1
|
Farraha M, Kumar S, Chong J, Cho HC, Kizana E. Gene Therapy Approaches to Biological Pacemakers. J Cardiovasc Dev Dis 2018; 5:jcdd5040050. [PMID: 30347716 PMCID: PMC6306875 DOI: 10.3390/jcdd5040050] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 10/16/2018] [Accepted: 10/17/2018] [Indexed: 01/01/2023] Open
Abstract
Bradycardia arising from pacemaker dysfunction can be debilitating and life threatening. Electronic pacemakers serve as effective treatment options for pacemaker dysfunction. They however present their own limitations and complications. This has motivated research into discovering more effective and innovative ways to treat pacemaker dysfunction. Gene therapy is being explored for its potential to treat various cardiac conditions including cardiac arrhythmias. Gene transfer vectors with increasing transduction efficiency and biosafety have been developed and trialed for cardiovascular disease treatment. With an improved understanding of the molecular mechanisms driving pacemaker development, several gene therapy targets have been identified to generate the phenotypic changes required to correct pacemaker dysfunction. This review will discuss the gene therapy vectors in use today along with methods for their delivery. Furthermore, it will evaluate several gene therapy strategies attempting to restore biological pacing, having the potential to emerge as viable therapies for pacemaker dysfunction.
Collapse
Affiliation(s)
- Melad Farraha
- Centre for Heart Research, the Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW 2145, Australia.
- Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia.
| | - Saurabh Kumar
- Department of Cardiology, Westmead Hospital, Westmead, NSW 2145, Australia.
| | - James Chong
- Centre for Heart Research, the Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW 2145, Australia.
- Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia.
- Department of Cardiology, Westmead Hospital, Westmead, NSW 2145, Australia.
| | - Hee Cheol Cho
- Departments of Pediatrics and Biomedical Engineering, Emory University, Atlanta, GA 30322, USA.
| | - Eddy Kizana
- Centre for Heart Research, the Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW 2145, Australia.
- Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia.
- Department of Cardiology, Westmead Hospital, Westmead, NSW 2145, Australia.
| |
Collapse
|
2
|
Penny WF, Hammond HK. Randomized Clinical Trials of Gene Transfer for Heart Failure with Reduced Ejection Fraction. Hum Gene Ther 2017; 28:378-384. [PMID: 28322590 PMCID: PMC5444414 DOI: 10.1089/hum.2016.166] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Despite improvements in drug and device therapy for heart failure, hospitalization rates and mortality have changed little in the past decade. Randomized clinical trials using gene transfer to improve function of the failing heart are the focus of this review. Four randomized clinical trials of gene transfer in heart failure with reduced ejection fraction (HFrEF) have been published. Each enrolled patients with stable symptomatic HFrEF and used either intracoronary delivery of a virus vector or endocardial injection of a plasmid. The initial CUPID trial randomized 14 subjects to placebo and 25 subjects to escalating doses of adeno-associated virus type 1 encoding sarcoplasmic reticulum calcium ATPase (AAV1.SERCA2a). AAV1.SERCA2a was well tolerated, and the high-dose group met a 6 month composite endpoint. In the subsequent CUPID-2 study, 243 subjects received either placebo or the high dose of AAV1.SERCA2a. AAV1.SERCA2a administration, while safe, failed to meet the primary or any secondary endpoints. STOP-HF used plasmid endocardial injection of stromal cell-derived factor-1 to promote stem-cell recruitment. In a 93-subject trial of patients with ischemic etiology heart failure, the primary endpoint (symptoms and 6 min walk distance) failed, but subgroup analyses showed improvements in subjects with the lowest ejection fractions. A fourth trial randomized 14 subjects to placebo and 42 subjects to escalating doses of adenovirus-5 encoding adenylyl cyclase 6 (Ad5.hAC6). There were no safety concerns, and patients in the two highest dose groups (combined) showed improvements in left ventricular function (left ventricular ejection fraction and -dP/dt). The safety data from four randomized clinical trials of gene transfer in patients with symptomatic HFrEF suggest that this approach can be conducted with acceptable risk, despite invasive delivery techniques in a high-risk population. Additional trials are necessary before the approach can be endorsed for clinical practice.
Collapse
Affiliation(s)
- William F Penny
- 1 VA San Diego Healthcare System, San Diego, California.,2 Department of Medicine, University of California , San Diego, San Diego, California
| | - H Kirk Hammond
- 1 VA San Diego Healthcare System, San Diego, California.,2 Department of Medicine, University of California , San Diego, San Diego, California
| |
Collapse
|
3
|
Abstract
Atrial fibrillation is a prominent cause of morbidity and mortality in developed countries. Treatment strategies center on controlling atrial rhythm or ventricular rate. The need for anticoagulation is an independent decision from the rate versus rhythm control debate. This review discusses novel biological strategies that have potential utility in the management of atrial fibrillation. Rate controlling strategies predominately rely on G-protein gene transfer to enhance cholinergic or suppress adrenergic signaling pathways in the atrioventricular node. Calcium channel blocking gene therapy and fibrosis enhancing cell therapy have also been reported. Rhythm controlling strategies focus on disrupting reentry by enhancing conduction or suppressing repolarization. Efforts to suppress inflammation and apoptosis are also under study. Resistance to blood clot formation has been shown with thrombomodulin. These strategies are in various stages of preclinical development.
Collapse
|
4
|
Farraha M, Chong JJ, Kizana E. Therapeutic Prospects of Gene Therapy for Atrial Fibrillation. Heart Lung Circ 2016; 25:808-13. [DOI: 10.1016/j.hlc.2016.04.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 04/17/2016] [Indexed: 01/01/2023]
|
5
|
Hammond HK, Penny WF, Traverse JH, Henry TD, Watkins MW, Yancy CW, Sweis RN, Adler ED, Patel AN, Murray DR, Ross RS, Bhargava V, Maisel A, Barnard DD, Lai NC, Dalton ND, Lee ML, Narayan SM, Blanchard DG, Gao MH. Intracoronary Gene Transfer of Adenylyl Cyclase 6 in Patients With Heart Failure: A Randomized Clinical Trial. JAMA Cardiol 2016; 1:163-71. [PMID: 27437887 PMCID: PMC5535743 DOI: 10.1001/jamacardio.2016.0008] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
IMPORTANCE Gene transfer has rarely been tested in randomized clinical trials. OBJECTIVE To evaluate the safety and efficacy of intracoronary delivery of adenovirus 5 encoding adenylyl cyclase 6 (Ad5.hAC6) in heart failure. DESIGN, SETTING, AND PARTICIPANTS A randomized, double-blind, placebo-controlled, phase 2 clinical trial was conducted in US medical centers (randomization occurred from July 19, 2010, to October 30, 2014). Participants 18 to 80 years with symptomatic heart failure (ischemic and nonischemic) and an ejection fraction (EF) of 40% or less were screened; 86 individuals were enrolled, and 56 were randomized. Data analysis was of the intention-to-treat population. Participants underwent exercise testing and measurement of left ventricular EF (echocardiography) and then cardiac catheterization, where left ventricular pressure development (+dP/dt) and decline (-dP/dt) were recorded. Participants were randomized (3:1 ratio) to receive 1 of 5 doses of intracoronary Ad5.hAC6 or placebo. Participants underwent a second catheterization 4 weeks later for measurement of dP/dt. Exercise testing and EF were assessed 4 and 12 weeks after randomization. INTERVENTIONS Intracoronary administration of Ad5.hAC6 (3.2 × 109 to 1012 virus particles) or placebo. MAIN OUTCOMES AND MEASURES Primary end points included exercise duration and EF before and 4 and 12 weeks after randomization and peak rates of +dP/dt and -dP/dt before and 4 weeks after randomization. Fourteen placebo participants were compared (intention to treat) with 24 Ad5.hAC6 participants receiving the highest 2 doses (D4 + 5). RESULTS Fifty-six individuals were randomized and monitored for up to 1 year. Forty-two participants (75%) received Ad5.hAC6 (mean [SE] age, 63 [1] years; EF, 30% [1%]), and 14 individuals (25%) received placebo (age, 62 [1] years; EF, 30% [2%]). Exercise duration showed no significant group differences (4 weeks, P = .27; 12 weeks, P = .47, respectively). The D4 + 5 participants had increased EF at 4 weeks (+6.0 [1.7] EF units; n = 21; P < .004), but not 12 weeks (+3.0 [2.4] EF units; n = 21; P = .16). Placebo participants showed no increase in EF at 4 weeks or 12 weeks. Exercise duration showed no between-group differences (4-week change from baseline: placebo, 27 [36] seconds; D4 + 5, 44 [25] seconds; P = .27; 12-week change from baseline: placebo, 44 [28] seconds; D4 + 5, 58 [29 seconds, P = .47). AC6 gene transfer increased basal left ventricular peak -dP/dt (4-week change from baseline: placebo, +93 [51] mm Hg/s; D4 + 5, -39 [33] mm Hg/s; placebo [n = 21]; P < .03); AC6 did not increase arrhythmias. The admission rate for patients with heart failure was 9.5% (4 of 42) in the AC6 group and 28.6% (4 of 14) in the placebo group (relative risk, 0.33 [95% CI, 0.08-1.36]; P = .10). CONCLUSIONS AND RELEVANCE AC6 gene transfer safely increased LV function beyond standard heart failure therapy, attainable with one-time administration. Larger trials are warranted. TRIAL REGISTRATION clinicaltrials.gov Identifier: NCT00787059.
Collapse
Affiliation(s)
- H Kirk Hammond
- Veterans Affairs San Diego Healthcare System, San Diego California2Department of Medicine, University of California, San Diego
| | - William F Penny
- Veterans Affairs San Diego Healthcare System, San Diego California2Department of Medicine, University of California, San Diego
| | | | - Timothy D Henry
- Minneapolis Heart Institute, Minneapolis, Minnesota4now with Cedars Sinai Heart Institute, Los Angeles, California
| | | | - Clyde W Yancy
- Department of Medicine, Northwestern University, Chicago, Illinois
| | - Ranya N Sweis
- Department of Medicine, Northwestern University, Chicago, Illinois
| | - Eric D Adler
- Department of Medicine, University of California, San Diego
| | - Amit N Patel
- Department of Medicine, University of Utah, Salt Lake City
| | - David R Murray
- Department of Medicine, University of Wisconsin, Madison
| | - Robert S Ross
- Veterans Affairs San Diego Healthcare System, San Diego California2Department of Medicine, University of California, San Diego
| | - Valmik Bhargava
- Veterans Affairs San Diego Healthcare System, San Diego California2Department of Medicine, University of California, San Diego
| | - Alan Maisel
- Veterans Affairs San Diego Healthcare System, San Diego California2Department of Medicine, University of California, San Diego
| | - Denise D Barnard
- Veterans Affairs San Diego Healthcare System, San Diego California2Department of Medicine, University of California, San Diego
| | - N Chin Lai
- Veterans Affairs San Diego Healthcare System, San Diego California2Department of Medicine, University of California, San Diego
| | - Nancy D Dalton
- Department of Medicine, University of California, San Diego
| | - Martin L Lee
- Department of Biostatistics, UCLA (University of California at Los Angeles)
| | - Sanjiv M Narayan
- Veterans Affairs San Diego Healthcare System, San Diego California2Department of Medicine, University of California, San Diego10now with School of Medicine, Stanford University, Palo Alto, California
| | | | - Mei Hua Gao
- Veterans Affairs San Diego Healthcare System, San Diego California2Department of Medicine, University of California, San Diego
| |
Collapse
|
6
|
Affiliation(s)
- J Kevin Donahue
- Division of Cardiovascular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| |
Collapse
|
7
|
Abstract
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.
Collapse
|
8
|
Abstract
Recent advances in our understanding of the pathophysiology of myocardial dysfunction in the setting of congestive heart failure have created a new opportunity in developing nonpharmacological approaches to treatment. Gene therapy has emerged as a powerful tool in targeting the molecular mechanisms of disease by preventing the ventricular remodeling and improving bioenergetics in heart failure. Refinements in vector technology, including the creation of recombinant adeno-associated viruses, have allowed for safe and efficient gene transfer. These advancements have been coupled with evolving delivery methods that include vascular, pericardial, and direct myocardial approaches. One of the most promising targets, SERCA2a, is currently being used in clinical trials. The recent success of the Calcium Upregulation by Percutaneous Administration of Gene Therapy in Cardiac Disease phase 2 trials using adeno-associated virus 1-SERCA2a in improving outcomes highlights the importance of gene therapy as a future tool in treating congestive heart failure.
Collapse
|
9
|
|
10
|
Tang T, Hammond HK. Gene transfer for congestive heart failure: update 2013. Transl Res 2013; 161:313-20. [PMID: 23261978 PMCID: PMC3602385 DOI: 10.1016/j.trsl.2012.11.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Revised: 11/19/2012] [Accepted: 11/27/2012] [Indexed: 01/08/2023]
Abstract
Congestive heart failure is a major cause of morbidity and mortality with increasing social and economic costs. There have been no new high impact therapeutic agents for this devastating disease for more than a decade. However, many pivotal regulators of cardiac function have been identified using cardiac-directed transgene expression and gene deletion in preclinical studies. Some of these increase function of the failing heart. Altering the expression of these pivotal regulators using gene transfer is now either being tested in clinical gene transfer trials, or soon will be. In this review, we summarize recent progress in cardiac gene transfer for clinical congestive heart failure.
Collapse
Affiliation(s)
- Tong Tang
- Department of Medicine, University of California San Diego, and VA San Diego Healthcare System, San Diego, Calif., USA
| | | |
Collapse
|
11
|
Mechanistic, technical, and clinical perspectives in therapeutic stimulation of coronary collateral development by angiogenic growth factors. Mol Ther 2013; 21:725-38. [PMID: 23403495 DOI: 10.1038/mt.2013.13] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
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.
Collapse
|
12
|
Minimally invasive closed-chest ultrasound-guided substance delivery into the pericardial space in mice. Naunyn Schmiedebergs Arch Pharmacol 2012; 386:227-38. [PMID: 23250337 PMCID: PMC3570759 DOI: 10.1007/s00210-012-0815-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Accepted: 11/13/2012] [Indexed: 01/13/2023]
Abstract
Organ-directed gene transfer remains an attractive method for both gaining a better understanding of heart disease and for cardiac therapy. However, virally mediated transfer of gene products into cardiac cells requires prolonged exposure of the myocardium to the viral substrate. Pericardial injection of viral vectors has been proposed and used with some success to achieve myocardial transfection and may be a suitable approach for transfection of atrial myocardium. Indeed, such an organ-specific method would be particularly useful to reverse phenotypes in young and adult genetically altered murine models of cardiac disease. We therefore sought to develop a minimally invasive technique for pericardial injection of substances in mice. Pericardial access in anaesthetised, spontaneously breathing mice was achieved using continuous high-resolution ultrasound guidance. We could demonstrate adequate delivery of injected substances into the murine pericardium. Atrial epicardial and myocardial cells were transfected in approximately one third of mice injected with enhanced green fluorescent protein-expressing adenovirus. Cellular expression rates within individual murine atria were limited to a maximum of 20 %; therefore, expression efficiency needs to be further improved. Minimally invasive, ultrasound-guided injection of viral material appears a technically challenging yet feasible method for selective transfection of atrial epi- and myocardium. This pericardial injection method may be useful in the evaluation of potential genetic interventions aimed at rescuing atrial phenotypes in transgenic mouse models.
Collapse
|
13
|
Fang H, Lai NC, Gao MH, Miyanohara A, Roth DM, Tang T, Hammond HK. Comparison of Adeno-Associated Virus Serotypes and Delivery Methods for Cardiac Gene Transfer. Hum Gene Ther Methods 2012. [DOI: 10.1089/hum.2012.105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
14
|
Fang H, Lai NC, Gao MH, Miyanohara A, Roth DM, Tang T, Hammond HK. Comparison of adeno-associated virus serotypes and delivery methods for cardiac gene transfer. Hum Gene Ther Methods 2012; 23:234-41. [PMID: 22966786 PMCID: PMC3555516 DOI: 10.1089/hgtb.2012.105] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Accepted: 07/08/2012] [Indexed: 01/28/2023] Open
Abstract
Cardiac gene transfer is a potentially useful strategy for cardiovascular diseases. The adeno-associated virus (AAV) is a common vector to obtain transgene expression in the heart. Initial studies conducted in rodents used indirect intracoronary delivery for cardiac gene transfer. More recently AAV vectors with so-called cardiac tropism have enabled significant cardiac transgene expression following intravenous injection. However, a direct comparison of intravenous versus intracoronary delivery with rigorous quantification of cardiac transgene expression has not been conducted. In the present study we tested the hypothesis that intracoronary AAV delivery would be superior to intravenous delivery vis-à-vis cardiac transgene expression. We compared intravenous and intracoronary delivery of AAV5, AAV6, and AAV9 (5×10(11) genome copies per mouse). Using enhanced green fluorescent protein as a reporter, we quantified transgene expression by fluorescence intensity and Western blotting. Quantitative polymerase chain reaction (PCR) was also performed to assess vector DNA copies, employing primers against common sequences on AAV5, AAV6, and AAV9. Intracoronary delivery resulted in 2.6- to 28-fold higher transgene protein expression in the heart 3 weeks after AAV injection compared to intravenous delivery depending on AAV serotype. The highest level of cardiac gene expression was achieved following intracoronary delivery of AAV9. Intracoronary delivery of AAV9 is a preferred method for cardiac gene transfer.
Collapse
Affiliation(s)
- Hongfei Fang
- Department of Medicine, University of California San Diego, San Diego, California
- VA San Diego Healthcare System, San Diego, California
| | - Ngai Chin Lai
- Department of Medicine, University of California San Diego, San Diego, California
- VA San Diego Healthcare System, San Diego, California
| | - Mei Hua Gao
- Department of Medicine, University of California San Diego, San Diego, California
- VA San Diego Healthcare System, San Diego, California
| | - Atsushi Miyanohara
- Department of Medicine, University of California San Diego, San Diego, California
| | - David M. Roth
- VA San Diego Healthcare System, San Diego, California
- Department of Anesthesiology, University of California San Diego, San Diego, California
| | - Tong Tang
- Department of Medicine, University of California San Diego, San Diego, California
- VA San Diego Healthcare System, San Diego, California
| | - H. Kirk Hammond
- Department of Medicine, University of California San Diego, San Diego, California
- VA San Diego Healthcare System, San Diego, California
| |
Collapse
|
15
|
Shi W, Schmarkey LS, Jiang R, Bone CC, Condit ME, Dillehay DL, Engler RL, Rubanyi GM, Vinten-Johansen J. Ischemia-reperfusion increases transfection efficiency of intracoronary adenovirus type 5 in pig heart in situ. Hum Gene Ther Methods 2012; 23:204-12. [PMID: 22816318 DOI: 10.1089/hgtb.2012.048] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
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.
Collapse
Affiliation(s)
- Weiwei Shi
- Cardiothoracic Research Laboratory, Carlyle Fraser Heart Center, Emory University, Atlanta, GA 30308, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Karakikes I, Hadri L, Rapti K, Ladage D, Ishikawa K, Tilemann L, Yi GH, Morel C, Gwathmey JK, Zsebo K, Weber T, Kawase Y, Hajjar RJ. Concomitant intravenous nitroglycerin with intracoronary delivery of AAV1.SERCA2a enhances gene transfer in porcine hearts. Mol Ther 2012; 20:565-71. [PMID: 22215018 DOI: 10.1038/mt.2011.268] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
SERCA2a gene therapy improves contractile and energetic function of failing hearts and has been shown to be associated with benefits in clinical outcomes, symptoms, functional status, biomarkers, and cardiac structure in a phase 2 clinical trial. In an effort to enhance the efficiency and homogeneity of gene uptake in cardiac tissue, we examined the effects of nitroglycerin (NTG) in a porcine model following AAV1.SERCA2a gene delivery. Three groups of Göttingen minipigs were assessed: (i) group A: control intracoronary (IC) AAV1.SERCA2a (n = 6); (ii) group B: a single bolus IC injection of NTG (50 µg) immediately before administration of intravenous (IV) AAV1.SERCA2a (n = 6); and (iii) group C: continuous IV NTG (1 µg/kg/minute) during the 10 minutes of AAV1.SERCA2a infusion (n = 6). We found that simultaneous IV infusion of NTG and AAV1.SERCA2a resulted in increased viral transduction efficiency, both in terms of messenger RNA (mRNA) as well as SERCA2a protein levels in the whole left ventricle (LV) compared to control animals. On the other hand, IC NTG pretreatment did not result in enhanced gene transfer efficiency, mRNA or protein levels when compared to control animals. Importantly, the transgene expression was restricted to the heart tissue. In conclusion, we have demonstrated that IV infusion of NTG significantly improves cardiac gene transfer efficiency in porcine hearts.
Collapse
Affiliation(s)
- Ioannis Karakikes
- The Cardiovascular Research Center, Mount Sinai School of Medicine, New York, New York 10029, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Rapti K, Chaanine AH, Hajjar RJ. Targeted gene therapy for the treatment of heart failure. Can J Cardiol 2011; 27:265-83. [PMID: 21601767 PMCID: PMC5902317 DOI: 10.1016/j.cjca.2011.02.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2010] [Revised: 02/10/2011] [Accepted: 02/11/2011] [Indexed: 12/18/2022] Open
Abstract
Chronic heart failure is one of the leading causes of morbidity and mortality in Western countries and is a major financial burden to the health care system. Pharmacologic treatment and implanting devices are the predominant therapeutic approaches. They improve survival and have offered significant improvement in patient quality of life, but they fall short of producing an authentic remedy. Cardiac gene therapy, the introduction of genetic material to the heart, offers great promise in filling this void. In-depth knowledge of the underlying mechanisms of heart failure is, obviously, a prerequisite to achieve this aim. Extensive research in the past decades, supported by numerous methodological breakthroughs, such as transgenic animal model development, has led to a better understanding of the cardiovascular diseases and, inadvertently, to the identification of several candidate genes. Of the genes that can be targeted for gene transfer, calcium cycling proteins are prominent, as abnormalities in calcium handling are key determinants of heart failure. A major impediment, however, has been the development of a safe, yet efficient, delivery system. Nonviral vectors have been used extensively in clinical trials, but they fail to produce significant gene expression. Viral vectors, especially adenoviral, on the other hand, can produce high levels of expression, at the expense of safety. Adeno-associated viral vectors have emerged in recent years as promising myocardial gene delivery vehicles. They can sustain gene expression at a therapeutic level and maintain it over extended periods of time, even for years, and, most important, without a safety risk.
Collapse
Affiliation(s)
- Kleopatra Rapti
- Cardiovascular Research Center, Mount Sinai School of Medicine, New York, New York, USA
| | | | | |
Collapse
|
18
|
Gao MH, Hammond HK. Unanticipated signaling events associated with cardiac adenylyl cyclase gene transfer. J Mol Cell Cardiol 2011; 50:751-8. [PMID: 21354173 DOI: 10.1016/j.yjmcc.2011.02.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2010] [Revised: 02/04/2011] [Accepted: 02/07/2011] [Indexed: 12/31/2022]
Abstract
The published papers on the effects of increased cardiac expression of adenylyl cyclase type 6 (AC6) are reviewed. These include the effects of AC on normal and failing left ventricle in several pathophysiological models in mice and pigs. In addition, the effects of increased expression of AC6 in cultured neonatal and adult rat cardiac myocytes are discussed in the context of attempting to establish mechanisms for the unanticipated beneficial effects of AC6 on the failing heart. This article is part of a Special Section entitled "Special Section: Cardiovascular Gene Therapy".
Collapse
Affiliation(s)
- Mei Hua Gao
- VA San Diego Healtcare System and University of California San Diego, San Diego, CA, USA
| | | |
Collapse
|
19
|
Davis J, Westfall MV, Townsend D, Blankinship M, Herron TJ, Guerrero-Serna G, Wang W, Devaney E, Metzger JM. Designing heart performance by gene transfer. Physiol Rev 2008; 88:1567-651. [PMID: 18923190 DOI: 10.1152/physrev.00039.2007] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The birth of molecular cardiology can be traced to the development and implementation of high-fidelity genetic approaches for manipulating the heart. Recombinant viral vector-based technology offers a highly effective approach to genetically engineer cardiac muscle in vitro and in vivo. This review highlights discoveries made in cardiac muscle physiology through the use of targeted viral-mediated genetic modification. Here the history of cardiac gene transfer technology and the strengths and limitations of viral and nonviral vectors for gene delivery are reviewed. A comprehensive account is given of the application of gene transfer technology for studying key cardiac muscle targets including Ca(2+) handling, the sarcomere, the cytoskeleton, and signaling molecules and their posttranslational modifications. The primary objective of this review is to provide a thorough analysis of gene transfer studies for understanding cardiac physiology in health and disease. By comparing results obtained from gene transfer with those obtained from transgenesis and biophysical and biochemical methodologies, this review provides a global view of cardiac structure-function with an eye towards future areas of research. The data presented here serve as a basis for discovery of new therapeutic targets for remediation of acquired and inherited cardiac diseases.
Collapse
Affiliation(s)
- Jennifer Davis
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota 55455, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Kapur NK, Rade JJ. Fibroblast growth factor 4 gene therapy for chronic ischemic heart disease. Trends Cardiovasc Med 2008; 18:133-41. [PMID: 18555186 DOI: 10.1016/j.tcm.2008.03.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2008] [Revised: 03/17/2008] [Accepted: 03/20/2008] [Indexed: 11/16/2022]
Abstract
Therapeutic myocardial angiogenesis and arteriogenesis represent a novel treatment strategy for patients with angina refractory to traditional medical and surgical therapies. The fibroblast growth factors are a family of proteins that are known mediators of angio-/arteriogenesis. Based on promising preclinical animal data, a series of four randomized placebo-controlled clinical trials have been conducted to determine the safety and efficacy of local delivery of fibroblast growth factor 4 with the use of adenovirus-vector-mediated gene transfer to induce myocardial angio-/arteriogenesis in patients with stable angina. This review describes the scientific rationale underlying these clinical trials, provides an overview of their results, and discusses the implications for future studies.
Collapse
Affiliation(s)
- Navin K Kapur
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | | |
Collapse
|
21
|
Phan HM, Gao MH, Lai NC, Tang T, Hammond HK. New signaling pathways associated with increased cardiac adenylyl cyclase 6 expression: implications for possible congestive heart failure therapy. Trends Cardiovasc Med 2008; 17:215-21. [PMID: 17936202 DOI: 10.1016/j.tcm.2007.07.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2007] [Revised: 07/30/2007] [Accepted: 07/31/2007] [Indexed: 01/08/2023]
Abstract
Congestive heart failure (CHF) affects more than five million people in the United States and results in considerable morbidity, mortality, and economic costs. Patients with class III and IV CHF have a 40% to 50% probability of dying 5 years after symptom onset despite optimal therapy, a prognosis worse than many cancers. A variety of drugs and devices have improved survival-the 50% survival time in 1980 was just 18 months-but the outlook for patients remains dismal and the prevalence of CHF continues to increase. This unmet medical need underscores the importance of developing new approaches for the treatment of CHF. This brief review focuses on data from preclinical experiments regarding the effects of increased adenylyl cyclase type 6 (AC6) expression on cellular and cardiac function, and possible mechanisms for the unexpected favorable effects of increased AC6 content on the failing heart.
Collapse
Affiliation(s)
- Huy M Phan
- Department of Medicine, University of California at San Diego, CA 92093, USA
| | | | | | | | | |
Collapse
|
22
|
Affiliation(s)
- David M. Kaye
- Heart Failure Research Group, Baker Heart Research Institute, Melbourne, Victoria 8008, Australia;
| | - Masahiko Hoshijima
- Institute of Molecular Medicine, University of California, San Diego, La Jolla, California 92093-0346
| | - Kenneth R. Chien
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Stem Cell Institute, Harvard Medical School, Richard B. Simches Research Centre, Boston, Massachusetts 02114;
| |
Collapse
|
23
|
Kaye DM, Preovolos A, Marshall T, Byrne M, Hoshijima M, Hajjar R, Mariani JA, Pepe S, Chien KR, Power JM. Percutaneous cardiac recirculation-mediated gene transfer of an inhibitory phospholamban peptide reverses advanced heart failure in large animals. J Am Coll Cardiol 2007; 50:253-60. [PMID: 17631218 DOI: 10.1016/j.jacc.2007.03.047] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2006] [Revised: 03/22/2007] [Accepted: 03/28/2007] [Indexed: 12/13/2022]
Abstract
OBJECTIVES The purpose of this study was to develop a clinically applicable high-efficiency percutaneous means of therapeutic gene delivery to the failing heart. BACKGROUND Substantial advances in the understanding of the cellular and molecular basis of heart failure (HF) have recently fostered interest in the potential utility of gene and cell therapy as novel therapeutic approaches. However, successful clinical translation is currently limited by the lack of safe, efficient, and selective delivery systems. METHODS We developed a novel percutaneous closed-loop recirculatory system that provides homogeneous myocardial delivery for gene transfer in the failing large animal heart. After 4 weeks' rapid pacing in adult sheep to induce HF, the animals were randomly allocated to receive either adenovirus expressing a pseudophosphorylated mutant (AdS16E) of phospholamban (PLN) or Ad-beta-galactosidase (AdLacZ). RESULTS Two weeks after gene delivery, in the presence of continued pacing, left ventricular (LV) ejection fraction had significantly improved in the AdS16E-treated animals (27 +/- 3% to 50 +/- 4%; p < 0.001), whereas a further decline occurred in the AdLacZ group (34 +/- 4% to 27 +/- 3%; p < 0.05). In conjunction, AdS16E delivery resulted in significant reductions in LV filling pressures and end-diastolic diameter (both p < 0.05). In conjunction, AdS16E-treated animals showed significant improvement in the expression of PLN and Ca2+-adenosine triphosphatase activity. In separate animals, recirculating AdLacZ delivery was shown to achieve superior myocardial gene expression in contrast to intracoronary delivery and was associated with lower systemic expression. CONCLUSIONS We report the development of a novel closed-loop system for cardiac gene therapy. Using this approach delivery of AdS16E reversed HF progression in a large animal HF model.
Collapse
Affiliation(s)
- David M Kaye
- Baker Heart Research Institute, Melbourne, Australia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
The rationale for gene transfer of adenylyl cyclase type VI (AC(VI)) for clinical congestive heart failure (CHF) is based on recent experimental studies that have extended from cultured cardiac myocytes to preclinical studies in animal models of CHF. Over the past several years substantial data have indicated an unexpected and pronounced favorable effect of AC(VI) expression in cardiovascular disease. Preclinical studies have shown that increased cardiac AC content improves left ventricular function and attenuates deleterious remodeling in the failing heart, and reduces mortality in heart failure and in acute myocardial infarction. A brief review of the preclinical studies that have examined changes associated with increased AC expression in the heart is presented here.
Collapse
Affiliation(s)
- H Kirk Hammond
- Univerrsity of California-San Diego, La Jolla, CA 92037, USA.
| |
Collapse
|
25
|
Sasano T, Kikuchi K, McDonald AD, Lai S, Donahue JK. Targeted high-efficiency, homogeneous myocardial gene transfer. J Mol Cell Cardiol 2007; 42:954-61. [PMID: 17484913 PMCID: PMC1976378 DOI: 10.1016/j.yjmcc.2007.02.004] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2006] [Revised: 02/06/2007] [Accepted: 02/07/2007] [Indexed: 11/23/2022]
Abstract
Myocardial gene therapy continues to show promise as a tool for investigation and treatment of cardiac disease. Progress toward clinical approval has been slowed by limited in vivo delivery methods. We investigated the problem in a porcine model, with an objective of developing a method for high efficiency, homogeneous myocardial gene transfer that could be used in large mammals, and ultimately in humans. Eighty-one piglets underwent coronary catheterization for delivery of viral vectors into the left anterior descending artery and/or the great cardiac vein. The animals were followed for 5 or 28 days, and then transgene efficiency was quantified from histological samples. The baseline protocol included treatment with VEGF, nitroglycerin, and adenosine followed by adenovirus infusion into the LAD. Gene transfer efficiency varied with choice of viral vector, with use of VEGF, adenosine, or nitroglycerin, and with calcium concentration. The best results were obtained by manipulation of physical parameters. Simultaneous infusion of adenovirus through both left anterior descending artery and great cardiac vein resulted in gene transfer to 78+/-6% of myocytes in a larger target area. This method was well tolerated by the animals. We demonstrate targeted, homogeneous, high efficiency gene transfer using a method that should be transferable for eventual human usage.
Collapse
Affiliation(s)
- Tetsuo Sasano
- Heart and Vascular Research Center, MetroHealth Hospital, Case Western Reserve University School of Medicine, Cleveland, OH 44109, USA
| | | | | | | | | |
Collapse
|