Abstract
Cardiovascular diseases remain a large global health problem. Although several conventional small-molecule treatments are available for common cardiovascular problems, gene therapy is a potential treatment option for acquired and inherited cardiovascular diseases that remain with unmet clinical needs. Among potential targets for gene therapy are severe cardiac and peripheral ischemia, heart failure, vein graft failure, and some forms of dyslipidemias. The first approved gene therapy in the Western world was indicated for lipoprotein lipase deficiency, which causes high plasma triglyceride levels. With improved gene delivery methods and more efficient vectors, together with interventional transgene strategies aligned for a better understanding of the pathophysiology of these diseases, new approaches are currently tested for safety and efficacy in clinical trials. In this article, we integrate a historical perspective with recent advances that will likely affect clinical development in this research area.
Keywords: cardiovascular gene therapy, clinical trials, hyperlipidemias, ischemia, heart failure, endpoints, trial design
Ylä-Herttuala and Baker summarize recent advances and the current status of cardiovascular gene therapy with special emphasis on new clinical initiatives and lessons learned from previous clinical trials, including a discussion of the selection of patient populations, endpoints, and best available treatment strategies.
Main Text
The potential of cardiovascular gene therapy became evident in the late 1980s, when it was shown that direct intra-arterial gene transfer was possible with endovascular catheter techniques.1 At the same time, hyperlipidemias became a target for gene therapy,2 and conditions like in-stent restenosis, vein graft stenosis, heart failure, arrhythmias, refractory angina, and peripheral vascular disease were recognized as potential targets for gene therapy. In the clinics, the pioneering work of Isner et al.3 used plasmid gene transfer to treat severe peripheral vascular disease, and adenoviral vectors were, for the first time, used for local endovascular catheter-mediated gene therapy in humans.4
In spite of great enthusiasm and positive preclinical results, clinical translation of cardiovascular gene therapy has not yet been very successful. There are many factors that might contribute to this negativity, including insufficient gene delivery to the target site, undermining the potential of transgenes; too short a duration of transgene expression from particular transduction strategies; insufficient knowledge about underlying pathophysiological mechanisms, leading to a retrospectively poor strategy; and, often, underpowered clinical trial design. Several prominent trials that have produced negative results and failures have sent investigators back to the laboratory to re-evaluate and design better vectors and gene therapy approaches for cardiovascular diseases.5, 6 This bench-bedside iteration has been fruitful in other gene therapy arenas but has so far failed to have a significant impact in the cardiovascular space.
In spite of these difficulties, during the last 2–3 years, significant clinical and conceptual progress has been made in cardiovascular gene therapy. The first gene drug approved in the Western world, Glybera, is indicated for the treatment of severe lipoprotein lipase deficiency. Even though this condition is ultra-rare, it was an important milestone for the entire field of gene therapy.7 Additionally, gene delivery techniques have been significantly improved, particularly with respect to catheter-based approaches, and targeting of powerful new therapeutic genes to the myocardium has recently produced promising results.8 Thus, a new generation of cardiovascular clinical trials is primed to evaluate the potential of gene therapy in carefully selected patient populations. This wave of trials must include carefully documented gene transfer effects and objectively measurable changes in parameters like blood flow, metabolic activity, and cardiac function9 because the impact of these results cannot be underestimated. Potential targets for cardiovascular gene therapy are presented in Figure 1.
Figure 1.
Potential Targets for Cardiovascular Gene Therapy
Coronary Heart Disease
Coronary heart disease is caused by insufficient blood flow to the myocardium because of atherosclerotic stenoses in the main coronary arteries. It is well known that some patients can develop collateral arteries that rescue the myocardium in spite of the stenoses. Therefore, it is logical that this condition could be treated by providing new blood flow to the ischemic muscle.10 Coronary bypass surgery and angioplasty techniques are excellent examples of this treatment principle. However, these approaches are not possible for all patients, and there is an increasing group of severe angina pectoris patients, so-called refractory angina, who cannot be treated with these conventional approaches.11
Because the basic mechanisms of angiogenesis and blood vessel formation are already well known, therapeutic vascular growth, which tries to grow new blood vessels in the ischemic myocardium, is a potential new treatment option. Several growth factors have been used to achieve this goal in the past, but, until recently, no clinically significant results have been obtained in phase II/III clinical trials.6 The most promising candidates for therapeutic vascular growth seem to be members of the vascular endothelial growth factor (VEGF) and fibroblast growth factor (FGF) families, hepatocyte growth factor (HGF), and gene therapy approaches combined with cell therapy.12, 13
During the last decade, several randomized controlled trials have tested either naked plasmids or adenoviral vectors for the treatment of severe coronary heart disease. Trials like EuroinjectOne,14 KAT,15 REVASC,16 NOTHERN,17 NOVA,18 VEGF-Neupogen,19 and GENASIS20 have tested the intramyocardial delivery of VEGFs (Table 1). Gene transfer has also been used during bypass surgery and via minithoracotomy using epicardial injections.21, 22 Importantly, safety in these trials has been very good, even after 10-year follow-up.23, 24, 25 At the moment, there are five angiogenic gene therapy trials that are either ongoing or have recently reported final results (Table 1). Two of these trials are testing a novel VEGF-DdNdC in refractory angina patients using percutaneous endocardial delivery of adenoviral vectors.8 Targeting to the ischemic hibernating myocardium is done using a combination of electroanatomical mapping and 15O-H2O-positron emission tomography (PET), which makes it possible to treat areas that suffer from stress-induced ischemia.9 Crucially, absolute myocardial bloodflow can be measured with 15O-H2O-PET and correlated to potential clinical benefits. Because VEGF-DdNdC is both an angiogenic and lymphangiogenic growth factor, it represents a new strategy (i.e., therapeutic vascular growth) to treat refractory angina in addition to angiogenic effects: stimulation of the lymphatic circulation will help to reduce myocardial edema, which is a common side effect after proangiogenic therapies.14, 15, 16, 17, 18 VEGF-DdNdC can also potentially induce regenerative effects by inducing stem and progenitor cells in the treated muscles.26 A new trial based on intramyocardial injections via thoracotomy has also been planned. This study is based on an adenovirus expressing three major vascular endothelial growth factor-A (VEGF-A) isoforms, which should lead to a better angiogenic response by generating more natural growth factor gradients between ischemic and normoxic parts of the myocardium.27
Table 1.
Ongoing or Recent Trials in CAD and PAD
| Trial | Disease | Vector | Therapeutic Agent | Delivery | Study Design | n | Primary Endpoint | Main Result | Reference |
|---|---|---|---|---|---|---|---|---|---|
| KAT301 | CAD | Ad | VEGF-DdNdC | percutaneous i.my. NOGA/PET guided injections | phase I, RCT | 30 | safety, perfusion reserve in ischemic area (PET) | positive, increased perfusion reserve in treated areas at 1 year | 8 |
| ReGenHeart | CAD | Ad | VEGF-DdNdC | percutaneous i.my. NOGA/PET guided injections | phase II, RCT | 180 | 6-min walking test, perfusion reserve in ischemic area (PET/SPECT) | NA | NCT03039751 |
| ASPIRE | CAD | Ad | FGF-4 | percutaneous i.c. injections | phase III, open-label | 100 | reversible perfusion defect (SPECT) | NA | 29 |
| AWARE | CAD | Ad | FGF-4 | percutaneous i.c. injections | phase III, RCT | 300 | ischemic ECG changes on ETT | NA | NCT00438867 |
| VEGF-A116A | CAD | Ad | VEGF-A116A | thoracotomy i.my. injections | phase I/II, open-label | 41 | time to 1 mm ST depression on ETT | NA | NCT01757223 |
| HGF-X7 | CAD | Ad | HGF | percutaneous i.my. injections | phase I, open-label, no controls | 12 | safety | NA | 30 |
| MULTIGENE ANGIO | PAD | RV | VEGF-A165, Ang1 with cell therapy | percutaneous i.a. injections | phase I, open-label, no controls | 23 | safety, amputation-free survival | positive, amputation-free survival 72% at 1 year | 51 |
| JVS-100 | PAD | Pl | SDF-1 | i.m. injection | phase II | 48 | safety, amputation rate | NA | NCT01410331 |
| HGF-X7 (VM202) | PAD | Pl | HGF | i.m. injections | phase II | 50 | visual analog scale | NA | NCT01064440 |
| HGF-X7 (NL003) | PAD | Pl | HGF | i.m. injections | phase II, RCT | 200 | ulcer area, visual analog scale | NA | NCT01548378 |
| KAT-PAD101 | PAD | Ad | VEGF-DdNdC | i.m. injections | phase I, RCT | 30 | safety, perfusion in treated area (PET) | NA | EudraCT 001019-22 |
| Neovasculgen | PAD | Pl | VEGF-A165 | i.m. injections | phase II/III, RCT | 100 | pain-free walking distance | positive, pain-free walking distance increased 167% at 1 year | 43 |
| Previous Trials in CAD and PAD | |||||||||
| KAT | CAD | Ad, Pl | VEGF-A165 | percutaneous i.c. injection at angioplasty site | phase II, RCT | 103 | improved myocardial perfusion (SPECT) | positive, increased perfusion at 6 months (only adenovirus) | 15 |
| REVASC | CAD | Ad | VEGF-A121 | thoracotomy i.my. injections | phase II, RCT | 67 | time to 1-min ST depression on ETT | positive | 16 |
| VEGF thoracotomy | CAD | Ad | VEGF-A121 | thoracotomy i.my. injections | phase I, open-label, no controls | 21 | safety, angina classification | positive, improvement in angina classification at day 30 | 21 |
| EuroinjectOne | CAD | Pl | VEGF-A165 | percutaneous i.my. NOGA guided injections | phase II, RCT | 74 | improved myocardial perfusion (SPECT) | negative | 14 |
| Genasis | CAD | Pl | VEGF-2 | percutaneous i.my. injections | phase III, RCT | 295 | ETT | negative | 20 |
| NORTHERN | CAD | Pl | VEGF-A165 | percutaneous i.my. NOGA guided injections | phase II, RCT | 93 | change in myocardial perfusion (SPECT) | negative | 17 |
| NOVA | CAD | Ad | VEGF-A121 | percutaneous i.my. NOGA guided injections | phase I/II, RCT | 17 | ETT | negative | 18 |
| VEGF-Neupogen trial | CAD | Pl | VEGF-A165, hGCSF | percutaneous i.my. NOGA guided injections and systemic hGCSF administration | phase II, RCT | 48 | change in myocardial perfusion (SPECT) | negative | 19 |
| AGENT-3 | CAD | Ad | FGF-4 | percutaneous i.c. injections | phase III, RCT | 416 | ETT | negative (subgroup of >55-year-old women positive) | 28 |
| AGENT-4 | CAD | Ad | FGF-4 | percutaneous i.c. injections | phase III, RCT | 116 | ETT | negative | 28 |
| VEGF peripheral trial | PAD | Ad, Pl | VEGF-A165 | percutaneous i.a. injection at angioplasty site | phase II, RCT | 54 | increased vascularity in angiography | positive | 40 |
| RAVE | PAD | Ad | VEGF-A121 | i.m. injections | phase II, RCT | 105 | peak walking time | negative | 42 |
| WALK | PAD | Ad | HIF-1α/VP16 | i.m. injections | phase III, RCT | 289 | peak walking time | negative | 48 |
| Delta-1 | PAD | Pl | Del-1 | i.m. injections | phase II, RCT | 105 | peak walking time | negative | 47 |
| Groningen trial | PAD | Pl | VEGF-A165 | i.m. injections | phase II, RCT | 54 | decrease in amputation rate | negative | 41 |
| HGF-STAT | PAD | Pl | HGF | i.m. injections | phase II, RCT | 104 | limb perfusion measured as TcPO2 | positive, TcPO2 increased in high-dose group at 6 months | 53 |
| HGF study | PAD | Pl | HGF | i.m. injections | phase II, RCT | 40 | improvement in rest pain, reduction in ulcer size | positive, primary endpoint improved 70% | 54 |
| TALISMAN | PAD | Pl | FGF-1 | i.m. injections | phase II, RCT | 125 | ulcer healing | negative (secondary endpoint amputation rate positive) | 44 |
| TAMARIS | PAD | Pl | FGF-1 | i.m. injections | phase III, RCT | 525 | time to major amputation or death | negative | 45 |
CAD, coronary heart disease; PAD, peripheral vascular disease; RCT, randomized controlled trial; i.a., intra-arterial; i.c., intracoronary; i.m., intramuscular; i.my., intramyocardial; ECG, electrocardiogram; RV, retrovirus; Pl, plasmid; ETT, exercise tolerance test; hGCSF, human Granulocyte colony stimulating factor; TcPO2, transcutaneous oxygen tension; ST, ECG S-T segment; NA, not available.
Adenoviral intracoronary delivery of FGF-4 has been thoroughly tested in a series of AGENT trials.28 Currently, the ASPIRE trial will compare standard of care without a placebo group in an open-label design with a primary endpoint based on relative perfusion changes in the myocardium, as measured with 99Tc-SPECT imaging at 8 weeks.29 The randomized, double-blinded, placebo-controlled AWARE trial has also been planned with intracoronary adenovirus (Ad) FGF-4 in women with stable angina (Table 1). Both the ASPIRE and AWARE trials are based on significant positive effects in an exercise tolerance test found in AGENT 3 and 4 trials in women older than 55.28 HGF has been used both as plasmid and adenoviral constructs in the treatment of coronary heart disease, but, so far, only small open-label studies have been reported.30, 31
Potential new treatments for refractory angina include upregulation of hypoxia-inducible factor 1α, thymosin B4, modified stabilized RNAs, GalNac-antisense oligonucleotides, nanoparticles, promoter-activating small interfering RNAs (siRNAs), and exosomes, which could deliver both growth factors and microRNAs (miRNAs) to ischemic myocardium.6, 32, 33, 34, 35, 36, 37, 38 No trials are currently testing these new approaches for the treatment of severe coronary heart disease. However, thymosin B4 is an interesting candidate because it activates partly different signaling cascades than previously used growth factors.34 Also, epicardial activation of cardiomyocyte proliferation and tissue repair could prevent harmful remodeling and be a useful approach for therapy.35
Peripheral Vascular Disease
Peripheral vascular disease is caused by atherosclerotic occlusions in the main lower extremity arteries, producing symptoms like claudication, rest pain, and insufficient ulcer healing. Patients are usually older than those with coronary symptoms, which makes this population very difficult to treat. Current pharmacological treatments are not very effective for critical limb ischemia or severe peripheral arterial disease, and not all patients are suitable for endovascular operations. Therapeutic vascular growth with the same factors used in coronary heart disease gene therapy has been tested with both intra-arterial and intramuscular delivery routes. However, in a recent meta-analysis of randomized controlled trials, no consistent benefits were found of angiogenic gene therapy in peripheral vascular disease.39 This illustrates the challenges ahead for this population, but the driver for innovation is the large unmet clinical need. Some positive reports are available from both plasmid- and adenovirus-mediated VEGF-A gene therapy.40 Also, improvements in oxygenation as well as walking time and ulcer healing have been reported,41, 42 and a plasmid-based VEGF-A product has been approved for clinical use in peripheral arterial disease in Russia.43 Two large FGF-1 plasmid trials, TALISMAN44 and TAMARIS,45 did not find functional improvements in the treated patients, although the TALISMAN trial showed a beneficial effect on the amputation rate in critical ischemia patients. FGF-2 is currently tested in peripheral vascular disease (PAD) using a Sendai virus vector, but results have not been reported.46 Of the other large previous trials, plasmid Del-1 (Delta-1 trial)47 and adenoviral HIF-1α/vp16 (WALK trial),48 aiming to stimulate angiogenic signaling pathways, gave negative results.
Problems like long atherosclerotic stenoses in the main arteries, shunting and a stealing effect of the increased blood flow to less ischemic areas, unstable neovessels, and too short a time of transgene expression have been listed as potential reasons for failure.49 It appears that some basic physiological aspects of peripheral muscle perfusion have not been fully taken into account when designing previous trials, and it is likely that factors like overdilated capillaries, alterations in capillary blood transit time, and insufficient transfer of oxygen into peripheral muscles need to be taken into account when designing future clinical trials.6, 50 A recent trial aimed to improve the functionality of the induced neovessels by using a combination of retroviral VEGF-A and angiopoietin-1 gene transfers to endothelial and smooth muscle cells isolated from patients with peripheral vascular disease, whereafter the cells were injected back into the patients.51 A combination with surgical revascularization is tested in an ongoing KAT-PAD101 trial, where AdVEGF-DdNdC is given 1–2 days prior to vascular surgery to dilate capillaries and small arterioles to improve distal runoff after the vascular surgery operation.52 In addition, AdVEGF-DdNdC should induce both angiogenic and lymphangiogenic growth in the treated legs (Table 1). Two trials have tested HGF in critical limb ischemia,53, 54 and a current trial is ongoing using a plasmid that expresses two HGF isoforms. Multiple intramuscular injections at several time points are used for gene delivery. The SDF-1 plasmid is also tested in a similar setup for critical limb ischemia (Table 1).
It appears that, in the future, critical limb ischemia patients should be studied separately from less severe claudicants and that more emphasis should be given to accurate imaging and metabolic and functional studies as surrogates for the treatment effect because walking distance, amputation rate, or ulcer healing may not be able to capture improvements in the frequency of hospital admissions or related variables in these patient populations.55 It is possible that only a subgroup of peripheral vascular disease patients will benefit from therapeutic vascular growth, whereas others may actually have problems other than insufficient angiogenesis.6 For future clinical trials, it would be important to identify patient subgroups that are most likely to respond positively to the new treatments.
Heart Failure and Arrhythmias
Heart failure is an increasingly common problem in the elderly population because of the improved therapies for coronary heart disease and acute myocardial infarction. Advances in the understanding of the pathogenesis of heart failure have led to recent gene therapy trials (Table 2).5 The CUPID2 trial tested adeno-associated virus (AAV)1-sarcoplasmic reticulum calcium ATPase 2a (SERCA2a) in patients with chronic systolic heart failure or non-ischemic cardiomyopathy.56 Previous smaller trials had indicated positive effects; therefore, it was a great surprise that all endpoints in the CUPID2-trial were negative. The AAV1 vector was given with one-time intracoronary infusion, but, apparently, the gene transfer efficacy remained too low to induce any measurable positive effects.57 The AGENT-HF and SERCA-LVAD trials will test the same AAV1-SERCA2a in congestive heart failure and in patients with a left ventricle assist device (LVAD), respectively (Table 2).
Table 2.
Ongoing, Planned, or Recent Trials in HF, Hyperlipidemias, Restenosis, or Vein Graft Disease
| Trial | Disease | Vector | Therapeutic Agent | Delivery | Study Design | n | Primary Endpoint | Main Result | Reference |
|---|---|---|---|---|---|---|---|---|---|
| CUPID 2 | heart failure | AAV1 | SERCA2a | percutaneous i.c. injection | phase II, RCT | 250 | time to recurrent cardiovascular events | negative | 56 |
| SERCA-LVAD | chronic heart failure in patients with LVAD | AAV1 | SERCA2a | percutaneous i.c. injection | phase II, RCT | 24 | safety and feasibility | NA | NCT00534703 |
| AGENT-HF | heart failure | AAV1 | SERCA2a | percutaneous i.c. injections | phase II, RCT | 44 | changes in left ventricular end-systolic volume | NA | NCT01966887 |
| STOP-HF | heart failure | Pl | SDF-1 | percutaneous i.my. helical infusion catheter-mediated injections | phase II, RCT | 93 | 6-min. walking distance | negative | 58 |
| RETRO-HF | heart failure | Pl | SDF-1 | percutaneous retrograde injection via coronary vein | phase I/II, open-label part and RCT part | 52 | 6-min. walking distance | NA | NCT01961726 |
| AC6 | heart failure | Ad | adenylyl cyclase type 6 | percutaneous i.c. injections | phase I/II, RCT | 56 | combined ETT and cardiac function before and during doputamine stress | NA | NCT00787059 |
| Lp(a) | high lipoprotein Lp(a) levels | as-oligonucleotide | as-oligonucleotide against Lp(a) | weekly s.c. injections | phase II, RCT | 64 | change in plasma Lp(a) concentration | positive, 67% reduction in plasma Lp(a) | 67 |
| Lp(a)-L | high lipoprotein Lp(a) levels | GalNac-conjugated as-oligonucleotide | liver targeted as-oligonucleotide against Lp(a) | weekly s.c. injections | phase I/II, RCT | 58 | change in plasma Lp(a) concentration | positive, 66%–92% reduction in plasma Lp(a) | 67 |
| PCSK9 | high LDL cholesterol levels | siRNA | siRNA against PCSK9 mRNA | weekly/monthly s.c. injections | phase I, RCT (healthy volunteers) | 69 | change in plasma LDL cholesterol | positive, plasma LDL cholesterol reduction 51%–60% | 68 |
| apoC-III | hypertriglyceridemia | as-oligonucleotide | as-oligonucleotide against apolipoprotein C-III | weekly s.c. injections | phase II, RCT | 85 | change in plasma apoC-III level | positive, apoC-III plasma levels decreased 40%-80% | 69 |
| AAV FH | homozygous familial hypercholesterolemia because of LDL receptor mutation | AAV | LDL receptor | liver-directed delivery | phase I, open-label | 10 | safety, decrease in plasma LDL cholesterol | NA | NCT02651675 |
| Familial hypercholesterolemia trial | homozygous familial hypercholesterolemia because of LDL receptor mutation | RV | LDL receptor | ex vivo gene transfer to patient hepatocytes that were then injected back into the patient | phase I, open-label | 5 | safety, decrease in plasma LDL cholesterol level | moderate decrease in LDL cholesterol in three patients | 2 |
| Previous Trials in HF, Hyperlipidemias, Restenosis, or Vein Graft Disease | |||||||||
| Glybera trials | lipoprotein lipase deficiency | AAV1 | lipoprotein lipaseS447X | i.m. injections | phase I/II, open-label | 19 | post-Brandial hypertriglyceridemia, severe pancreatitis | positive, 50% decrease in pancreatitis, improvement in post-Brandial chylomicron metabolism | 65, 66 |
| Mipomersen trials | severe hypercholesterolemia | as-oligonucleotide | as-oligonucleotide against apolipoprotein B | weekly s.c. injections | phase III, RCT | 158 | change in plasma LDL cholesterol | positive, plasma LDL cholesterol reduction 37% | 70 |
| Italics | in-stent restenosis | as-oligonucleotide | as-oligonucleotide against c-myc | percutaneous i.c. local delivery after stent placement | phase II, RCT | 85 | percent neointimal volume obstruction by IVUS | negative | 76 |
| Prevent III | vein graft failure in PAD | oligonucleotide | E2F oligonucleotide decoy | ex vivo pressure-mediated delivery to vein graft | phase III, RCT | 1,404 | time to graft reintervention or amputation because of graft failure | negative | 79 |
| Prevent IV | vein graft failure in CABG | oligonucleotide | E2F oligonucleotide decoy | ex vivo pressure-mediated delivery to vein graft | phase III, RCT | 2,400 | angiographic vein graft failure | negative | 80 |
as, anti-sense; FH, familial hyoercholesterolemia; IVUS, intravascular ultrasound; s.c., subcutaneous; CABG, coronary artery by-pass grafting.
The STOP-HF trial tested the SDF-1 plasmid in heart failure. Therapy was given as several endomyocardial injections using a catheter device. Six-minute walking distance was used as the primary endpoint, but this trial was also negative.58 It appears that gene transfer efficiency was too low to achieve measurable positive outcomes. The RETRO-HF trial will test a similar SDF-1 plasmid using a retrograde delivery to the heart muscle. Adenoviral adenylyl cyclase 6 will be tested in congestive heart failure patients using a dose escalation format. The primary endpoints will include exercise tolerance tests and cardiac function measurements. However, the results of these trials are not yet available (Table 2). An obvious concern for the adenylyl cyclase trial is the short expression time of the adenovirus and potential immunological reactions, especially if repeated dosing will be required.
It is interesting to note that, even though heart failure is caused most commonly by advanced coronary heart disease, treatment genes for heart failure have been very different from those used for coronary heart disease and myocardial ischemia, targeting primarily excitation-contraction coupling and adverse remodeling. In the future, combination with short-term angiogenic therapies could help to balance altered cardiac metabolism and microcirculatory blood flow in failing myocardium. VEGF-B has shown several useful effects, such as cardiac-specific angiogenic activity and improvement in cardiac energy metabolism,59, 60, 61, 62 which could be tested in clinical trials. Combined gene and cell therapies could also potentially be used in future trials to achieve positive treatment outcomes.12
Heart failure develops gradually and, while in the early phase, targeting myocardial force, contractility, and adverse remodeling is useful for therapy; in the later stages, apoptosis of cardiomyocytes, fibrosis, and arrhythmias bring significant challenges for the late-state therapies. Because heart failure requires treatment of the majority of ventricular cardiomyocytes, very efficient vectors and gene delivery techniques are required for these indications. AAVs have shown significant long-term efficacy in preclinical models, but this still needs to be proven clinically. Also, the high prevalence of pre-existing antibodies against many AAV serotypes remains a concern. Recently, transfer of nucleic acids with slowly degrading nanoparticles and exosomes have become potential therapeutic approaches, and new treatment genes, such as S100A1, and vectors with better cardiac tropism are being developed for heart failure therapy.36, 37, 38, 63, 64 The best scenario would obviously be an efficient early treatment of myocardial infarction so that remodeling and progression to heart failure could be prevented. Improving blood flow to salvageable myocardium and evoking cardiomyocyte proliferation after acute myocardial infarction could be useful in this regard.
Arrhythmias are a significant cause of cardiac morbidity and mortality. Atrial fibrillation is a very common problem and can lead to significant clinical sequelae. The most severe ventricular arrhythmias and conduction system defects could theoretically be amenable to gene therapy. For example, a malfunctioning sino-atrial node could be replaced with a locally induced new focus, which could control cardiac rhythm. However, these approaches have turned out to be very difficult in vivo, and, so far, no clinical tests have been conducted in this area.
Hyperlipidemias, Lipoprotein Metabolism, and Atherosclerosis
Gene therapy could potentially be used for the treatment of severe inherited and acquired disorders of lipoprotein metabolism. The first approved gene drug in the Western world was Glybera, which is an AAV1 vector expressing lipoprotein lipase. This treatment is indicated for severe lipoprotein lipase deficiency, which causes high postprandial plasma triglyceride levels and severe pancreatitis attacks. The treatment is given by multiple intramuscular injections7, 65, 66 (Table 2).
Historically, homozygous familial hypercholesterolemia, which is caused by a mutation in low-density lipoprotein (LDL) receptor, was the first disorder where gene therapy was tested in the clinics.2 The first trial was very laborious, with a large liver resection and harvesting of hepatocytes, followed by retrovirus-mediated ex vivo gene transfer of LDL receptor and return of the cells back to the patients. The results were not very convincing, although a few patients showed a moderate reduction in plasma cholesterol level. Currently, a clinical trial is planned with an AAV vector expressing LDL receptor to treat homozygous familiar hypercholesterolemia (Table 2). Based on good clinical experiences with AAV vectors in the treatment of hemophilia, this approach sounds promising, although potential immune responses against AAV vectors remain a concern.
Promising results have been obtained with stabilized, liver-targeted antisense oligonucleotides against the lipoprotein (Lp)a. Lp(a) is an independent risk factor for coronary heart disease, and subcutaneous injections of antisense oligonucleotides have been reported to be safe and very efficient in reducing atherogenic Lp(a) levels.67 Other targets to reduce atherogenic lipoproteins are PCSK968 and apolipoprotein C-III,69 which are currently tested for human therapy based on either antisense oligonucleotides or RNAi technology (Table 2). Mipomersen is already an approved antisense drug to reduce apolipoprotein B-100 levels in familial hypercholesterolemia and in some other types of severe hypercholesterolemias.70 However, significant side effects have also been reported with mipomersen, and this drug is not used very often for therapy. Nevertheless, the liver seems to be an excellent target for antisense and RNAi therapy for lipid and lipoprotein disorders.
Some dyslipidemias and even atherosclerosis could be treated by increasing apolipoprotein A-1 levels71 because apoA-1 is a key component in antiatherogenic high-density lipoprotein (HDL) particles. Lecithin cholesterol acyl transferase is a key enzyme in HDL maturation and a potential target for therapy. Also, inhibition of microsomal triglyceride transfer proteins has shown potential to control severe atherogenic lipoprotein profiles. Because oxidized LDL is a key pathogenetic factor for the accumulation of lipids in the arterial wall,72 soluble decoy scavenger receptors, which can bind oxidized LDL and sequester it for degradation in the liver,73 or vectors expressing antibodies against oxidized LDL74 could theoretically be used as antiatherogenic therapy. However, it is clear that gene therapy approaches to control dyslipidemias and atherosclerosis can only potentially be used in very severe cases where well characterized and effective conventional therapies, such as statins and inhibitors of cholesterol absorption, cannot achieve the desired treatment effects.
Restenosis, In-stent Restenosis, and Vein Graft Disease
Intravascular interventions to target occlusions in the coronary arteries inevitably cause marked damage to the endothelium and arterial medial layer, exposing thrombogenic molecules. Activation of platelets, thrombosis, leukocyte adhesion, smooth muscle cell proliferation, matrix accumulation, and vascular remodeling are all involved in the pathogenesis of restenosis; i.e., the response to injury, leading to re-narrowing of the vessel lumen.75 Drug-eluting stents have significantly improved treatment outcomes and reduced remodeling, recoil, and acute obstruction of the treated arteries, although late stent thrombosis still occurs in some patients. However, it is difficult to see how gene therapy could be competitive with current drug-eluting stents. Also, a previous clinical trial in restenosis gave negative results76 (Table 2).
Vein graft stenosis is another common problem after bypass surgery. With most bypass operations still using the autologous saphenous vein as a conduit for bypass, there remains a significant risk of failure in the long term. The rationale for a gene therapy approach is to prevent pathological vein graft remodeling in the immediate time after implantation as the vein adapts to higher blood pressure and surgical handling. Vein grafts offer an excellent opportunity for local intraluminal or perivascular gene transfer during the preparation of grafts ex vivo.77, 78 Tissue inhibitor of metalloproteinases (TIMPs) have been suggested as a potential treatment,77, 78 but no clinical results are yet available from this approach because of problems in manufacturing clinical-grade AdTIMP-3. E2F transcription factor antisense decoys have been tried in clinics, but the results were disappointing79, 80 (Table 2). Vascular grafts and dialysis anastomosis stenosis could also be treated using gene transfer from the adventitial surface of the vessels. Phase I/II studies have indicated that local transduction with VEGF could reduce stenosis, presumably by increasing nitric oxide and prostacyclin production in the grafts. However, final clinical trial results are not available from these studies.81
Lessons Learned and Future Perspectives
Gene Delivery Vectors and Transduction Efficiency
It appears that the gene transfer efficiency in many previous cardiovascular trials has been too low to achieve meaningful clinical effects. Usually, in preclinical animal models, much higher doses per kilogram of gene transfer vectors have been used than what is possible in human trials.82 Intra-arterial delivery methods seem to be much less efficient than intramyocardial or intramuscular injections.4 As a consequence, it is likely that the concentration of therapeutic proteins in the target tissues has not reached sufficient levels and/or has not persisted long enough to achieve biological effects. Secreted therapeutic proteins have a better likelihood to give positive effects because, currently, it is very difficult to achieve more than 10%–20% transduction efficiency in human heart or peripheral muscles. Factors like volume per injection and matrix binding properties of the vectors and transgenes affect the distribution of transgene products in the treated tissues. It appears that physical (i.e., by surgery or via a catheter) or, possibly, genetic targeting of vectors would be desirable. In therapeutic vascular growth, the expression time of a transgene should be long enough to cause biological effects, but too long an expression time of powerful factors like VEGF will be detrimental to the tissue architecture.83 In the future, it would be important to develop vectors where transgene expression could be regulated by small molecules or physiological stimuli. Integrating vectors can best be used for applications where genetic defects require a lifelong treatment effect. It would be important to determine, in preclinical studies and in phase 1 trials, that the transgene product can really be measured either in the plasma or in the target tissues and that a dose effect can be demonstrated.84 Troubleshooting of the main problems in clinical gene therapy trials is presented in Table 3.
Table 3.
Troubleshooting of Clinical Cardiovascular Gene Therapy Trials
| Reasons | Potential Solutions | |
|---|---|---|
| General Problems | ||
| Lack of clinical efficacy in randomized controlled trials |
low gene transfer efficiency in target tissues (especially with naked plasmids and intra-arterial gene transfer routes) | intramyocardial or intramuscular gene transfer routes |
| adenoviral or AAV dose too low in i.m. delivery | optimized viral dose, determine dose-response in the clinics | |
| short half-life of transduced therapeutic factors | enhance spreading of gene transfer vectors in the target tissues using sufficient injection volumes and number of injections | |
| difficult or unresponsive patient populations | use of gene transfer as adjuvant therapy in combination with conventional treatments | |
| strong placebo effects | use of less severely affected patients who can potentially respond to new therapies | |
| randomized, controlled, blinded study design | ||
| Specific Problems on Measured Variables | ||
| No detectable effects on clinical symptoms, organ function, metabolism, or imaging endpoints | gene transfer efficiency too low to cause physiological effects | perfusion measurements shortly after gene transfer to show proof of concept |
| insufficient distribution of vector or transgene product because of binding to extracellular matrix | careful dose optimization in preclinical and early clinical studies | |
| regression of new vessels too fast after transient transgene expression | use of gene transfer vectors that produce long-term and/or regulated gene expression | |
| increased blood flow may cause shunting in target tissues | development and validation of more sensitive surrogate endpoints for imaging, organ function, and target tissue metabolism | |
| lack of validated, sensitive surrogate endpoints | understanding and controlling immunoresponses against vectors and transgenes | |
| wrong time points for endpoint measurements | combination of gene therapy with established treatment procedures | |
| immune responses against vectors or transgenes | ||
| pathophysiology still incompletely understood | ||
Patient Populations
In many previous trials, severe “no option” patients who are no longer eligible for any other kind of treatments have been recruited. However, in future ischemia and heart failure trials, healthier patients should be recruited. It is possible that, in severe ischemia patients, one of the main reasons for the difficult clinical situation is that endogenous angiogenesis has failed in the first place and that these patients are no longer able to react to angiogenic therapies. It is likely that only some subgroups of patients will respond positively to gene therapy approaches.8 It would be highly desirable to find biomarkers or other characteristics that would help to select the most optimal patients for future clinical trials.6
The placebo effect has been very strong in many previous cardiovascular gene therapy trials, and only a randomized, blinded, controlled trial design can give reliable results regarding clinical benefits.82 The reasons for the strong placebo effect are partly unknown, but patients usually go through a heavy pre-evaluation screening process with multiple laboratory and imaging tests, and gene therapy itself raises high expectations in severely affected patients. Therefore, it is not surprising that placebo groups have reacted positively. It should also be kept in mind that, in intramuscular injections, tissue injury caused by the needle or catheter can lead to local inflammation and production of growth factors and cause some positive effects. These factors should be taken into account when planning control groups for future clinical trials (Table 3).
Endpoints
Whether technical or pharmacological shortcomings of gene drugs in previous trials have caused clinical failures remains unknown. However, traditional endpoints, such as survival, exercise tolerance, amputation frequency, or ulcer healing, seem to be very demanding in severely affected patients.6 There is a clear need to develop validated surrogate endpoints for cardiovascular trials based on tissue perfusion, collateral flow, metabolic improvements, and reduced burden of the use of hospital services that could better capture potential therapeutic benefits. This, together with selection of patients who are most likely to benefit from new therapies, should be taken into account in future clinical trials.6, 8
An evaluation of pre-existing antibody levels and immunological responses to the treatments should be included in every trial and correlated with the potential treatment results. In future trials, gene therapies should also be combined with existing therapies, such as bypass surgery or angioplasty as adjuvant therapies. This would allow new approaches for therapy, such as in peripheral vascular disease, where gene transfer is given prior to bypass operation to open peripheral capillaries and to avoid a poor run-off syndrome, which often leads to disappointing results in these patients.6, 50, 52 Other co-morbidities can significantly affect therapeutic outcomes, and, for this reason, patients should be accurately stratified for diseases like diabetes, rheumatoid arthritis, or other autoimmune diseases to avoid confounding results. Most therapeutic approaches have so far been based on single-dose applications. It should be evaluated whether repeated dosing could be applied because many chronic cardiovascular diseases are likely to progress, and the need for future therapeutic interventions is quite obvious.
As far as the safety profile of cardiovascular gene therapy is concerned, most trials have shown a very good safety profile even after 10-year follow-up.23, 24, 25 This should encourage clinical testing of new therapeutic approaches with improved vectors and gene delivery methods. Current imaging methods can be used to guide therapeutic approaches so that very low doses could be applied to targets like ischemic myocardium in combination with standard therapies. Because a regulatory pathway for the approval of gene drugs has now been established, it is expected that several novel treatments for cardiovascular diseases will enter clinical testing in the near future.
Acknowledgments
This review was supported by the Finnish Academy Center of Excellence in Cardiovascular and Metabolic Diseases, the British Heart Foundation Chair of Translational Cardiovascular Sciences, and ERC advanced grants (VASCMIR and CleverGenes). The authors thank Ms. Marjo-Riitta Salminkoski for preparing the manuscript.
References
- 1.Nabel E.G., Plautz G., Nabel G.J. Site-specific gene expression in vivo by direct gene transfer into the arterial wall. Science. 1990;249:1285–1288. doi: 10.1126/science.2119055. [DOI] [PubMed] [Google Scholar]
- 2.Grossman M., Rader D.J., Muller D.W., Kolansky D.M., Kozarsky K., Clark B.J., 3rd, Stein E.A., Lupien P.J., Brewer H.B., Jr., Raper S.E. A pilot study of ex vivo gene therapy for homozygous familial hypercholesterolaemia. Nat. Med. 1995;1:1148–1154. doi: 10.1038/nm1195-1148. [DOI] [PubMed] [Google Scholar]
- 3.Isner J.M., Pieczek A., Schainfeld R., Blair R., Haley L., Asahara T., Rosenfield K., Razvi S., Walsh K., Symes J.F. Clinical evidence of angiogenesis after arterial gene transfer of phVEGF165 in patient with ischaemic limb. Lancet. 1996;348:370–374. doi: 10.1016/s0140-6736(96)03361-2. [DOI] [PubMed] [Google Scholar]
- 4.Laitinen M., Mäkinen K., Manninen H., Matsi P., Kossila M., Agrawal R.S., Pakkanen T., Luoma J.S., Viita H., Hartikainen J. Adenovirus-mediated gene transfer to lower limb artery of patients with chronic critical leg ischemia. Hum. Gene Ther. 1998;9:1481–1486. doi: 10.1089/hum.1998.9.10-1481. [DOI] [PubMed] [Google Scholar]
- 5.Hulot J.S., Ishikawa K., Hajjar R.J. Gene therapy for the treatment of heart failure: promise postponed. Eur. Heart J. 2016;37:1651–1658. doi: 10.1093/eurheartj/ehw019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Ylä-Herttuala S., Bridges C., Katz M.G., Korpisalo P. Angiogenic gene therapy in cardiovascular diseases: dream or vision? Eur. Heart J. 2017;2017:10. doi: 10.1093/eurheartj/ehw547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Ylä-Herttuala S. Endgame: glybera finally recommended for approval as the first gene therapy drug in the European union. Mol. Ther. 2012;20:1831–1832. doi: 10.1038/mt.2012.194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Hartikainen, J., Hassinen, I., Hedman, A., Kivelä, A., Saraste, A., Knuuti, J., Husso, M., Mussalo, H., Hedman, M., Rissanen, T.T., et al. (2017) Adenoviral intramyocardial VEGF-DΔNΔC gene transfer increases myocardial perfusion reserve in refractory angina patients. A phase I/IIa study with one year follow-up. Submitted. [DOI] [PMC free article] [PubMed]
- 9.Hassinen I., Kivelä A., Hedman A., Saraste A., Knuuti J., Hartikainen J.E., Ylä-Herttuala S. Intramyocardial gene therapy directed to hibernating heart muscle using a combination of electromechanical mapping and positron emission tomography. Hum. Gene Ther. 2016;27:830–834. doi: 10.1089/hum.2016.131. [DOI] [PubMed] [Google Scholar]
- 10.Ylä-Herttuala S., Alitalo K. Gene transfer as a tool to induce therapeutic vascular growth. Nat. Med. 2003;9:694–701. doi: 10.1038/nm0603-694. [DOI] [PubMed] [Google Scholar]
- 11.Henry T.D., Satran D., Jolicoeur E.M. Treatment of refractory angina in patients not suitable for revascularization. Nat. Rev. Cardiol. 2014;11:78–95. doi: 10.1038/nrcardio.2013.200. [DOI] [PubMed] [Google Scholar]
- 12.Gyöngyösi M., Wojakowski W., Lemarchand P., Lunde K., Tendera M., Bartunek J., Marban E., Assmus B., Henry T.D., Traverse J.H., ACCRUE Investigators Meta-Analysis of Cell-based CaRdiac stUdiEs (ACCRUE) in patients with acute myocardial infarction based on individual patient data. Circ. Res. 2015;116:1346–1360. doi: 10.1161/CIRCRESAHA.116.304346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Banovic M., Loncar Z., Behfar A., Vanderheyden M., Beleslin B., Zeiher A., Metra M., Terzic A., Bartunek J. Endpoints in stem cell trials in ischemic heart failure. Stem Cell Res. Ther. 2015;6:159. doi: 10.1186/s13287-015-0143-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Kastrup J., Jørgensen E., Rück A., Tägil K., Glogar D., Ruzyllo W., Bøtker H.E., Dudek D., Drvota V., Hesse B., Euroinject One Group Direct intramyocardial plasmid vascular endothelial growth factor-A165 gene therapy in patients with stable severe angina pectoris A randomized double-blind placebo-controlled study: the Euroinject One trial. J. Am. Coll. Cardiol. 2005;45:982–988. doi: 10.1016/j.jacc.2004.12.068. [DOI] [PubMed] [Google Scholar]
- 15.Hedman M., Hartikainen J., Syvänne M., Stjernvall J., Hedman A., Kivelä A., Vanninen E., Mussalo H., Kauppila E., Simula S. Safety and feasibility of catheter-based local intracoronary vascular endothelial growth factor gene transfer in the prevention of postangioplasty and in-stent restenosis and in the treatment of chronic myocardial ischemia: phase II results of the Kuopio Angiogenesis Trial (KAT) Circulation. 2003;107:2677–2683. doi: 10.1161/01.CIR.0000070540.80780.92. [DOI] [PubMed] [Google Scholar]
- 16.Stewart D.J., Hilton J.D., Arnold J.M., Gregoire J., Rivard A., Archer S.L., Charbonneau F., Cohen E., Curtis M., Buller C.E. Angiogenic gene therapy in patients with nonrevascularizable ischemic heart disease: a phase 2 randomized, controlled trial of AdVEGF(121) (AdVEGF121) versus maximum medical treatment. Gene Ther. 2006;13:1503–1511. doi: 10.1038/sj.gt.3302802. [DOI] [PubMed] [Google Scholar]
- 17.Stewart D.J., Kutryk M.J.B., Fitchett D., Freeman M., Camack N., Su Y., Siega A.D., Bilodeau L., Burton J.R., Proulx G. VEGF gene therapy fails to improve perfusion of ischemic myocardium in patients with advanced coronary disease: results of the NORTHERN trial. Mol. Ther. 2009;17:1109–1115. doi: 10.1038/mt.2009.70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Kastrup J., Jørgensen E., Fuchs S., Nikol S., Bøtker H.E., Gyöngyösi M., Glogar D., Kornowski R. A randomised, double-blind, placebo-controlled, multicentre study of the safety and efficacy of BIOBYPASS (AdGVVEGF121.10NH) gene therapy in patients with refractory advanced coronary artery disease: the NOVA trial. EuroIntervention. 2011;6:813–818. doi: 10.4244/EIJV6I7A140. [DOI] [PubMed] [Google Scholar]
- 19.Ripa R.S., Wang Y., Jørgensen E., Johnsen H.E., Hesse B., Kastrup J. Intramyocardial injection of vascular endothelial growth factor-A165 plasmid followed by granulocyte-colony stimulating factor to induce angiogenesis in patients with severe chronic ischaemic heart disease. Eur. Heart J. 2006;27:1785–1792. doi: 10.1093/eurheartj/ehl117. [DOI] [PubMed] [Google Scholar]
- 20.Corautus Genetics (2006). Corautus announces termination of patient enrollment in GENASIS severe angina clinical trial. http://www.corautus.com.
- 21.Rosengart T.K., Lee L.Y., Patel S.R., Sanborn T.A., Parikh M., Bergman G.W., Hachamovitch R., Szulc M., Kligfield P.D., Okin P.M. Angiogenesis gene therapy: phase I assessment of direct intramyocardial administration of an adenovirus vector expressing VEGF121 cDNA to individuals with clinically significant severe coronary artery disease. Circulation. 1999;100:468–474. doi: 10.1161/01.cir.100.5.468. [DOI] [PubMed] [Google Scholar]
- 22.Kilian E.G., Sadoni S., Vicol C., Kelly R., van Hulst K., Schwaiger M., Kupatt C., Boekstegers P., Pillai R., Channon K. Myocardial transfection of hypoxia inducible factor-1alpha via an adenoviral vector during coronary artery bypass grafting. - A multicenter phase I and safety study - Circ. J. 2010;74:916–924. doi: 10.1253/circj.cj-09-0594. [DOI] [PubMed] [Google Scholar]
- 23.Hedman M., Muona K., Hedman A., Kivelä A., Syvänne M., Eränen J., Rantala A., Stjernvall J., Nieminen M.S., Hartikainen J., Ylä-Herttuala S. Eight-year safety follow-up of coronary artery disease patients after local intracoronary VEGF gene transfer. Gene Ther. 2009;16:629–634. doi: 10.1038/gt.2009.4. [DOI] [PubMed] [Google Scholar]
- 24.Rosengart T.K., Bishawi M.M., Halbreiner M.S., Fakhoury M., Finnin E., Hollmann C., Shroyer A.L., Crystal R.G. 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. 2013;24:203–208. doi: 10.1089/hum.2012.137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.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. 2012;19:392–395. doi: 10.1038/gt.2011.109. [DOI] [PubMed] [Google Scholar]
- 26.Koponen J.K., Kekarainen T., Heinonen S.E., Laitinen A., Nystedt J., Laine J., Ylä-Herttuala S. Umbilical cord blood-derived progenitor cells enhance muscle regeneration in mouse hindlimb ischemia model. Mol. Ther. 2007;15:2172–2177. doi: 10.1038/sj.mt.6300302. [DOI] [PubMed] [Google Scholar]
- 27.Kaminsky S.M., Quach L., Chen S., Pierre-Destine L., Van de Graaf B., Monette S., Rosenberg J.B., De B.P., Sondhi D., Hackett N.R. Safety of direct cardiac administration of AdVEGF-All6A+, a replication-deficient adenovirus vector cDNA/genomic hybrid expressing all three major isoforms of human vascular endothelial growth factor, to the ischemic myocardium of rats. Hum. Gene Ther. Clin. Dev. 2013;24:38–46. doi: 10.1089/humc.2013.054. [DOI] [PubMed] [Google Scholar]
- 28.Henry T.D., Grines C.L., Watkins M.W., Dib N., Barbeau G., Moreadith R., Andrasfay T., Engler R.L. Effects of Ad5FGF-4 in patients with angina: an analysis of pooled data from the AGENT-3 and AGENT-4 trials. J. Am. Coll. Cardiol. 2007;50:1038–1046. doi: 10.1016/j.jacc.2007.06.010. [DOI] [PubMed] [Google Scholar]
- 29.Kaski J.C., Consuegra-Sanchez L. Evaluation of ASPIRE trial: a Phase III pivotal registration trial, using intracoronary administration of Generx (Ad5FGF4) to treat patients with recurrent angina pectoris. Expert Opin. Biol. Ther. 2013;13:1749–1753. doi: 10.1517/14712598.2013.827656. [DOI] [PubMed] [Google Scholar]
- 30.Yang Z.J., Zhang Y.R., Chen B., Zhang S.L., Jia E.Z., Wang L.S., Zhu T.B., Li C.J., Wang H., Huang J. Phase I clinical trial on intracoronary administration of Ad-hHGF treating severe coronary artery disease. Mol. Biol. Rep. 2009;36:1323–1329. doi: 10.1007/s11033-008-9315-3. [DOI] [PubMed] [Google Scholar]
- 31.Kim J.S., Hwang H.Y., Cho K.R., Park E.A., Lee W., Paeng J.C., Lee D.S., Kim H.K., Sohn D.W., Kim K.B. Intramyocardial transfer of hepatocyte growth factor as an adjunct to CABG: phase I clinical study. Gene Ther. 2013;20:717–722. doi: 10.1038/gt.2012.87. [DOI] [PubMed] [Google Scholar]
- 32.Turunen M.P., Lehtola T., Heinonen S.E., Assefa G.S., Korpisalo P., Girnary R., Glass C.K., Väisänen S., Ylä-Herttuala S. Efficient regulation of VEGF expression by promoter-targeted lentiviral shRNAs based on epigenetic mechanism: a novel example of epigenetherapy. Circ. Res. 2009;105:604–609. doi: 10.1161/CIRCRESAHA.109.200774. [DOI] [PubMed] [Google Scholar]
- 33.Ziegler T., Hinkel R., Stöhr A., Eschenhagen T., Laugwitz K.L., le Noble F., David R., Hansen A., Kupatt C. Thymosin β4 Improves Differentiation and Vascularization of EHTs. Stem Cells Int. 2017;2017:6848271. doi: 10.1155/2017/6848271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Hinkel R., Trenkwalder T., Kupatt C. Molecular and cellular mechanisms of thymosin β4-mediated cardioprotection. Ann. N Y Acad. Sci. 2012;1269:102–109. doi: 10.1111/j.1749-6632.2012.06693.x. [DOI] [PubMed] [Google Scholar]
- 35.Smart N., Bollini S., Dubé K.N., Vieira J.M., Zhou B., Davidson S., Yellon D., Riegler J., Price A.N., Lythgoe M.F. De novo cardiomyocytes from within the activated adult heart after injury. Nature. 2011;474:640–644. doi: 10.1038/nature10188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Henri O., Pouehe C., Houssari M., Galas L., Nicol L., Edwards-Lévy F., Henry J.P., Dumesnil A., Boukhalfa I., Banquet S. Selective Stimulation of Cardiac Lymphangiogenesis Reduces Myocardial Edema and Fibrosis Leading to Improved Cardiac Function Following Myocardial Infarction. Circulation. 2016;133:1484–1497. doi: 10.1161/CIRCULATIONAHA.115.020143. discussion 1497. [DOI] [PubMed] [Google Scholar]
- 37.Beltrami C., Besnier M., Shantikumar S., Shearn A.I., Rajakaruna C., Laftah A., Sessa F., Spinetti G., Petretto E., Angelini G.D., Emanueli C. Human Pericardial Fluid Contains Exosomes Enriched with Cardiovascular-Expressed MicroRNAs and Promotes Therapeutic Angiogenesis. Mol. Ther. 2017;25:679–693. doi: 10.1016/j.ymthe.2016.12.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Wang C., Zhang C., Liu L., A X., Chen B., Li Y., Du J. Macrophage-Derived mir-155-Containing Exosomes Suppress Fibroblast Proliferation and Promote Fibroblast Inflammation during Cardiac Injury. Mol. Ther. 2017;25:192–204. doi: 10.1016/j.ymthe.2016.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Hammer A., Steiner S. Gene therapy for therapeutic angiogenesis in peripheral arterial disease - a systematic review and meta-analysis of randomized, controlled trials. Vasa. 2013;42:331–339. doi: 10.1024/0301-1526/a000298. [DOI] [PubMed] [Google Scholar]
- 40.Mäkinen K., Manninen H., Hedman M., Matsi P., Mussalo H., Alhava E., Ylä-Herttuala S. Increased vascularity detected by digital subtraction angiography after VEGF gene transfer to human lower limb artery: a randomized, placebo-controlled, double-blinded phase II study. Mol. Ther. 2002;6:127–133. doi: 10.1006/mthe.2002.0638. [DOI] [PubMed] [Google Scholar]
- 41.Kusumanto Y.H., van Weel V., Mulder N.H., Smit A.J., van den Dungen J.J.A.M., Hooymans J.M., Sluiter W.J., Tio R.A., Quax P.H., Gans R.O. Treatment with intramuscular vascular endothelial growth factor gene compared with placebo for patients with diabetes mellitus and critical limb ischemia: a double-blind randomized trial. Hum. Gene Ther. 2006;17:683–691. doi: 10.1089/hum.2006.17.683. [DOI] [PubMed] [Google Scholar]
- 42.Rajagopalan S., Mohler E.R., 3rd, Lederman R.J., Mendelsohn F.O., Saucedo J.F., Goldman C.K., Blebea J., Macko J., Kessler P.D., Rasmussen H.S., Annex B.H. Regional angiogenesis with vascular endothelial growth factor in peripheral arterial disease: a phase II randomized, double-blind, controlled study of adenoviral delivery of vascular endothelial growth factor 121 in patients with disabling intermittent claudication. Circulation. 2003;108:1933–1938. doi: 10.1161/01.CIR.0000093398.16124.29. [DOI] [PubMed] [Google Scholar]
- 43.Deev R.V., Bozo I.Y., Mzhavanadze N.D., Voronov D.A., Gavrilenko A.V., Chervyakov Y.V., Staroverov I.N., Kalinin R.E., Shvalb P.G., Isaev A.A. pCMV-vegf165 Intramuscular Gene Transfer is an Effective Method of Treatment for Patients With Chronic Lower Limb Ischemia. J. Cardiovasc. Pharmacol. Ther. 2015;20:473–482. doi: 10.1177/1074248415574336. [DOI] [PubMed] [Google Scholar]
- 44.Nikol S., Baumgartner I., Van Belle E., Diehm C., Visoná A., Capogrossi M.C., Ferreira-Maldent N., Gallino A., Wyatt M.G., Wijesinghe L.D. Therapeutic angiogenesis with intramuscular NV1FGF improves amputation-free survival in patients with critical limb ischemia. Mol. Ther. 2008;16:972–978. doi: 10.1038/mt.2008.33. [DOI] [PubMed] [Google Scholar]
- 45.Belch J., Hiatt W.R., Baumgartner I., Driver I.V., Nikol S., Norgren L., Van Belle E., TAMARIS Committees and Investigators Effect of fibroblast growth factor NV1FGF on amputation and death: a randomised placebo-controlled trial of gene therapy in critical limb ischaemia. Lancet. 2011;377:1929–1937. doi: 10.1016/S0140-6736(11)60394-2. [DOI] [PubMed] [Google Scholar]
- 46.Yonemitsu Y., Matsumoto T., Maehara Y. Gene Therapy for peripheral arterial disease using Sendai Virus vector: from preclinical studies to the Phase I/IIa clinical trial. In: Nagai Y., editor. Sendai Virus Vector. First Edition. Springer; Japan: 2013. pp. 183–201. [Google Scholar]
- 47.Grossman P.M., Mendelsohn F., Henry T.D., Hermiller J.B., Litt M., Saucedo J.F., Weiss R.J., Kandzari D.E., Kleiman N., Anderson R.D. Results from a phase II multicenter, double-blind placebo-controlled study of Del-1 (VLTS-589) for intermittent claudication in subjects with peripheral arterial disease. Am. Heart J. 2007;153:874–880. doi: 10.1016/j.ahj.2007.01.038. [DOI] [PubMed] [Google Scholar]
- 48.Creager M.A., Olin J.W., Belch J.J.F., Moneta G.L., Henry T.D., Rajagopalan S., Annex B.H., Hiatt W.R. Effect of hypoxia-inducible factor-1alpha gene therapy on walking performance in patients with intermittent claudication. Circulation. 2011;124:1765–1773. doi: 10.1161/CIRCULATIONAHA.110.009407. [DOI] [PubMed] [Google Scholar]
- 49.Ylä-Herttuala S., Rissanen T.T., Vajanto I., Hartikainen J. Vascular endothelial growth factors: biology and current status of clinical applications in cardiovascular medicine. J. Am. Coll. Cardiol. 2007;49:1015–1026. doi: 10.1016/j.jacc.2006.09.053. [DOI] [PubMed] [Google Scholar]
- 50.Dragneva G., Korpisalo P., Ylä-Herttuala S. Promoting blood vessel growth in ischemic diseases: challenges in translating preclinical potential into clinical success. Dis. Model. Mech. 2013;6:312–322. doi: 10.1242/dmm.010413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Flugelman M.Y., Halak M., Yoffe B., Schneiderman J., Rubinstein C., Bloom A.I., Weinmann E., Goldin I., Ginzburg V., Mayzler O. Phase Ib Safety, Two-Dose Study of MultiGeneAngio in Patients with Chronic Critical Limb Ischemia. Mol. Ther. 2017;25:816–825. doi: 10.1016/j.ymthe.2016.12.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Vajanto I., Korpisalo P., Karjalainen J., Hakala T., Mäkinen K., Ylä-Herttuala S. Antegrade flow and peripheral resistance determine the level of endogenous arteriogenesis in patients with superficial femoral artery occlusion. Eur. J. Clin. Invest. 2009;39:1048–1054. doi: 10.1111/j.1365-2362.2009.02208.x. [DOI] [PubMed] [Google Scholar]
- 53.Powell R.J., Simons M., Mendelsohn F.O., Daniel G., Henry T.D., Koga M., Morishita R., Annex B.H. Results of a double-blind, placebo-controlled study to assess the safety of intramuscular injection of hepatocyte growth factor plasmid to improve limb perfusion in patients with critical limb ischemia. Circulation. 2008;118:58–65. doi: 10.1161/CIRCULATIONAHA.107.727347. [DOI] [PubMed] [Google Scholar]
- 54.Shigematsu H., Yasuda K., Iwai T., Sasajima T., Ishimaru S., Ohashi Y., Yamaguchi T., Ogihara T., Morishita R. Randomized, double-blind, placebo-controlled clinical trial of hepatocyte growth factor plasmid for critical limb ischemia. Gene Ther. 2010;17:1152–1161. doi: 10.1038/gt.2010.51. [DOI] [PubMed] [Google Scholar]
- 55.Halonen P.J., Nurro J., Kuivanen A., Ylä-Herttuala S. Current gene therapy trials for vascular diseases. Expert Opin. Biol. Ther. 2014;14:327–336. doi: 10.1517/14712598.2014.872237. [DOI] [PubMed] [Google Scholar]
- 56.Greenberg B., Butler J., Felker G.M., Ponikowski P., Voors A.A., Desai A.S., Barnard D., Bouchard A., Jaski B., Lyon A.R. Calcium upregulation by percutaneous administration of gene therapy in patients with cardiac disease (CUPID 2): a randomised, multinational, double-blind, placebo-controlled, phase 2b trial. Lancet. 2016;387:1178–1186. doi: 10.1016/S0140-6736(16)00082-9. [DOI] [PubMed] [Google Scholar]
- 57.Ylä-Herttuala S. Gene Therapy for Heart Failure: Back to the Bench. Mol. Ther. 2015;23:1551–1552. doi: 10.1038/mt.2015.158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Chung E.S., Miller L., Patel A.N., Anderson R.D., Mendelsohn F.O., Traverse J., Silver K.H., Shin J., Ewald G., Farr M.J. Changes in ventricular remodelling and clinical status during the year following a single administration of stromal cell-derived factor-1 non-viral gene therapy in chronic ischaemic heart failure patients: the STOP-HF randomized Phase II trial. Eur. Heart J. 2015;36:2228–2238. doi: 10.1093/eurheartj/ehv254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Karpanen T., Bry M., Ollila H.M., Seppänen-Laakso T., Liimatta E., Leskinen H., Kivelä R., Helkamaa T., Merentie M., Jeltsch M. Overexpression of vascular endothelial growth factor-B in mouse heart alters cardiac lipid metabolism and induces myocardial hypertrophy. Circ. Res. 2008;103:1018–1026. doi: 10.1161/CIRCRESAHA.108.178459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Lähteenvuo J.E., Lähteenvuo M.T., Kivelä A., Rosenlew C., Falkevall A., Klar J., Heikura T., Rissanen T.T., Vähäkangas E., Korpisalo P. Vascular endothelial growth factor-B induces myocardium-specific angiogenesis and arteriogenesis via vascular endothelial growth factor receptor-1- and neuropilin receptor-1-dependent mechanisms. Circulation. 2009;119:845–856. doi: 10.1161/CIRCULATIONAHA.108.816454. [DOI] [PubMed] [Google Scholar]
- 61.Hagberg C.E., Falkevall A., Wang X., Larsson E., Huusko J., Nilsson I., van Meeteren L.A., Samen E., Lu L., Vanwildemeersch M. Vascular endothelial growth factor B controls endothelial fatty acid uptake. Nature. 2010;464:917–921. doi: 10.1038/nature08945. [DOI] [PubMed] [Google Scholar]
- 62.Woitek F., Zentilin L., Hoffman N.E., Powers J.C., Ottiger I., Parikh S., Kulczycki A.M., Hurst M., Ring N., Wang T. Intracoronary Cytoprotective Gene Therapy: A Study of VEGF-B167 in a Pre-Clinical Animal Model of Dilated Cardiomyopathy. J. Am. Coll. Cardiol. 2015;66:139–153. doi: 10.1016/j.jacc.2015.04.071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Pleger S.T., Shan C., Ksienzyk J., Bekeredjian R., Boekstegers P., Hinkel R., Schinkel S., Leuchs B., Ludwig J., Qiu G. Cardiac AAV9-S100A1 gene therapy rescues post-ischemic heart failure in a preclinical large animal model. Sci. Transl. Med. 2011;3:92ra64. doi: 10.1126/scitranslmed.3002097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Ishikawa K., Fish K.M., Tilemann L., Rapti K., Aguero J., Santos-Gallego C.G., Lee A., Karakikes I., Xie C., Akar F.G. Cardiac I-1c overexpression with reengineered AAV improves cardiac function in swine ischemic heart failure. Mol. Ther. 2014;22:2038–2045. doi: 10.1038/mt.2014.127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Carpentier A.C., Frisch F., Labbé S.M., Gagnon R., de Wal J., Greentree S., Petry H., Twisk J., Brisson D., Gaudet D. Effect of alipogene tiparvovec (AAV1-LPL(S447X)) on postprandial chylomicron metabolism in lipoprotein lipase-deficient patients. J. Clin. Endocrinol. Metab. 2012;97:1635–1644. doi: 10.1210/jc.2011-3002. [DOI] [PubMed] [Google Scholar]
- 66.Gaudet D., Stroes E.S., Méthot J., Brisson D., Tremblay K., Bernelot Moens S.J., Iotti G., Rastelletti I., Ardigo D., Corzo D. Long-Term Retrospective Analysis of Gene Therapy with Alipogene Tiparvovec and Its Effect on Lipoprotein Lipase Deficiency-Induced Pancreatitis. Hum. Gene Ther. 2016;27:916–925. doi: 10.1089/hum.2015.158. [DOI] [PubMed] [Google Scholar]
- 67.Viney N.J., van Capelleveen J.C., Geary R.S., Xia S., Tami J.A., Yu R.Z., Marcovina S.M., Hughes S.G., Graham M.J., Crooke R.M. Antisense oligonucleotides targeting apolipoprotein(a) in people with raised lipoprotein(a): two randomised, double-blind, placebo-controlled, dose-ranging trials. Lancet. 2016;388:2239–2253. doi: 10.1016/S0140-6736(16)31009-1. [DOI] [PubMed] [Google Scholar]
- 68.Fitzgerald K., White S., Borodovsky A., Bettencourt B.R., Strahs A., Clausen V., Wijngaard P., Horton J.D., Taubel J., Brooks A. A Highly Durable RNAi Therapeutic Inhibitor of PCSK9. N. Engl. J. Med. 2017;376:41–51. doi: 10.1056/NEJMoa1609243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Gaudet D., Alexander V.J., Baker B.F., Brisson D., Tremblay K., Singleton W., Geary R.S., Hughes S.G., Viney N.J., Graham M.J. Antisense Inhibition of Apolipoprotein C-III in Patients with Hypertriglyceridemia. N. Engl. J. Med. 2015;373:438–447. doi: 10.1056/NEJMoa1400283. [DOI] [PubMed] [Google Scholar]
- 70.Thomas G.S., Cromwell W.C., Ali S., Chin W., Flaim J.D., Davidson M. Mipomersen, an apolipoprotein B synthesis inhibitor, reduces atherogenic lipoproteins in patients with severe hypercholesterolemia at high cardiovascular risk: a randomized, double-blind, placebo-controlled trial. J. Am. Coll. Cardiol. 2013;62:2178–2184. doi: 10.1016/j.jacc.2013.07.081. [DOI] [PubMed] [Google Scholar]
- 71.Wacker B.K., Dronadula N., Zhang J., Dichek D.A. Local Vascular Gene Therapy With Apolipoprotein A-I to Promote Regression of Atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2017;37:316–327. doi: 10.1161/ATVBAHA.116.308258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Ylä-Herttuala S., Palinski W., Rosenfeld M.E., Parthasarathy S., Carew T.E., Butler S., Witztum J.L., Steinberg D. Evidence for the presence of oxidatively modified low density lipoprotein in atherosclerotic lesions of rabbit and man. J. Clin. Invest. 1989;84:1086–1095. doi: 10.1172/JCI114271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Mäkinen P.I., Lappalainen J.P., Heinonen S.E., Leppänen P., Lähteenvuo M.T., Aarnio J.V., Heikkilä J., Turunen M.P., Ylä-Herttuala S. Silencing of either SR-A or CD36 reduces atherosclerosis in hyperlipidaemic mice and reveals reciprocal upregulation of these receptors. Cardiovasc. Res. 2010;88:530–538. doi: 10.1093/cvr/cvq235. [DOI] [PubMed] [Google Scholar]
- 74.Tsimikas S., Miyanohara A., Hartvigsen K., Merki E., Shaw P.X., Chou M.Y., Pattison J., Torzewski M., Sollors J., Friedmann T. Human oxidation-specific antibodies reduce foam cell formation and atherosclerosis progression. J. Am. Coll. Cardiol. 2011;58:1715–1727. doi: 10.1016/j.jacc.2011.07.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.DeYoung M.B., Dichek D.A. Gene therapy for restenosis: are we ready? Circ. Res. 1998;82:306–313. doi: 10.1161/01.res.82.3.306. [DOI] [PubMed] [Google Scholar]
- 76.Kutryk M.J., Foley D.P., van den Brand M., Hamburger J.N., van der Giessen W.J., deFeyter P.J., Bruining N., Sabate M., Serruys P.W., ITALICS Trial Local intracoronary administration of antisense oligonucleotide against c-myc for the prevention of in-stent restenosis: results of the randomized investigation by the Thoraxcenter of antisense DNA using local delivery and IVUS after coronary stenting (ITALICS) trial. J. Am. Coll. Cardiol. 2002;39:281–287. doi: 10.1016/s0735-1097(01)01741-7. [DOI] [PubMed] [Google Scholar]
- 77.Turunen P., Puhakka H.L., Heikura T., Romppanen E., Inkala M., Leppänen O., Ylä-Herttuala S. Extracellular superoxide dismutase with vaccinia virus anti-inflammatory protein 35K or tissue inhibitor of metalloproteinase-1: Combination gene therapy in the treatment of vein graft stenosis in rabbits. Hum. Gene Ther. 2006;17:405–414. doi: 10.1089/hum.2006.17.405. [DOI] [PubMed] [Google Scholar]
- 78.George S.J., Wan S., Hu J., MacDonald R., Johnson J.L., Baker A.H. Sustained reduction of vein graft neointima formation by ex vivo TIMP-3 gene therapy. Circulation. 2011;124(11, Suppl):S135–S142. doi: 10.1161/CIRCULATIONAHA.110.012732. [DOI] [PubMed] [Google Scholar]
- 79.Conte M.S., Bandyk D.F., Clowes A.W., Moneta G.L., Seely L., Lorenz T.J., Namini H., Hamdan A.D., Roddy S.P., Belkin M., PREVENT III Investigators Results of PREVENT III: a multicenter, randomized trial of edifoligide for the prevention of vein graft failure in lower extremity bypass surgery. J. Vasc. Surg. 2006;43:742–751. doi: 10.1016/j.jvs.2005.12.058. discussion 751. [DOI] [PubMed] [Google Scholar]
- 80.Alexander J.H., Hafley G., Harrington R.A., Peterson E.D., Ferguson T.B., Jr., Lorenz T.J., Goyal A., Gibson M., Mack M.J., Gennevois D., PREVENT IV Investigators Efficacy and safety of edifoligide, an E2F transcription factor decoy, for prevention of vein graft failure following coronary artery bypass graft surgery: PREVENT IV: a randomized controlled trial. JAMA. 2005;294:2446–2454. doi: 10.1001/jama.294.19.2446. [DOI] [PubMed] [Google Scholar]
- 81.Ark Therapeutics. (2009) Ark Therapeutics completes potency test qualification enabling Trinam Phase III trial to begin. Quick Facts RTTNews. 8.1.2009.
- 82.Rissanen T.T., Ylä-Herttuala S. Current status of cardiovascular gene therapy. Mol. Ther. 2007;15:1233–1247. doi: 10.1038/sj.mt.6300175. [DOI] [PubMed] [Google Scholar]
- 83.Karvinen H., Pasanen E., Rissanen T.T., Korpisalo P., Vähäkangas E., Jazwa A., Giacca M., Ylä-Herttuala S. Long-term VEGF-A expression promotes aberrant angiogenesis and fibrosis in skeletal muscle. Gene Ther. 2011;18:1166–1172. doi: 10.1038/gt.2011.66. [DOI] [PubMed] [Google Scholar]
- 84.Korpisalo P., Hytönen J.P., Laitinen J.T., Laidinen S., Parviainen H., Karvinen H., Siponen J., Marjomäki V., Vajanto I., Rissanen T.T., Ylä-Herttuala S. Capillary enlargement, not sprouting angiogenesis, determines beneficial therapeutic effects and side effects of angiogenic gene therapy. Eur. Heart J. 2011;32:1664–1672. doi: 10.1093/eurheartj/ehq433. [DOI] [PubMed] [Google Scholar]

