce angiogenesis is a viable therapeutic approach for CAD. Clinical studies of cardiac gene therapy with angiogenic factors have added substantially to the evidence for efficacy, but definitive studies have not yet led to commercial approval. This review provides the general concepts for angiogenesis-based therapeutic approaches for diffuse CAD and summarizes the results from key studies in the field with recommendations for refinement to a successful product design and evaluation.
Collapse
Affiliation(s)
- Stephen M Kaminsky
- 1 Department of Genetic Medicine, Weill Cornell Medical College , New York, NY 10065
| | | | | | | | | | | | | |
Collapse
|
9
|
Katz MG, Fargnoli AS, Williams RD, Bridges CR. Gene therapy delivery systems for enhancing viral and nonviral vectors for cardiac diseases: current concepts and future applications. Hum Gene Ther 2014; 24:914-27. [PMID: 24164239 DOI: 10.1089/hum.2013.2517] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Gene therapy is one of the most promising fields for developing new treatments for the advanced stages of ischemic and monogenetic, particularly autosomal or X-linked recessive, cardiomyopathies. The remarkable ongoing efforts in advancing various targets have largely been inspired by the results that have been achieved in several notable gene therapy trials, such as the hemophilia B and Leber's congenital amaurosis. Rate-limiting problems preventing successful clinical application in the cardiac disease area, however, are primarily attributable to inefficient gene transfer, host responses, and the lack of sustainable therapeutic transgene expression. It is arguable that these problems are directly correlated with the choice of vector, dose level, and associated cardiac delivery approach as a whole treatment system. Essentially, a delicate balance exists in maximizing gene transfer required for efficacy while remaining within safety limits. Therefore, the development of safe, effective, and clinically applicable gene delivery techniques for selected nonviral and viral vectors will certainly be invaluable in obtaining future regulatory approvals. The choice of gene transfer vector, dose level, and the delivery system are likely to be critical determinants of therapeutic efficacy. It is here that the interactions between vector uptake and trafficking, delivery route means, and the host's physical limits must be considered synergistically for a successful treatment course.
Collapse
Affiliation(s)
- Michael G Katz
- Sanger Heart and Vascular Institute , Cannon Research Center, Carolinas HealthCare System, Charlotte, NC 28203
| | | | | | | |
Collapse
|
10
|
Rosengart TK, Bishawi MM, Halbreiner MS, Fakhoury M, Finnin E, Hollmann C, Shroyer AL, Crystal RG. Long-term follow-up assessment of a phase 1 trial of angiogenic gene therapy using direct intramyocardial administration of an adenoviral vector expressing the VEGF121 cDNA for the treatment of diffuse coronary artery disease. Hum Gene Ther 2012; 24:203-8. [PMID: 23137122 DOI: 10.1089/hum.2012.137] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
On the basis of studies in experimental animals demonstrating that AdVEGF121, an E1(-)E3(-) serotype 5 adenovirus coding the 121 isoform of vascular endothelial growth factor (VEGF), could mediate the generation of new blood vessels and reverse coronary ischemia, a clinical study of direct myocardial administration of AdVEGF121 was initiated in patients with late-stage, diffuse coronary artery disease. This study provides long-term (median, 11.8 years) follow-up on these patients. From 1997 to 1999, AdVEGF121 was administered by direct myocardial injection to an area of reversible ischemia in 31 patients with severe coronary disease, either as an adjunct to conventional coronary artery bypass grafting (group A) or as minimally invasive sole (MIS) therapy, using a minithoracotomy (group B). There was no control group; the study participants served as the control subjects. The 5- and 10-year survival was 10 of 15 (67%) and 6 of 15 (40%) for the group A patients, and 11 of 16 (69%) and 5 of 16 (31%) for group B sole therapy patients, respectively. In comparison, maximal medical therapy in comparable groups in the literature have a 3- to 5-year survival rate of 52 to 59%. For the survivors, the angina score for group A was 3.4±0.5 at time 0 and 1.9±1.0 at last follow-up, and for group B it was 3.4±0.6 and 2.0±1.1, respectively. The incidences of malignancy and retinopathy were no greater than that expected for the age-matched general population. We conclude that adenovirus-mediated VEGF direct myocardial administration to patients with severe coronary artery disease is safe, and future larger trials are warranted to assess efficacy.
Collapse
Affiliation(s)
- Todd K Rosengart
- Division of Cardiothoracic Surgery, Department of Surgery, Stony Brook University Medical Center, Stony Brook, NY 11794, USA
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Nonviral delivery of genetic medicine for therapeutic angiogenesis. Adv Drug Deliv Rev 2012; 64:40-52. [PMID: 21971337 DOI: 10.1016/j.addr.2011.09.005] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Revised: 06/29/2011] [Accepted: 09/18/2011] [Indexed: 01/08/2023]
Abstract
Genetic medicines that induce angiogenesis represent a promising strategy for the treatment of ischemic diseases. Many types of nonviral delivery systems have been tested as therapeutic angiogenesis agents. However, their delivery efficiency, and consequently therapeutic efficacy, remains to be further improved, as few of these technologies are being used in clinical applications. This article reviews the diverse nonviral gene delivery approaches that have been applied to the field of therapeutic angiogenesis, including plasmids, cationic polymers/lipids, scaffolds, and stem cells. This article also reviews clinical trials employing nonviral gene therapy and discusses the limitations of current technologies. Finally, this article proposes a future strategy to efficiently develop delivery vehicles that might be feasible for clinically relevant nonviral gene therapy, such as high-throughput screening of combinatorial libraries of biomaterials.
Collapse
|
12
|
Muona K, Mäkinen K, Hedman M, Manninen H, Ylä-Herttuala S. 10-year safety follow-up in patients with local VEGF gene transfer to ischemic lower limb. Gene Ther 2011; 19:392-5. [DOI: 10.1038/gt.2011.109] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
13
|
Zeng R, Chen YC, Zeng Z, Liu WQ, Liu XX, Liu R, Qiang O, Li X. Different angiogenesis effect of mini-TyrRS/mini-TrpRS by systemic administration of modified siRNAs in rats with acute myocardial infarction. Heart Vessels 2010; 25:324-32. [DOI: 10.1007/s00380-009-1200-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2009] [Accepted: 08/27/2009] [Indexed: 11/29/2022]
|
14
|
Akhavani MA, Sivakumar B, Paleolog EM, Kang N. Angiogenesis and plastic surgery. J Plast Reconstr Aesthet Surg 2008; 61:1425-37. [PMID: 18835232 DOI: 10.1016/j.bjps.2008.05.041] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2008] [Revised: 03/27/2008] [Accepted: 05/29/2008] [Indexed: 01/23/2023]
Abstract
SUMMARY Angiogenesis, the formation of new blood vessels from an existing vascular bed, is a normal physiological process which also underpins many--apparently unrelated--pathological states. It is an integral factor in determining the success or failure of many procedures in plastic and reconstructive surgery. As a result, the ability to control the process would be of great therapeutic benefit. To appreciate the potential benefits and limitations of recent advances in our understanding of angiogenesis, it is important to comprehend the basic physiology of blood vessel formation. This review aims to summarise current knowledge of the way in which angiogenesis is controlled and to look at how disordered vessel development results in pathology relevant to plastic surgery. Through this we hope to provide a comprehensive overview of the recent advances in angiogenesis as they relate to plastic surgery, particularly the promotion of flap survival, tendon healing, nerve regeneration, fracture healing and ulcer treatments.
Collapse
|
15
|
Shim WSN, Li W, Zhang L, Li S, Ong HC, Song IC, Bapna A, Ge R, Lim YT, Chuah SC, Sim EKW, Wong P. Angiopoietin-1 promotes functional neovascularization that relieves ischemia by improving regional reperfusion in a swine chronic myocardial ischemia model. J Biomed Sci 2006; 13:579-91. [PMID: 16547766 DOI: 10.1007/s11373-006-9082-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2005] [Accepted: 02/22/2006] [Indexed: 10/24/2022] Open
Abstract
This study investigates the long-term angiogenic effects of ANG-1 and VEGF in a swine chronic myocardial ischemia model. Four-weeks after gradual occlusion of the left circumflex coronary artery by ameroid constrictor, animals were injected with recombinant adenoviral vectors carrying either human ANG-1 (n=9), human VEGF(165) (n=10) or empty vector (n=7) into the left ventricle free wall supplied by the constricted artery. Left ventricular perfusion in animals that received AdANG-1 (3.25+/-0.16 ml/min/g, p<0.05) recovered robustly 4 weeks after gene transfer while ischemia persisted in the AdVEGF (1.09+/-0.13 ml/min/g) and empty vector (1.20+/-0.03 ml/min/g) groups. Microvascular densities in the left ventricles of animals that received AdANG-1 (19.61+/-1.76/0.572 mm(2) myocardial tissue, p<0.05) and AdVEGF (18.17+/-1.43/0.572 mm(2) myocardial tissue, p<0.05) were significantly higher than animals that received empty vector (13.53+/-0.92/0.572 mm(2) myocardial tissue) 12 weeks after gene transfer. ANG-1, but not VEGF, contributed to enhanced regional perfusion by increasing arteriolar density (1.9+/-0.4/0.572 mm(2) myocardial tissue vs. 0.7+/-0.2/0.572 mm(2) myocardial tissue, p<0.05) of large-sized (50-100 microm) arterioles. These data demonstrate that gene transfer of ANG-1 and VEGF enhances angiogenesis, but ANG-1 promotes sustained improvement of ventricular perfusion that expedites recovery of ischemic myocardium via arteriogenesis.
Collapse
Affiliation(s)
- Winston S N Shim
- Research and Development Unit, National Heart Center, 17 Third Hospital Avenue, Singapore 168752, Singapore.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Kikuchi K, McDonald AD, Sasano T, Donahue JK. Targeted modification of atrial electrophysiology by homogeneous transmural atrial gene transfer. Circulation 2005; 111:264-70. [PMID: 15642761 DOI: 10.1161/01.cir.0000153338.47507.83] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Safe and effective myocardial gene transfer remains elusive. Heterogeneous ventricular gene delivery has been achieved in small mammals but generally with methods not readily transferable to the clinic. Atrium-specific gene transfer has not yet been reported. We hypothesized that homogeneous atrial gene transfer could be achieved by direct application of adenoviral vectors to the epicardial surface, use of poloxamer gel to increase virus contact time, and mild trypsinization to increase virus penetration. METHODS AND RESULTS We "painted" recombinant adenovirus encoding the reporter gene Escherichia coli beta-galactosidase directly onto porcine atria. Investigational variables included poloxamer use, trypsin concentration, and safety. Using the painting method, we modified the atrial phenotype with an adenovirus expressing HERG-G628S, a long-QT-syndrome mutant. Our results showed that application of virus with poloxamer alone resulted in diffuse epicardial gene transfer with negligible penetration into the myocardium. Dilute trypsin concentrations allowed complete transmural gene transfer. After trypsin exposure, echocardiographic left atrial diameter did not change. Left atrial function decreased on postoperative day 3 but returned to baseline by day 7. Tissue tensile strength was affected only in the 1% trypsin group. HERG-G628S gene transfer prolonged atrial action potential duration and refractory period without affecting ventricular electrophysiology. CONCLUSIONS We show complete transmural atrial gene transfer by this novel painting method. Adaptation of the method could allow application to other tissue targets. Use with functional proteins in the atria could cure or even prevent diseases such as atrial fibrillation or sinus node dysfunction.
Collapse
Affiliation(s)
- Kan Kikuchi
- Johns Hopkins University School of Medicine, Baltimore, Md 21205, USA
| | | | | | | |
Collapse
|
17
|
Gurunluoglu R, Meirer R, Shafighi M, Huemer GM, Yilmaz B, Piza-Katzer H. Gene therapy with adenovirus-mediated VEGF enhances skin flap prefabrication. Microsurgery 2005; 25:433-41. [PMID: 16032725 DOI: 10.1002/micr.20142] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
We investigated the feasibility in rats of enhancing skin-flap prefabrication with subdermal injections of adenovirus-encoding vascular endothelial growth factor (Ad-VEGF). The left saphenous vascular pedicle was used as a source for vascular induction. A peninsular abdominal flap (8 x 8 cm) was elevated as distally based, keeping the epigastric vessels intact on both sides. After the vascular pedicle was tacked underneath the abdominal flap, 34 rats were randomly divided into three groups according to treatment protocol. The implantation site around the pedicle was injected with Ad-VEGF in group I (n = 10), with adenovirus-encoding green fluorescent protein (Ad-GFP) in control group I (n = 14), and with saline in control group II (n = 10). All injections were given subdermally at four points around the implanted vessel by an individual blinded to the treatment protocol. The peninsular flap was sutured in its place, and 4 weeks later, an abdominal island flap based solely on the implanted vessels was elevated. The prefabricated island flap was sutured back, and flap viability was evaluated on day 7. Skin specimens were stained with hematoxylin and eosin for histological evaluation. In two rats from each group, microangiography was performed to visualize the vascularity of the prefabricated flaps. There was a significant increase in survival of prefabricated flaps in the Ad-VEGF group compared to the control groups: Ad-VEGF, 88.9 +/- 6.1% vs. Ad-GFP, 65.6 +/- 9.4% (P < 0.05) and saline, 56.0 +/- 3.4% (P < 0.05). Sections from four prefabricated flaps treated with Ad-GFP revealed multiple sites of shiny deposits of green fluorescent protein around the area of local administration 1 day and 3 weeks after gene therapy. Histological examination done under high-power magnification (x400) with a light microscope revealed increased vascularity and mild inflammation surrounding the implanted vessel in all groups. However, we were unable to demonstrate any significant quantitative difference with respect to vascularity and inflammatory infiltrates in prefabricated flaps treated with Ad-VEGF compared with controls. Microangiographic studies showed increased vascularity around the implanted pedicle, which was similar in all groups. However, vascularization was distributed in a larger area in the prefabricated flaps treated with Ad-VEGF. In this study, the authors demonstrated that adenovirus-mediated VEGF gene therapy increased the survival of prefabricated flaps, suggesting that it may allow prefabrication of larger flaps and have the potential to reduce the time required for flap maturation.
Collapse
Affiliation(s)
- Raffi Gurunluoglu
- Department of Plastic, Reconstructive, and Aesthetic Surgery, Acibadem Hospital, Kadikoy, Istanbul, Turkey.
| | | | | | | | | | | |
Collapse
|
18
|
Katsube K, Bishop AT, Friedrich PF. Transduction of rabbit saphenous artery: a comparison of naked DNA, liposome complexes, and adenovirus vectors. J Orthop Res 2004; 22:1290-5. [PMID: 15475211 DOI: 10.1016/j.orthres.2004.05.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The methods and efficiency of gene transfer into rabbit saphenous artery were examined in this study. The purpose was to develop an animal model capable of evaluating the use of angiogenic gene therapy to revascularize necrotic bone more rapidly and completely than by surgical implantation of blood vessels alone. The success of transduction using adenovirus vectors, liposome/DNA complexes, and naked DNA was evaluated with delivery to both intra-luminal and adventitial sites. Intra-luminal and adventitial (extra-luminal) application was used for the viral and liposome methods. Naked DNA was evaluated only in the intra-luminal site, based upon previous reports. Relative transduction success was expressed as the percentage of total cells with beta-galactosidase activity. A 20-mm length of saphenous artery exposed surgically was targeted for lacZ gene transfer. Two days after transduction, the arteries were harvested and stained with X-gal for beta-galactosidase activity. The percentage of endothelial, media and adventitial cells with beta-galactosidase activity was determined. Intra-arterial injection of adenovirus vector transduced the largest amount of cells in all three areas of the vessel (endothelium, media and adventitia). The adenovirus vectors when applied to the adventitia only transduced adventitial cells. Following intra-arterial injection of liposome/DNA complexes transduction was detected only in endothelium. Extra-luminal liposome and intra-arterial naked DNA delivery resulted in no detectable gene transfer. Intra-arterial delivery of an adenovirus vector would likely provide optimal gene transfer for possible angiogenic gene therapy.
Collapse
Affiliation(s)
- Kousuke Katsube
- Department of Orthopedic Surgery, Microvascular Research Laboratory, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | | | | |
Collapse
|
19
|
Degabriele NM, Griesenbach U, Sato K, Post MJ, Zhu J, Williams J, Jeffery PK, Geddes DM, Alton EWFW. Critical appraisal of the mouse model of myocardial infarction. Exp Physiol 2004; 89:497-505. [PMID: 15131069 DOI: 10.1113/expphysiol.2004.027276] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In order to critically evaluate the utility of a mouse model of myocardial infarction (MI) for therapeutic studies, we investigated survival, haemodynamic measurements and histopathology in mice with an occluding suture placed at one of three distinct sites along the left anterior descending coronary artery. The suture was placed at the atrioventricular juncture (High), or at two sites more distally towards the base (Middle and Low). In the High group, only 33% of animals survived 7 days after MI (P < 0.05 compared to all other groups). Only the Middle group had significantly reduced haemodynamics compared to sham-operated animals (maximum left ventricular pressure: 55.9 +/- 3.5 versus 80.8 +/- 5.1 mmHg, maximum change in pressure over time : 2003 +/- 172 versus 4402 +/- 491, P < 0.01). Histological examination showed morphological changes in all MI groups. The Middle group had larger lesions than the Low group (P < 0.05). Lesions in the anterior and lateral walls correlated, albeit weakly, with cardiac function. Power calculations indicated that, despite a certain amount of intragroup variation, the Middle Suture model may be useful for therapeutic studies to assess the effects of treatment on cardiac function and overall lesion size.
Collapse
Affiliation(s)
- Naomi M Degabriele
- Department of Gene Therapy, Faculty of Medicine at the National Heart & Lung Institute, Imperial College, London, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Rissanen TT, Rutanen J, Ylä-Herttuala S. Gene Transfer for Therapeutic Vascular Growth in Myocardial and Peripheral Ischemia. ADVANCES IN GENETICS 2004; 52:117-64. [PMID: 15522734 DOI: 10.1016/s0065-2660(04)52004-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Therapeutic vascular growth in the treatment of peripheral and myocardial ischemia has not yet fulfilled its expectations in clinical trials. Randomized, double-blinded placebo-controlled trials have predominantly shown the safety and feasibility but not the clear-cut clinically relevant efficacy of angiogenic gene or recombinant growth factor therapy. It is likely that growth factor levels achieved with single injections of recombinant protein or naked plasmid DNA are too low to induce any relevant angiogenic effects. Also, the route of administration of gene transfer vectors has not been optimal in many cases leading to low gene-transfer efficacy. Animal experiments using intramuscular or intramyocardial injections of adenovirus encoding vascular endothelial growth factor (VEGF, VEGF-A), the mature form of VEGF-D, and fibroblast growth factors (FGF-1, -2, and -4) have shown high angiogenic efficacy. Adenoviral overexpression of VEGF receptor-2 ligands, VEGF-A and the mature form of VEGF-D, enlarge the preexisting capillaries in skeletal muscle and myocardium via nitric oxide(NO)-mediated mechanisms and via proliferation of both endothelial cells and pericytes, resulting in markedly increased tissue perfusion. VEGF also enhances collateral growth, which is probably secondary to increased peripheral capillary blood flow and shear stress. As a side effect of VEGF overexpression and rapid microvessel enlargement, vascular permeability increases and may result in substantial tissue edema and pericardial effusion in the heart. Because of the transient adenoviral gene expression, the majority of angiogenic effects and side effects return to baseline by 2 weeks after the gene transfer. In contrast, VEGF overexpression lasting over 4 weeks has been shown to induce the growth of a persistent vascular network in preclinical models. To improve efficacy, the choice of the vascular growth factor, gene transfer vector, and route of administration should be optimized in future clinical trials. This review is focused on these issues.
Collapse
Affiliation(s)
- Tuomas T Rissanen
- Department of Biotechnology and Molecular Medicine, A. I. Virtanen Institute, Kuopio University, Kuopio, Finland
| | | | | |
Collapse
|
21
|
Chu D, Sullivan CC, Weitzman MD, Du L, Wolf PL, Jamieson SW, Thistlethwaite PA. Direct comparison of efficiency and stability of gene transfer into the mammalian heart using adeno-associated virus versus adenovirus vectors. J Thorac Cardiovasc Surg 2003; 126:671-9. [PMID: 14502138 DOI: 10.1016/s0022-5223(03)00082-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
OBJECTIVE Recent gene therapy strategies have relied on the use of adenovirus or plasmid as vehicles for gene delivery to the heart. These approaches have been limited by low transduction frequencies and transient transgene expression. We sought to determine whether adeno-associated virus produces more stable, higher efficiency gene expression in the rodent heart than did previous conventional methods. METHODS Two recombinant viral constructs were made: an adeno-associated virus containing the lacZ gene under the control of the cytomegalovirus promoter (AAV-lacZ) and an adenovirus expressing lacZ under the control of the same promoter (Adeno-lacZ). Twenty rats were injected (into the ventricular apex) with 1 x 10(7-8) genomic particles of each virus. Animals were put to death at serial time points and transgene expression quantitated by beta-galactosidase activity, myocardial staining, and Western blot protein analysis. RESULTS Three months after adeno-associated virus gene transfer, animals demonstrated stable beta-galactosidase expression in 60% of cardiomyocytes without evidence of myocardial inflammation/necrosis. The distribution and degree of protein expression and number of positive cells at 3 months were equivalent to transgene expression at 4 weeks. Adeno-associated virus was not detected in organs other than the heart. In contrast, Adeno-lacZ animals displayed transient beta-galactosidase activity in 60% of cardiomyocytes, which was undetectable 4 weeks after gene transfer. Adenovirus-treated animals manifest significant myocardial inflammation and had transgene expression in other organs. CONCLUSION Direct intramyocardial injection of an adeno-associated virus vector programs stable, long-term, cardiac-specific transgene expression in the rodent heart for up to 3 months. Our results suggest adeno-associated virus has significant advantages for long-term transgene expression in the heart compared to adenovirus vectors.
Collapse
Affiliation(s)
- Danny Chu
- Division of Cardiothoracic Surgery, University of California at San Diego, 200 West Arbor Drive, San Diego, CA 92103-8892, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Klueh U, Dorsky DI, Kreutzer DL. Use of vascular endothelial cell growth factor gene transfer to enhance implantable sensor function
in vivo. J Biomed Mater Res A 2003; 67:1072-86. [PMID: 14666925 DOI: 10.1002/jbm.a.20041] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
In the current study, we developed and validated a simple, rapid and safe in vivo model to test gene transfer and sensor function in vivo. Using the model, we tested the specific hypothesis that in vivo gene transfer of angiogenic factors at sites of biosensor implantation would induce neovascularization surrounding the sensor and thereby enhance biosensor function in vivo. As the in vivo site for testing of our gene transfer cell and biosensor function systems, the developing chorioallantoic membrane (CAM) of the embryo was utilized. Vascular endothelial cell growth factor (VEGF) was used as a prototype for angiogenic factor gene transfer. A helper-independent retroviral vector derived from Rous sarcoma virus (RSV), designated RCAS, was used for gene transfer of the murine VEGF (mVEGF) gene (mVEGF:RCAS) into the DF-1 chicken cell line (designated mVEGF:DF-1). Initially, the ability of VEGF:DF-1 cells to produce VEGF and RCAS viral vectors containing the mVEGF gene (mVEGF:RCAS) was validated in vitro and in vivo, as was the ability of the mVEGF:DF-1 cells to induce neovascularization in the ex ova CAM model. Using the system, we determined the ability of mVEGF:DF-1 cells to enhance acetaminophen sensor function in vivo, by inducing neovascularization at sites of sensor implantation in the ex ova CAM model. For these studies, acetaminophen sensors were placed on 8-day-old ex ova CAMs, followed by addition of media or cells (mVEGF:DF-1 cells or GFP:DF-1 cells) at the sites of biosensor implantation on the CAM. At 4 to 10 days after sensor placement, the biosensor function was determined by measuring sensor response to an intravenous injection of acetaminophen. Sensors implanted on CAMs with buffer or control cells (GFP:DF-1 cells) displayed no induced neovascularization around the sensor and had minimal/baseline sensor responses to intravenous acetaminophen injection (media, 133.33 +/- 27.64 nA; GFP:DF-1, 187.50 +/- 55.43 nA). Alternatively, the sensors implanted with mVEGF:DF-1 cells displayed massive neovascularization and equally massive sensor response to intravenous injection of acetaminophen (VEGF:DF-1, 1387.50 +/- 276.42 nA). These data clearly demonstrate that enhancing vessel density (i.e., neovascularization) around an implanted sensor dramatically enhances sensor function in vivo.
Collapse
Affiliation(s)
- U Klueh
- Center for Molecular Tissue Engineering, University of Connecticut, School of Medicine, Farmington 06030, USA
| | | | | |
Collapse
|
23
|
Crottogini A, Meckert PC, Vera Janavel G, Lascano E, Negroni J, Del Valle H, Dulbecco E, Werba P, Cuniberti L, Martínez V, De Lorenzi A, Telayna J, Mele A, Fernández JL, Marangunich L, Criscuolo M, Capogrossi MC, Laguens R. Arteriogenesis Induced by Intramyocardial Vascular Endothelial Growth Factor 165 Gene Transfer in Chronically Ischemic Pigs. Hum Gene Ther 2003; 14:1307-18. [PMID: 14503966 DOI: 10.1089/104303403322319390] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Exogenous vascular endothelial growth factor (VEGF) improves tissue perfusion in large animals and humans with chronic myocardial ischemia. Because tissue perfusion is mainly dependent on the arteriolar tree, we hypothesized that the neovascularizing effect of VEGF should include arteriogenesis, an effect not as yet described in large mammalian models of myocardial ischemia. In the present study we investigated the effect of intramyocardial plasmid-mediated human VEGF(165) gene transfer (pVEGF(165)) on the proliferation of vessels with smooth muscle in a pig model of myocardial ischemia. In addition, we assessed the effect of treatment on capillary growth, myocardial perfusion, myocardial function and collateralization. Three weeks after positioning of an Ameroid constrictor (Research Instruments SW, Escondido, CA) in the left circumflex artery, pigs underwent basal perfusion (single-photon emission computed tomography [SPECT] with (99m)Tc-sestamibi) and regional function (echocardiography) studies at rest and under dobutamine stress, and were then randomly assigned to receive transepicardial injection of pVEGF(165) 3.8 mg (n = 8) or placebo (empty plasmid, n = 8). All experimental steps and data analysis were done in a blinded fashion. Five weeks later, pVEGF(165)-treated pigs showed a significantly higher density of small (8-50 microm in diameter) vessels with smooth muscle, higher density of capillaries, and improved myocardial perfusion. These results indicate an arteriogenic effect of VEGF in a large mammalian model of myocardial ischemia and encourage the use of VEGF to promote arteriolar growth in patients with severe coronary artery disease.
Collapse
Affiliation(s)
- Alberto Crottogini
- Department of Physiology, Favaloro University, 1078 Buenos Aires, Argentina.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Emani SM, Shah AS, Bowman MK, Emani S, Wilson K, Glower DD, Koch WJ. Catheter-based intracoronary myocardial adenoviral gene delivery: importance of intraluminal seal and infusion flow rate. Mol Ther 2003; 8:306-13. [PMID: 12907153 DOI: 10.1016/s1525-0016(03)00149-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
Although percutaneous, adenoviral-mediated intracoronary gene delivery to the heart has been demonstrated in some species, consistent and safe methodology is needed before clinical applicability is possible. In this study, we examine the effects of altering intracoronary flow rate and obtaining an adequate seal between the catheter and the coronary lumen on successful cardiac gene delivery and myocardial injury in both piglets and adult rabbits. To study the efficacy of in vivo myocardial gene transfer, we utilized adenoviral vectors containing either the beta(2)-adrenergic receptor or beta-galactosidase. The left circumflex coronary artery of piglets and the right coronary artery of rabbits were catheterized under fluoroscopic guidance and adenovirus solutions were injected using varying flow rates with or without balloon inflation. Successful transgene delivery to the heart was determined approximately 1 week after coronary infusions. Histologic analysis was also performed in all animals to determine the extent of myocardial injury. Our results indicate that efficient and reproducible cardiac transgene expression utilizing intracoronary delivery is dependent upon the infusion flow rate and, in larger animals, requires an intraluminal seal. Excessive flow rate is associated with greater myocardial injury. Thus, conditions can be established and controlled to improve future investigational and clinical application of catheter-based intracoronary myocardial gene therapy.
Collapse
|
25
|
Therapeutic angiogenesis induced by human hepatocyte growth factor (HGF) gene in rat myocardial ischemia models. CHINESE SCIENCE BULLETIN-CHINESE 2003. [DOI: 10.1007/bf03183297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
26
|
Francis SC, Katovich MJ, Gelband CH, Raizada MK. Gene therapy in cardiovascular disease. Current status. AMERICAN JOURNAL OF PHARMACOGENOMICS : GENOMICS-RELATED RESEARCH IN DRUG DEVELOPMENT AND CLINICAL PRACTICE 2002; 1:55-66. [PMID: 12173315 DOI: 10.2165/00129785-200101010-00007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Cardiovascular disease is the leading cause of mortality and morbidity in developed countries. Most conventional therapy is often inefficacious and tends to treat the symptoms rather than the underlying causes of the disorder. Gene therapy offers a novel approach for prevention and treatment of cardiovascular diseases. Technical advances in viral vector systems and the development of fusigenic liposome vectors have been crucial to the development of effective gene therapy strategies directed at the vasculature and myocardium in animal models. Gene transfer techniques are being evaluated as potential treatment alternatives for both genetic (familial hypercholesterolemia) and acquired occlusive vascular diseases (atherosclerosis, restenosis, arterial thrombosis) as well as for cardiac disorders including heart failure, myocardial ischemia, graft coronary arteriosclerosis and hypertension. Continued technologic advances in vector systems and promising results in human and animal gene transfer studies make the use of gene therapy a promising strategy for the treatment of cardiovascular disorders.
Collapse
Affiliation(s)
- S C Francis
- Department of Physiology, College of Medicine, University of Florida Brain Institute, Gainesville, Florida, USA
| | | | | | | |
Collapse
|
27
|
Gurunluoglu R, Ozer K, Skugor B, Lubiatowski P, Carnevale K, Siemionow M. Effect of transfection time on the survival of epigastric skin flaps pretreated with adenovirus encoding the VEGF gene. Ann Plast Surg 2002; 49:161-9. [PMID: 12187344 DOI: 10.1097/00000637-200208000-00009] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
An experimental study was conducted to investigate the effect of time of adenovirus-mediated vascular endothelial growth factor (VEGF) gene therapy on the viability of epigastric skin flaps. Eighty-four male Sprague-Dawley rats were used. Skin flaps measuring 8 x 8 cm were marked on the ventral abdominal wall. The upper border of the flap was 1 cm above the costal margin, and the lower border was at the pubis and the inguinal fold. The lateral borders of the flap corresponded to the location of the distinct conversion of the thin ventral skin to the thick dorsal skin. Seven sites in the predicted area of necrosis on the outlined skin flaps were chosen for subdermal injections. All injections were administered by an individual who was blinded to the different treatment groups. The rats received either saline (control group I, N = 28) or adenovirus encoding green fluorescent protein (Ad-GFP; group II, N = 28) or Ad-VEGF (group III, N = 28). The epigastric island skin flaps based solely on the right inferior epigastric vessels were elevated either on the same day of injection (day 0 = 12 hours after transfection, N = 7) or on day 3 (N = 7), day 7 (N = 7), or day 14 (N = 7) after subdermal gene therapy. Flaps were sutured back to their native configuration. Flap viability was evaluated on day 7 after surgery. Sections of the flaps were examined histologically after undergoing hematoxylin-eosin staining. There was a significant reduction in mean percentage of necrotic flap area by 56%, 67%, 70%, and 54% in flaps transfected with Ad-VEGF, 12 hours, 3 days, 7 days, and 14 days before flap elevation, respectively ( < 0.05). There was no evidence that the mean percentage of skin necrosis in the Ad-GFP group was different than in the control group ( = 0.26). There was evidence of mild inflammation in flaps pretreated with Ad-GFP and Ad-VEGF compared with the control group. The authors demonstrated that adenovirus-mediated gene therapy of the abdominal skin after subdermal injections was technically feasible. This was demonstrated by the visualization of GFP expression in control experiments using a fluorescence microscope. In this study, adenovirus-mediated VEGF gene therapy promoted epigastric flap survival, which was not related to the time of transfection. These findings raise the possibility that pretreatment with VEGF gene therapy using an adenovirus vector may be applicable in patients at risk for plastic surgery.
Collapse
Affiliation(s)
- Raffi Gurunluoglu
- Department of Plastic and Reconstructive Surgery, The Cleveland Clinic Foundation, OH 44195, USA
| | | | | | | | | | | |
Collapse
|
28
|
Lubiatowski P, Gurunluoglu R, Goldman CK, Skugor B, Carnevale K, Siemionow M. Gene therapy by adenovirus-mediated vascular endothelial growth factor and angiopoietin-1 promotes perfusion of muscle flaps. Plast Reconstr Surg 2002; 110:149-59. [PMID: 12087246 DOI: 10.1097/00006534-200207000-00026] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
An experimental study was conducted to investigate the potential use of intravascular gene therapy with adenovirus-mediated (Ad) vascular endothelial growth factor (VEGF) or angiopoietin-1 (Ang-1) for the enhancement of muscle flap perfusion and to evaluate the effect of therapy on microcirculatory hemodynamics and microvascular permeability in vivo by using a cremaster muscle flap model in the rat. The cremaster tube flap was left intact after isolation of the pudo-epigastric pedicle. A total of 90 male Sprague-Dawley rats were divided into five groups of 18 each, according to the type of intraarterial treatment. Control flaps received phosphate-buffered saline. Group 2 (the control gene encoding green fluorescent protein, Ad-GFP) served as the adenovirus control. In Groups 3, 4, and 5, flaps were pretreated with Ad-VEGF, Ad-Ang-1, and Ad-Ang-1 + Ad-VEGF, respectively. Flaps were preserved in a subcutaneous pocket in the hindlimb for evaluation of functional capillary density and microvascular permeability indices at 3, 7, and 14 days by intravital microscopy system. At day 7 and 14, Ad-VEGF, Ad-Ang-1, and combined treatment groups showed significantly higher numbers of capillary densities when compared with control and Ad-GFP groups (p < 0.05). At day 14, Ad-VEGF was the superior treatment group compared with Ad-Ang-1 and Ad-VEGF + Ad-Ang-1 (p < 0.05). Overall, there was a linear increase in the number of functional capillaries in all treatment groups (p < 0.05). At day 3 after Ad-Ang-1 therapy, a significantly lower permeability index was found when compared with Ad-VEGF + Ad-Ang-1 and Ad-VEGF alone treatment (p < 0.05). At day 7, the Ad-VEGF group had the highest score of permeability index compared with control, combined, and Ad-Ang-1 groups (p < 0.05). Histologic evaluation of muscle flaps demonstrated mild focal inflammation. There was evidence of mild vasculitis in all flaps except control muscles. Intravascular angiogenic therapy with Ad-VEGF or Ad-Ang-1 was technically feasible, as demonstrated by expression of the control gene, GFP, along the vascular tree. All treatment groups increased perfusion of the muscle flap over a period of 14 days, indicating a long-lasting effect of gene therapy. Ang-1 alone or in combination with VEGF was as effective as VEGF alone in augmenting muscle perfusion with more stable vessels 1 week after gene therapy.
Collapse
Affiliation(s)
- Przemyslaw Lubiatowski
- Department of Plastic and Reconstructive Surgery, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | | | | | | | | | | |
Collapse
|
29
|
Rosengart TK, Sanborn TA, Hackett N, Crystal RG. Reply to the letter. Catheter Cardiovasc Interv 2002. [DOI: 10.1002/ccd.10172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
30
|
Lubiatowski P, Goldman CK, Gurunluoglu R, Carnevale K, Siemionow M. Enhancement of epigastric skin flap survival by adenovirus-mediated VEGF gene therapy. Plast Reconstr Surg 2002; 109:1986-93. [PMID: 11994603 DOI: 10.1097/00006534-200205000-00031] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
A novel approach to treat ischemic tissues by using gene therapy has recently been introduced on the basis of the angiogenic potential of certain growth factors. The authors investigated the effect of adenovirus-mediated gene therapy with vascular endothelial growth factor (VEGF) delivered into the subdermal space to treat compromised skin flaps. For this purpose, the epigastric skin flap model in rats, based solely on the right inferior epigastric vessels, was used. Thirty male Sprague-Dawley rats were divided into five groups of six rats each. Viral transfection with 108 plaque-forming units was performed 2 days before the epigastric flap elevation. Rats received subdermal injections of adenovirus encoding VEGF (Ad-VEGF) or green fluorescent protein (Ad-GFP) as treatment control. Another set of animals (n = 6) received no injections and were designated as control. To determine whether site of injection had an impact on flap viability, injections were given into the predicted local ischemic area (Ad-VEGF local, n = 6; Ad-GFP local, n = 6) and into the midline of the flap (Ad-VEGF midline, n = 6; Ad-GFP midline, n = 6). A flap measuring 8 x 8 cm was outlined on the abdominal skin extending from the xiphoid process proximally and the pubic region distally, to the anterior axillary lines bilaterally. Then, the epigastric flap was elevated as an island on the right inferior epigastric vessels and sutured back to its bed. Flap viability was evaluated at 7 and 14 days after the first operation. The epigastric flaps were scanned to the computer and areas of hypoxic and/or necrotic zones relative to total flap surface area were measured and expressed as percentages by using Image Pro Plus software. Specimens were taken for histologic evaluation at day 14 before the animals were killed. Combined area of necrotic and hypoxic zones as well as necrotic zone were decreased to 9.7 +/- 1.4 percent and 1.4 +/- 0.9 percent in Ad-VEGF local, and 11.8 +/- 1.9 percent and 3.5 +/- 1.64 percent in Ad-VEGF midline compared with the control and Ad-GFP treatment groups (control, 23 +/- 3.6 percent and 20.1 +/- 3.3 percent; Ad-GFP local, 24.8 +/- 4.8 percent and 16.2 +/- 5.9 percent; and Ad-GFP midline, 23.4 +/- 6.9 percent and 19.5 +/- 7.7 percent; p < 0.05). Histologic evaluation by light microscopy failed to demonstrate any quantitative difference in vascularity of skin flaps between the treatment groups. In this study, the authors demonstrated that adenovirus-mediated gene therapy using VEGF enhanced epigastric skin flap survival, as confirmed by the significant reduction in combined area of necrotic and hypoxic zones of the flap. Compared with the control, both local and midline subdermal injections of Ad-VEGF showed improvement in overall flap survival by 57.9 and 48.7 percent, respectively. The results of this study raise the possibility of using adenovirus-mediated therapeutic angiogenesis for safer flap surgery in high-risk patients.
Collapse
Affiliation(s)
- Przemyslaw Lubiatowski
- Department of Plastic and Reconstructive Surgery, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | | | | | | | | |
Collapse
|
31
|
Dulak J, Partyka L, Jozkowicz A, Heba G, Prager M, Neumayer C, Sobhian B, Thurnher M, Nanobashvili J, Fugl A, Ratajska A, Polterauer P, Pachinger O, Weidinger F, Dembinska-Kiec A, Redl H, Huk I. Gene Transfer of Naked VEGF Plasmid Induces the Formation of Microvessels but not Mature Collaterals in Ischaemic Limb Muscles. Eur Surg 2002. [DOI: 10.1046/j.1563-2563.2002.02034.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
32
|
Gurunluoglu R, Lubiatowski P, Goldman CK, Carnevale K, Siemionow M. Enhancement of muscle flap hemodynamics by angiopoietin-1. Ann Plast Surg 2002; 48:401-9. [PMID: 12068223 DOI: 10.1097/00000637-200204000-00011] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Angiopoietin-1 (Ang-1) constitutes a novel family of endothelial cell-specific angiogenic factors. Ang-1 functions mainly in remodeling, maturation, and stabilization of blood vessels. Its direct role in the process of angiogenesis remains unknown. The authors designed an experimental study to investigate the angiogenic potential of Ang-1 and to determine its hemodynamic effects on the cremaster muscle flap model in the rat. Adenovirus-mediated gene therapy was used for delivery of Ang-1. The study sample included 45 male Sprague-Dawley rats weighing 200 to 250 g. After the cremaster muscle tube flaps were prepared, rats were randomized into three different groups of 15 animals. In group I (the control), the flaps received phosphate-buffered saline (PBS). In group II, flaps were treated with adenovirus vector encoding Ang-1 (Ad-Ang-1). In group III, flaps received a control gene encoding green fluorescein protein (Ad-GFP). All treatments were administered via intra-arterial injections of either viral particles (10(8) placque-forming units) or PBS. The external iliac artery was used for this purpose. The cremaster tube flap was then preserved in a subcutaneous pocket in the lower limb. The tube flap was withdrawn from the limb on days 3, 7, and 14 after intra-arterial injection to evaluate microcirculatory measurements such as red blood cell velocity, vessel diameter, capillary density, and microvascular permeability by intravital microscopy. Evaluations were performed by an investigator who was blinded to treatment groups. In a series of control experiments performed with Ad-GFP, adenoviral gene expression was evidenced by the observation of shiny GFP deposits along the vessel walls under fluorescence microscopy throughout the whole cremaster flap 2 days after transfection. At day 3 there was no evidence of any differences in capillary density and permeability index (PI). At day 7, the functional capillary density was significantly higher in the Ad-Ang-1-treated group compared with the control and the Ad-GFP groups (10/hpf +/- 2 vs. 7/hpf +/- 0.5, p = 0.006; 5/hpf +/- 1.6, p = 0.0001). The PI in the Ad-Ang-1-treated group was significantly lower compared with the Ad-GFP-treated group (1.1/hpf +/- 0.1% vs. 1.4/hpf +/- 0.1%, p = 0.0005). At 14 days, the number of the flowing capillaries was significantly higher in the Ad-Ang-1-treated group compared with the control and the Ad-GFP-treated groups (13/hpf +/- 1.7 vs. 9/hpf +/- 2 and 6/hpf +/- 1.3, p = 0.0001). The microvascular PI was significantly lower in the Ad-Ang-1-treated group compared with the Ad-GFP-treated group (1.3/hpf +/- 0.2% vs. 1.8/hpf +/- 0.5%, p = 0.004). Histologically, the cremaster flaps revealed focal and mild inflammation regardless of the treatment and time point of evaluation. There was evidence of vasculitis in muscles pretreated with Ad-GFP and Ad-Ang-1. In summary, in the Ad-Ang-1-treated cremaster flaps, functional capillary density increased from 46% at day 7 to 98% at day 14 when compared with the control group (p < 0.0001). In conclusion, in this experimental muscle flap model, Ad-Ang-1 treatment proved to be a successful method of angiogenic therapy, providing a long-lasting angiogenic effect over a period of 14 days. The increased capillary perfusion accompanied by the formation of more stable and mature vessels resistant to fluorescein isothiocyanate-conjugated albumin leakage may serve as in vivo evidence that Ang-1 therapy improves skeletal muscle flap hemodynamics. These exciting findings raise the possibility that Ang-1 may have implications for therapeutic angiogenesis. To the authors' knowledge, their study demonstrates for the first time the feasibility of intravascular gene therapy using a virus vector in an attempt to enhance muscle flap hemodynamics.
Collapse
Affiliation(s)
- Raffi Gurunluoglu
- Department of Plastic and Reconstructive Surgery, The Cleveland Clinic Foundation, OH 44195, USA
| | | | | | | | | |
Collapse
|
33
|
Crystal RG, Harvey BG, Wisnivesky JP, O'Donoghue KA, Chu KW, Maroni J, Muscat JC, Pippo AL, Wright CE, Kaner RJ, Leopold PL, Kessler PD, Rasmussen HS, Rosengart TK, Hollmann C. Analysis of risk factors for local delivery of low- and intermediate-dose adenovirus gene transfer vectors to individuals with a spectrum of comorbid conditions. Hum Gene Ther 2002; 13:65-100. [PMID: 11779413 DOI: 10.1089/10430340152712647] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
In this study we analyze the adverse events and abnormal laboratory parameters following local administration of low (<10(9) particle units) and intermediate (10(9)-10(11) particle units) single and repetitive doses (140 total) of E1(-)E3(-) adenovirus (Ad) gene transfer vectors administered to the respiratory epithelium, solid tumors, skin, myocardium, and skeletal muscle in eight gene transfer trials since April 1993. In the accompanying paper by Harvey et al., (Hum. Gene Ther. 2002; 13:15-63), we conclude that for the total group, no deaths were attributable to the Ad vectors per se, and the incidence of major adverse events likely caused by an Ad vector was 0.7%. The present study analyzes the trials as a group to evaluate risk factors for the adverse events, abnormal values among laboratory parameters, and known deaths. Ten putative risk factors were assessed, including "patient-related" (age, sex, comorbid index and pretherapy anti-Ad antibodies), "vector-related" (dose, route, transgene, and number of vector administrations), and "trial-related" (trial in which the individual was enrolled, and whether surgery was part of the trial). While assessment of each factor individually suggested several possible associations with adverse events, abnormal laboratory parameters, or deaths, multivariate analysis identified only age, comorbid index, and surgery (comorbid index for death; age and surgery for non-death adverse events) as variables significantly associated with increased risk for a major (severity scale 3-4 of 4) adverse event for individuals enrolled in these gene transfer trials. Importantly, multivariate analysis suggested that vector-related parameters, including dose, route, transgene, or number of vector administrations at the doses and routes evaluated in these studies, do not appear to be significant risk factors for a major adverse event. With the caveat that these are phase I, uncontrolled trials, we conclude that (1) there is no definitive risk factor that will clearly predict a major adverse outcome resulting from local administration of low and intermediate doses of Ad gene transfer vectors; and (2) major adverse events in these gene transfer trials are associated primarily with the study population and/or trial procedures, not the Ad vectors themselves. This assessment is consistent with the concept that local administration of low and intermediate doses of Ad gene transfer vectors appears to be well tolerated.
Collapse
Affiliation(s)
- Ronald G Crystal
- Institute of Genetic Medicine and Division of Pulmonary and Critical Care Medicine, Weill Medical College of Cornell University, New York, NY 10021, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Gwon HC, Jeong JO, Kim HJ, Park SW, Lee SH, Park SJ, Huh JE, Lee Y, Kim S, Kim DK. The feasibility and safety of fluoroscopy-guided percutaneous intramyocardial gene injection in porcine heart. Int J Cardiol 2001; 79:77-88. [PMID: 11399344 DOI: 10.1016/s0167-5273(01)00410-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Catheter-based transendocardial gene injection would be useful for the delivery of genes into the heart. We examined the feasibility and safety of percutaneous intramyocardial gene injections with fluoroscopic guidance alone. METHODS We performed the procedure through an 8F arterial sheath inserted into the left carotid artery. In protocol 1, a mixture of India ink and normal saline was injected through a needle injection catheter in six pigs. We monitored blood pressure and ECG continuously during the procedure. Echocardiography, left ventriculography, and coronary angiography were performed. All pigs were sacrificed 2 days later and hearts were harvested. In protocol 2, a mixture of India ink and plasmid encoding CAT gene was injected in the same manner in eight pigs. Myocardial tissue was obtained 7 days after the procedure to assess gene expression. In protocol 3, four pigs were intentionally needle-perforated in the ventricular wall and were observed for 7 days. RESULTS In protocol 1, there was no significant hemodynamic changes or serious arrhythmias during the procedure. Echocardiography and angiography revealed no evidence indicating pericardial effusion or wall motion abnormalities. Harvested hearts revealed one intramyocardial hematoma in a total of 36 injection sites. In protocol 2, the gene expression could be identified in 39 sites out of 48 injections after 7 days. In protocol 3, no animal showed signs indicating cardiac tamponade during the observation period. CONCLUSIONS Our data suggest that fluoroscopy-guided percutaneous intramyocardial gene injection is a feasible and safe procedure, with no indication of associated significant hemodynamic changes, arrhythmias, or mortality.
Collapse
Affiliation(s)
- H C Gwon
- Department of Medicine, Sungkyunkwan University School of Medicine, Samsung Medical Center, Molecular Therapy Research Center, Samsung Biomedical Research Institute, Seoul, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Sanborn TA, Hackett NR, Lee LY, El-Sawy T, Blanco I, Tarazona N, Deutsch E, Crystal R, Rosengart TK. Percutaneous endocardial transfer and expression of genes to the myocardium utilizing fluoroscopic guidance. Catheter Cardiovasc Interv 2001; 52:260-6. [PMID: 11170342 DOI: 10.1002/1522-726x(200102)52:2<260::aid-ccd1061>3.0.co;2-t] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Experimental studies indicate that administration of angiogenic proteins or genes by the epicardial or intracoronary route can stimulate development of new collateral vessels and improve myocardial perfusion. An endocardial catheter-based approach to this therapy would obviate the need for surgery, while preserving the effectiveness of direct intramyocardial administration. Fluoroscopic guidance and prototype, preformed, coaxial catheters were used to examine the feasibility of percutaneous catheter-based adenovirus (Ad)-mediated gene transfer and expression in normal swine myocardium. The feasibility of intramyocardial administration (100 microl/injection) of a radiocontrast agent and black tissue dye to all regions of the left ventricle (septum, anterior, lateral, and inferior wall) was confirmed fluoroscopically and on postmortem examination. Injections of replication-deficient adenovirus (10 injections of 10(11) particle units/100 microl each) coding for beta-galactosidase (Adbetagal) or vascular endothelial growth factor (Ad(GV)VEGF121.10) were administered to the left ventricular free wall to examine endocardial based gene transfer and expression. beta-Galactosidase activity was detected by histochemical staining and quantitative assay in targeted regions of the myocardium. Regional VEGF expression was found to be significantly greater in targeted regions (1.3 +/- 0.4 ng/mg protein) as compared with non-targeted regions (0.3 +/- 0.1 ng/mg protein) or regions injected with control (Adbetagal) virus (0.2 +/- 0.03 ng/mg protein, P < 0.001). Catheter-based Ad mediated endocardial gene transfer and expression is feasible using percutaneous, fluoroscopically guided, preformed, coaxial catheters. This approach should be clinically useful to administer angiogenic genes to the ischemic myocardium.
Collapse
Affiliation(s)
- T A Sanborn
- Division of Cardiology, Weill Medical College of Cornell University, New York Presbyterian Hospital, New York, New York, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Affiliation(s)
- W C Russell
- Biomolecular Sciences Building, School of Biology, University of St Andrews, North Haugh, St Andrews, Fife KY16 9ST, UK1
| |
Collapse
|
37
|
Ramalingam R, Worgall S, Rafii S, Crystal RG. Downregulation of CXCR4 gene expression in primary human endothelial cells following infection with E1(-)E4(+) adenovirus gene transfer vectors. Mol Ther 2000; 2:381-6. [PMID: 11020354 DOI: 10.1006/mthe.2000.0131] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Infection of human endothelial cells with first-generation E1(-)E4(+) adenovirus (Ad) vectors leads to prolonged cell survival and changes in the cell phenotype to a more quiescent stage. Based on the concept that the CXCR4, the receptor for the endothelial chemoattractant stromal-derived factor-&alpha (SDF-alpha), is constitutively expressed by quiescent, resting endothelial cells, the present study analyzes the effect of Ad vector infection on CXCR4 expression and SDF-alpha responses of human umbilical vein endothelial cells (HUVEC). CXCR4 transcripts were markedly downregulated in E1(-)E4(+) Ad-infected cells 48 h following infection, but not in uninfected control cells or when the cells were infected with an E1(-)E4(-) Ad vector. Analysis of surface CXCR4 expression by flow cytometry demonstrated marked reduction of the CXCR4 receptor on cells infected with E1(-)E4(+) Ad compared to uninfected control cells or E1(-)E4(-) Ad-infected cells. Infection of other cell types which express CXCR4, such as dendritic cells and myeloma cells, did not exhibit CXCR4 receptor downregulation following infection with E1(-)E4(+) Ad. Consistent with the observed downregulation of CXCR4 mRNA and surface protein, infection of the endothelial cells with an E1(-)E4(+) Ad rendered the cells unresponsive to the chemoattractant SDF-alpha compared to naive or E1(-)E4(-) Ad-infected cells. Together, the data suggest that first-generation Ad vectors, likely the E4 region, modify the ability of endothelial cells to respond to at least one important chemoattractant.
Collapse
Affiliation(s)
- R Ramalingam
- Division of Pulmonary and Critical Care Medicine, Weill Medical College of Cornell University-New York Presbyterian Hospital, New York, New York 94143, USA
| | | | | | | |
Collapse
|
38
|
Lee RJ, Springer ML, Blanco-Bose WE, Shaw R, Ursell PC, Blau HM. VEGF gene delivery to myocardium: deleterious effects of unregulated expression. Circulation 2000; 102:898-901. [PMID: 10952959 DOI: 10.1161/01.cir.102.8.898] [Citation(s) in RCA: 513] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND Vascular endothelial growth factor (VEGF) is being investigated for therapeutic angiogenesis in ischemic myocardium. Primarily, transient delivery systems have been tested. The goal of this study was to investigate the effects of continuous expression of VEGF in myocardium by use of myoblast-mediated delivery. METHODS AND RESULTS Primary murine myoblasts (5 x 10(5) cells in 10 microL of PBS with 0.5% BSA) expressing both the murine VEGF gene and the beta-galactosidase (beta-gal) gene from a retroviral promoter were implanted in the ventricular wall of immunodeficient mice (n=11) via a subdiaphragmatic approach. Control immunodeficient mice (n=12) were injected with the same number of myoblasts expressing only the beta-gal gene. Between days 14 and 16, surviving mice were euthanized and the hearts processed for histology. In the experimental group, 11 of 11 mice demonstrated failure to thrive by day 13; 5 deaths occurred between days 8 and 15. There were no complications in the control mice. Histochemistry documented successful implantation of myoblasts (positive beta-gal reaction product) in 6 of 6 surviving experimental mice and 12 of 12 controls. Histology disclosed intramural vascular tumors resembling hemangiomas in the VEGF-myoblast-injected myocardium in 6 of 6 surviving mice. beta-Gal-expressing cells were present at the site of the vascular tumors. Immunohistochemistry localized abundant endothelial nitric oxide synthase and CD31 (platelet and endothelial cell adhesion molecule) within the lesion, consistent with the presence of endothelial cells. CONCLUSIONS In this model, unregulated continuous expression of VEGF is associated with (1) a high rate of failure to thrive/death and (2) formation of endothelial cell-derived intramural vascular tumors in the implantation site. These results underscore the importance of regulating VEGF expression for therapeutic angiogenesis.
Collapse
Affiliation(s)
- R J Lee
- Department of Medicine and the Cardiovascular Research Institute, University of California, San Francisco, USA.
| | | | | | | | | | | |
Collapse
|
39
|
Lee LY, Patel SR, Hackett NR, Mack CA, Polce DR, El-Sawy T, Hachamovitch R, Zanzonico P, Sanborn TA, Parikh M, Isom OW, Crystal RG, Rosengart TK. Focal angiogen therapy using intramyocardial delivery of an adenovirus vector coding for vascular endothelial growth factor 121. Ann Thorac Surg 2000; 69:14-23; discussion 23-4. [PMID: 10654479 DOI: 10.1016/s0003-4975(99)01102-9] [Citation(s) in RCA: 104] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
BACKGROUND Adenovirus (Ad) vector-mediated gene therapy strategies have emerged as promising modalities for the "biological revascularization" of tissues. We hypothesized that direct intramyocardial, as opposed to intracoronary, administration of an Ad vector coding for the vascular endothelial growth factor 121 cDNA (Ad(GV)VEGF121.10) would provide highly focal Ad genome levels, and increases in VEGF, ideal for inducing localized therapeutic angiogenesis. METHODS Persistence and regional distribution of the vector were assessed by TaqMan real-time quantitative polymerase chain reaction technology and enzyme-linked immunosorbent assay, after intramyocardial Ad(GV)VEGF121.10 in the rat, and either intramyocardial or intracoronary (circumflex territory) vector in Yorkshire swine. Based on these results, we assessed the focal nature of the improved cardiac blood flow in a previously reported porcine myocardial ischemia model. RESULTS Intramyocardial delivery of Ad(GV)VEGF121.10 in the rat resulted in local persistence of the Ad genome that decreased 1,000-fold over 3 weeks, with peak myocardial VEGF expression 24 to 72 h after vector delivery. After intramyocardial Ad(GV)VEGF121.10 in the circumflex distribution of pigs, Ad vector genome and VEGF protein levels were more than 1,000-fold and more than 90-fold higher, respectively, in this distribution than in other myocardial regions. In comparison, intracoronary injection yielded maximum myocardial Ad genome and VEGF levels 33-fold and 9-fold lower, respectively, than that after intramyocardial delivery. Angiograms obtained 28 days after intramyocardial Ad(GV)VEGF121.10 demonstrated rapid circumflex reconstitution via collaterals localized to the region of vector administration. CONCLUSIONS These studies demonstrate that direct intramyocardial administration of Ad(GV)VEGF121.10 results in focal genome and VEGF levels, including focal angiogenesis, sufficient to normalize blood flow to the ischemic myocardium, findings that are relevant to designing human trials of gene therapy-mediated cardiac angiogenesis.
Collapse
Affiliation(s)
- L Y Lee
- Department of Cardiothoracic Surgery, The New York Hospital-Cornell Medical Center, New York, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|