1
|
Mani AM, Lamin V, Peach RC, Friesen EH, Wong T, Singh MV, Dokun AO. miRNA-6236 Regulation of Postischemic Skeletal Muscle Angiogenesis. J Am Heart Assoc 2024:e035923. [PMID: 39604034 DOI: 10.1161/jaha.124.035923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 10/22/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND Peripheral arterial disease affects >200 million people worldwide and is characterized by impaired blood flow to the lower extremities. There are no effective medical treatments available. Using the mouse hind-limb ischemia model and miRNA sequencing, we identified a novel miRNA, miR-6236, whose expression significantly elevated in ischemic mouse limbs compared with nonischemic limbs. The role of miR-6236 in general or in postischemic angiogenesis is not known. Here we describe its role using in vivo and in vitro models of peripheral arterial disease. METHODS AND RESULTS In primary mouse and human endothelial cells, we studied the effect of simulated ischemia on miR-6236 expression and assessed its role in cell viability, apoptosis, migration, and tube formation during ischemia. Furthermore, we developed miR-6236 null mice and tested its role in postischemic perfusion recovery using the hind-limb ischemia model. Lastly, using bioinformatics and gene expression analysis, we identified putative angiogenic miR-6236 targets. In vitro simulated ischemia-enhanced miR-6236 expression in mouse and human endothelial cells, whereas its inhibition improved viability, migration, tube formation, and reduced apoptosis. In vivo ischemic mouse skeletal muscle tissue showed higher miR-6236 expression compared with nonischemic muscles. Loss of miR-6236 improved impaired postischemic perfusion recovery and poor angiogenesis associated with streptozotocin-induced diabetes in mice. Six of the 8 miR-6236 predicted angiogenic target mRNAs showed expression consistent with regulation by miR-6236 in ischemic skeletal muscle. CONCLUSIONS Our results show for the first time that miR-6236 plays a key role in regulating postischemic perfusion recovery and angiogenesis.
Collapse
Affiliation(s)
- Arul M Mani
- Division of Endocrinology and Metabolism, Carver College of Medicine University of Iowa Iowa City IA USA
| | - Victor Lamin
- Division of Endocrinology and Metabolism, Carver College of Medicine University of Iowa Iowa City IA USA
| | - Ronan C Peach
- Division of Endocrinology and Metabolism, Carver College of Medicine University of Iowa Iowa City IA USA
| | - Eli H Friesen
- Division of Endocrinology and Metabolism, Carver College of Medicine University of Iowa Iowa City IA USA
| | - Thomas Wong
- Division of Endocrinology and Metabolism, Carver College of Medicine University of Iowa Iowa City IA USA
| | - Madhu V Singh
- Division of Endocrinology and Metabolism, Carver College of Medicine University of Iowa Iowa City IA USA
| | - Ayotunde O Dokun
- Division of Endocrinology and Metabolism, Carver College of Medicine University of Iowa Iowa City IA USA
- Fraternal Order of Eagles Diabetes Research Centre, Carver College of Medicine University of Iowa Iowa City IA USA
| |
Collapse
|
2
|
Kupczyńska D, Lubieniecki P, Antkiewicz M, Barć J, Frączkowska-Sioma K, Dawiskiba T, Dorobisz T, Małodobra-Mazur M, Baczyńska D, Pańczak K, Witkiewicz W, Janczak D, Skóra JP, Barć P. Complementary Gene Therapy after Revascularization with the Saphenous Vein in Diabetic Foot Syndrome. Genes (Basel) 2023; 14:1968. [PMID: 37895317 PMCID: PMC10606318 DOI: 10.3390/genes14101968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
Diabetic foot syndrome (DFS) is one of the most serious macroangiopathic complications of diabetes. The primary treatment option is revascularization, but complementary therapies are still being sought. The study group consisted of 18 patients diagnosed with ischemic ulcerative and necrotic lesions in DFS. Patients underwent revascularization procedures and, due to unsatisfactory healing of the lesions, were randomly allocated to two groups: a group in which bicistronic VEGF165/HGF plasmid was administered and a control group in which saline placebo was administered. Before gene therapy administration and after 7, 30, 90, and 180 days, color duplex ultrasonography (CDU) was performed, the ankle-brachial index (ABI) and transcutaneous oxygen pressure (TcPO2) were measured, and DFS changes were described and documented photographically. In the gene therapy group, four out of eight patients (50%) healed their DFS lesions before 12 weeks. During this time, the ABI increased by an average of 0.25 and TcPO2 by 30.4 mmHg. In the control group, healing of the lesions by week 12 occurred in six out of nine patients (66.67%), and the ABI increased by an average of 0.14 and TcPO2 by 27.1 mmHg. One major amputation occurred in each group. Gene therapy may be an attractive option for complementary treatment in DFS.
Collapse
Affiliation(s)
- Diana Kupczyńska
- Clinical Department of Vascular, General and Transplantation Surgery, Wroclaw Medical University, Borowska Street 213, 50-556 Wroclaw, Poland; (D.K.); (M.A.); (K.F.-S.); (T.D.); (T.D.); (D.J.); (J.P.S.); (P.B.)
| | - Paweł Lubieniecki
- Clinical Department of Diabetology and Internal Disease, Wroclaw Medical University, Borowska Street 213, 50-556 Wroclaw, Poland
| | - Maciej Antkiewicz
- Clinical Department of Vascular, General and Transplantation Surgery, Wroclaw Medical University, Borowska Street 213, 50-556 Wroclaw, Poland; (D.K.); (M.A.); (K.F.-S.); (T.D.); (T.D.); (D.J.); (J.P.S.); (P.B.)
| | - Jan Barć
- Faculty of Medicine, Medical University of Lublin, Aleje Racławickie 1, 20-059 Lublin, Poland;
| | - Katarzyna Frączkowska-Sioma
- Clinical Department of Vascular, General and Transplantation Surgery, Wroclaw Medical University, Borowska Street 213, 50-556 Wroclaw, Poland; (D.K.); (M.A.); (K.F.-S.); (T.D.); (T.D.); (D.J.); (J.P.S.); (P.B.)
| | - Tomasz Dawiskiba
- Clinical Department of Vascular, General and Transplantation Surgery, Wroclaw Medical University, Borowska Street 213, 50-556 Wroclaw, Poland; (D.K.); (M.A.); (K.F.-S.); (T.D.); (T.D.); (D.J.); (J.P.S.); (P.B.)
| | - Tadeusz Dorobisz
- Clinical Department of Vascular, General and Transplantation Surgery, Wroclaw Medical University, Borowska Street 213, 50-556 Wroclaw, Poland; (D.K.); (M.A.); (K.F.-S.); (T.D.); (T.D.); (D.J.); (J.P.S.); (P.B.)
| | - Małgorzata Małodobra-Mazur
- Department of Forensic Medicine, Division of Molecular Techniques, Wroclaw Medical University, Borowska Street 213, 50-556 Wroclaw, Poland;
| | - Dagmara Baczyńska
- Department of Molecular and Cellular Biology, Wroclaw Medical University, Borowska 211A, 50-556 Wrocław, Poland;
| | - Konrad Pańczak
- Lecran Wound Care Center, Trawowa 63a, 54-614 Wrocław, Poland;
| | - Wojciech Witkiewicz
- Research and Development Center, Regional Specialized Hospital in Wroclaw, Kamienskiego 73a, 51-124 Wroclaw, Poland;
| | - Dariusz Janczak
- Clinical Department of Vascular, General and Transplantation Surgery, Wroclaw Medical University, Borowska Street 213, 50-556 Wroclaw, Poland; (D.K.); (M.A.); (K.F.-S.); (T.D.); (T.D.); (D.J.); (J.P.S.); (P.B.)
| | - Jan Paweł Skóra
- Clinical Department of Vascular, General and Transplantation Surgery, Wroclaw Medical University, Borowska Street 213, 50-556 Wroclaw, Poland; (D.K.); (M.A.); (K.F.-S.); (T.D.); (T.D.); (D.J.); (J.P.S.); (P.B.)
| | - Piotr Barć
- Clinical Department of Vascular, General and Transplantation Surgery, Wroclaw Medical University, Borowska Street 213, 50-556 Wroclaw, Poland; (D.K.); (M.A.); (K.F.-S.); (T.D.); (T.D.); (D.J.); (J.P.S.); (P.B.)
| |
Collapse
|
3
|
Weeraman D, Jones DA, Hussain M, Beirne AM, Hadyanto S, Rathod KS, Whiteford JR, Reid AE, Bourantas CV, Ylä-Herttuala S, Baumbach A, Gersh BJ, Henry TD, Mathur A. Proangiogenic Growth Factor Therapy for the Treatment of Refractory Angina: A Meta-analysis. JOURNAL OF THE SOCIETY FOR CARDIOVASCULAR ANGIOGRAPHY & INTERVENTIONS 2023; 2:100527. [PMID: 39132540 PMCID: PMC11307391 DOI: 10.1016/j.jscai.2022.100527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/10/2022] [Accepted: 10/14/2022] [Indexed: 08/13/2024]
Abstract
Background Refractory angina (RFA; limiting angina despite optimal medical therapy) is a growing, global problem, with limited treatment options. Therefore, we conducted a systematic review of randomized controlled trials (RCTs) to evaluate the effect of proangiogenic growth factor therapy (in the form of vascular growth factors delivered either as recombinant proteins or gene therapy) in patients with RFA ineligible for revascularization. Methods We performed a meta-analysis (PROSPERO: CRD42018107283) of RCTs as per the Preferred Reporting Items for Systematic Reviews and Meta-Analyses methodology. A comprehensive search of the PubMed, CENTRAL, Embase, Cochrane, ClinicalTrials.gov and Google Scholar databases, as well as scientific session abstracts, were performed. The pooled outcomes included major adverse cardiac events (MACE), mortality, myocardial perfusion, and indices of angina severity (Canadian Cardiovascular Society angina class [CCS] and exercise tolerance). A prespecified subgroup analysis was performed for delivery method, vector, and protein type. The standardized mean difference (SMD) or odds ratio (OR) was calculated to assess relevant outcomes. We assessed heterogeneity using the χ2 and I2 tests. Results We included 16 RCTs involving 1607 patients (1052 received proangiogenic growth factor therapy and 555 received a placebo or optimal medical therapy). Our analysis showed a significant decreased risk of MACE (OR, 0.72; 95% confidence interval [CI], 0.55-0.93) and significantly improved CCS class (SMD, -0.55; 95% CI, -1.10 to 0.00), but not mortality (OR, 0.66; 95% CI, 0.28-1.54) or exercise tolerance (SMD, 0.47; 95% CI, -0.14 to 1.09), in treated patients compared to those in the control group. Conclusions Proangiogenic growth factor therapy is a promising treatment option for RFA, with beneficial effects seen on MACE and CCS class. The results of ongoing trials are needed before it can be considered for clinical practice.
Collapse
Affiliation(s)
- Deshan Weeraman
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
- Barts National Institute for Health and Care Research Biomedical Research Centre, Barts Heart Centre & Queen Mary University of London, London, United Kingdom
- Barts Heart Centre, Barts Health National Health Service Trust, London, United Kingdom
| | - Daniel A. Jones
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
- Barts National Institute for Health and Care Research Biomedical Research Centre, Barts Heart Centre & Queen Mary University of London, London, United Kingdom
- Barts Heart Centre, Barts Health National Health Service Trust, London, United Kingdom
| | - Mohsin Hussain
- Barts National Institute for Health and Care Research Biomedical Research Centre, Barts Heart Centre & Queen Mary University of London, London, United Kingdom
- Barts Heart Centre, Barts Health National Health Service Trust, London, United Kingdom
| | - Anne-Marie Beirne
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
- Barts Heart Centre, Barts Health National Health Service Trust, London, United Kingdom
| | - Steven Hadyanto
- Barts National Institute for Health and Care Research Biomedical Research Centre, Barts Heart Centre & Queen Mary University of London, London, United Kingdom
| | - Krishnaraj S. Rathod
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
- Barts National Institute for Health and Care Research Biomedical Research Centre, Barts Heart Centre & Queen Mary University of London, London, United Kingdom
- Barts Heart Centre, Barts Health National Health Service Trust, London, United Kingdom
| | - James R. Whiteford
- Centre for Microvascular Research, William Harvey Research Institute, Barts & The London Medical School, Queen Mary University of London, London, United Kingdom
| | - Alice E. Reid
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Christos V. Bourantas
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
- Barts National Institute for Health and Care Research Biomedical Research Centre, Barts Heart Centre & Queen Mary University of London, London, United Kingdom
- Barts Heart Centre, Barts Health National Health Service Trust, London, United Kingdom
| | | | - Andreas Baumbach
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
- Barts National Institute for Health and Care Research Biomedical Research Centre, Barts Heart Centre & Queen Mary University of London, London, United Kingdom
- Barts Heart Centre, Barts Health National Health Service Trust, London, United Kingdom
| | - Bernard J. Gersh
- Department of Cardiovascular Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Timothy D. Henry
- The Carl and Edyth Lindner Center for Research and Education at The Christ Hospital, Cincinnati, Ohio
| | - Anthony Mathur
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
- Barts National Institute for Health and Care Research Biomedical Research Centre, Barts Heart Centre & Queen Mary University of London, London, United Kingdom
- Barts Heart Centre, Barts Health National Health Service Trust, London, United Kingdom
| |
Collapse
|
4
|
Barć P, Antkiewicz M, Frączkowska-Sioma K, Kupczyńska D, Lubieniecki P, Witkiewicz W, Małodobra-Mazur M, Baczyńska D, Janczak D, Skóra JP. Two-Stage Gene Therapy (VEGF, HGF and ANG1 Plasmids) as Adjunctive Therapy in the Treatment of Critical Lower Limb Ischemia in Diabetic Foot Syndrome. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:12818. [PMID: 36232122 PMCID: PMC9564889 DOI: 10.3390/ijerph191912818] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/03/2022] [Accepted: 10/04/2022] [Indexed: 06/16/2023]
Abstract
One of the most serious problems in people with diabetes is diabetic foot syndrome. Due to the peripheral location of atherosclerotic lesions in the arterial system of the lower extremities, endovascular treatment plays a dominant role. However, carrying out these procedures is not always possible and does not always bring the expected results. Gene therapy, which stimulates angiogenesis, improves not only the inflow from the proximal limb but also the blood redistribution in individual angiosomes. Due to the encouraging results of sequential treatment consisting of intramuscular injections of VEGF/HGF bicistronic plasmids followed by a month of ANG1 plasmids, we decided to use the described method for the treatment of critical ischemia of the lower limbs in the course of diabetes and, more specifically, in diabetic foot syndrome. Twenty-four patients meeting the inclusion criteria were enrolled in the study. They were randomly divided into two equal groups. The first group of patients was subjected to gene therapy, where the patients received intramuscular injections of pIRES/VEGF165/HGF plasmids and 1 month of ANG-1 plasmids. The remaining patients constituted the control group. Gene therapy was well tolerated by most patients. The wounds healed significantly better in Group 1. The minimal value of ABI increased significantly in Group 1 from 0.44 ± 0.14 (± standard deviation) to 0.47 ± 0.12 (with p = 0.028) at the end of the study. There were no significant differences in the control group. In the gene treatment group, PtcO2 increased significantly (from 28.71 ± 10.89 mmHg to 33.9 ± 6.33 mmHg with p = 0.001), while in Group 2, no statistically significant changes were found. The observed resting pain decreased significantly in both groups (Group 1 decreased from 6.80 ± 1.48 to 2.10 ± 1.10; p < 0.001; the control group decreased from 7.44 ± 1.42 to 3.78 ± 1.64 with p < 0.001). In our study, we evaluated the effectiveness of gene therapy with the growth factors described above in patients with CLI in the course of complicated DM. The therapy was shown to be effective with minimal side effects. No serious complications were observed.
Collapse
Affiliation(s)
- Piotr Barć
- Department and Clinic of Vascular, General and Transplantation Surgery, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland
| | - Maciej Antkiewicz
- Department and Clinic of Vascular, General and Transplantation Surgery, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland
| | - Katarzyna Frączkowska-Sioma
- Department and Clinic of Vascular, General and Transplantation Surgery, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland
| | - Diana Kupczyńska
- Department and Clinic of Vascular, General and Transplantation Surgery, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland
| | - Paweł Lubieniecki
- Department of Angiology, Hypertension and Diabetology, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland
| | - Wojciech Witkiewicz
- Research and Development Center, Regional Specialized Hospital in Wroclaw, Kamienskiego 73a, 51-124 Wroclaw, Poland
| | - Małgorzata Małodobra-Mazur
- Department of Forensic Medicine, Division of Molecular Techniques, Wroclaw Medical University, M. Curie-Sklodowskiej 52, 50-369 Wroclaw, Poland
| | - Dagmara Baczyńska
- Department of Molecular and Cellular Biology, Wroclaw Medical University, Borowska 211A, 50-556 Wrocław, Poland
| | - Dariusz Janczak
- Department and Clinic of Vascular, General and Transplantation Surgery, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland
| | - Jan Paweł Skóra
- Department and Clinic of Vascular, General and Transplantation Surgery, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland
| |
Collapse
|
5
|
Leikas AJ, Hassinen I, Kivelä A, Hedman A, Mussalo H, Ylä-Herttuala S, Hartikainen JEK. Intramyocardial adenoviral vascular endothelial growth factor-D ∆N∆C gene therapy does not induce ventricular arrhythmias. J Gene Med 2022; 24:e3437. [PMID: 35750637 DOI: 10.1002/jgm.3437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 06/13/2022] [Accepted: 06/19/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Phase I KAT301 trial investigated the use of intramyocardial adenoviral vascular endothelial growth factor -DΔNΔC (AdVEGF-D) gene therapy (GT) to alleviate symptoms in refractory angina (RA) patients. In KAT301, 30 patients with RA were randomized to AdVEGF-D or control group in 4:1 ratio. The treatment was found feasible, increased myocardial perfusion, and reduced angina symptoms at 1-year follow-up. However, there is some evidence suggesting that intramyocardial delivery route and overexpression of VEGFs might induce ventricular arrhythmias. Thus, we investigated whether intramyocardial AdVEGF-D GT increases the risk of ventricular arrhythmias in patients treated for RA. METHODS We analyzed non-invasive risk predictors of ventricular arrhythmias from 12-lead electrocardiography (ECG) as well as heart rate variability (HRV) and the incidence of arrhythmias from 24 h ambulatory ECG at baseline and 3 and 12 months after the GT. In addition, we analyzed the incidence of new-onset arrhythmias and pacemaker implantations during 8.2-year (range 6.3 - 10.4 years) follow-up. RESULTS We found no significant increase in arrhythmias, including supraventricular and ventricular ectopic beats, atrial fibrillation, non-sustained ventricular tachycardias, and life-threatening tachycardias, nor changes in the non-invasive risk predictors of ventricular arrhythmias in the AdVEGF-D treated patients. Instead, we found a significant improvement in the very low and high-frequency bands of HRV suggestive of improved cardiac autonomic regulation after GT. CONCLUSIONS In conclusion, our results suggest that AdVEGF-D GT does not predispose to arrhythmias and might improve HRV metrics.
Collapse
Affiliation(s)
- Aleksi J Leikas
- Heart Center, Kuopio University Hospital, Kuopio, Finland.,A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.,Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland
| | - Iiro Hassinen
- Heart Center, Kuopio University Hospital, Kuopio, Finland.,Mikkeli Central Hospital, Mikkeli, Finland
| | - Antti Kivelä
- Heart Center, Kuopio University Hospital, Kuopio, Finland
| | - Antti Hedman
- Heart Center, Kuopio University Hospital, Kuopio, Finland
| | - Hanna Mussalo
- Center of Diagnostic Imaging, Kuopio University Hospital, Kuopio, Finland
| | - Seppo Ylä-Herttuala
- Heart Center, Kuopio University Hospital, Kuopio, Finland.,A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.,Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland
| | - Juha E K Hartikainen
- Heart Center, Kuopio University Hospital, Kuopio, Finland.,Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
6
|
Lamin V, Verry J, Eigner-Bybee I, Fuqua JD, Wong T, Lira VA, Dokun AO. Modulation of miR-29a and ADAM12 Reduces Post-Ischemic Skeletal Muscle Injury and Improves Perfusion Recovery and Skeletal Muscle Function in a Mouse Model of Type 2 Diabetes and Peripheral Artery Disease. Int J Mol Sci 2021; 23:429. [PMID: 35008854 PMCID: PMC8745107 DOI: 10.3390/ijms23010429] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/23/2021] [Accepted: 12/25/2021] [Indexed: 12/15/2022] Open
Abstract
Both Type 1 diabetes mellitus (DM1) and type 2 diabetes mellitus (DM2) are associated with an increased risk of limb amputation in peripheral arterial disease (PAD). How diabetes contributes to poor PAD outcomes is poorly understood but may occur through different mechanisms in DM1 and DM2. Previously, we identified a disintegrin and metalloproteinase gene 12 (ADAM12) as a key genetic modifier of post-ischemic perfusion recovery. In an experimental PAD, we showed that ADAM12 is regulated by miR-29a and this regulation is impaired in ischemic endothelial cells in DM1, contributing to poor perfusion recovery. Here we investigated whether miR-29a regulation of ADAM12 is altered in experimental PAD in the setting of DM2. We also explored whether modulation of miR-29a and ADAM12 expression can improve perfusion recovery and limb function in mice with DM2. Our result showed that in the ischemic limb of mice with DM2, miR-29a expression is poorly downregulated and ADAM12 upregulation is impaired. Inhibition of miR-29a and overexpression of ADAM12 improved perfusion recovery, reduced skeletal muscle injury, improved muscle function, and increased cleaved Tie 2 and AKT phosphorylation. Thus, inhibition of miR-29a and or augmentation of ADAM12 improves experimental PAD outcomes in DM2 likely through modulation of Tie 2 and AKT signalling.
Collapse
Affiliation(s)
- Victor Lamin
- Division of Endocrinology and Metabolism, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; (V.L.); (J.V.); (I.E.-B.); (T.W.)
| | - Joseph Verry
- Division of Endocrinology and Metabolism, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; (V.L.); (J.V.); (I.E.-B.); (T.W.)
| | - Isaac Eigner-Bybee
- Division of Endocrinology and Metabolism, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; (V.L.); (J.V.); (I.E.-B.); (T.W.)
| | - Jordan D. Fuqua
- Department of Health and Human Physiology, College of Liberal Arts and Sciences, University of Iowa, Iowa City, IA 52242, USA; (J.D.F.); (V.A.L.)
| | - Thomas Wong
- Division of Endocrinology and Metabolism, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; (V.L.); (J.V.); (I.E.-B.); (T.W.)
| | - Vitor A. Lira
- Department of Health and Human Physiology, College of Liberal Arts and Sciences, University of Iowa, Iowa City, IA 52242, USA; (J.D.F.); (V.A.L.)
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Ayotunde O. Dokun
- Division of Endocrinology and Metabolism, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; (V.L.); (J.V.); (I.E.-B.); (T.W.)
- Department of Health and Human Physiology, College of Liberal Arts and Sciences, University of Iowa, Iowa City, IA 52242, USA; (J.D.F.); (V.A.L.)
| |
Collapse
|
7
|
A Thermoresponsive Chitosan/β-Glycerophosphate Hydrogel for Minimally Invasive Treatment of Critical Limb Ischaemia. Polymers (Basel) 2021; 13:polym13203568. [PMID: 34685327 PMCID: PMC8539345 DOI: 10.3390/polym13203568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/12/2021] [Accepted: 10/14/2021] [Indexed: 11/17/2022] Open
Abstract
A reduction in blood supply to any limb causes ischaemia, pain and morbidity. Critical limb ischaemia is the most serious presentation of peripheral vascular disease. One in five patients with critical limb ischaemia will die within six months of diagnosis and one in three will require amputation in this time. Improving blood flow to the limb, via the administration of angiogenic agents, could relieve pain and avoid amputation. Herein, chitosan is combined with β-glycerophosphate to form a thermoresponsive formulation (chitosan/β-GP) that will flow through a syringe and needle at room temperature but will form a gel at body temperature. The chitosan/β-GP hydrogel, with or without the angiogenic molecule desferrioxamine (DFO), was injected into the mouse hind limb, following vessel ligation, to test the ability of the formulations to induce angiogenesis. The effects of the formulations were measured using laser Doppler imaging to determine limb perfusion and CD31 staining to quantify the number of blood vessels. Twenty-eight days following induction of ischaemia, the chitosan/β-GP and chitosan/β-GP + 100 µM DFO formulations had significantly (p < 0.001 and p < 0.05, respectively) improved blood flow in the ischaemic limb compared with an untreated control. Chitosan/β-GP increased vessel number by 1.7-fold in the thigh of the ischaemic limb compared with an untreated control, while chitosan/β-GP + 100 µM DFO increased vessel number 1.8-fold. Chitosan/β-GP represents a potential minimally invasive treatment for critical limb ischaemia.
Collapse
|
8
|
Leikas AJ, Hassinen I, Hedman A, Kivelä A, Ylä-Herttuala S, Hartikainen JEK. Long-term safety and efficacy of intramyocardial adenovirus-mediated VEGF-D ΔNΔC gene therapy eight-year follow-up of phase I KAT301 study. Gene Ther 2021; 29:289-293. [PMID: 34593990 PMCID: PMC9159942 DOI: 10.1038/s41434-021-00295-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 08/27/2021] [Accepted: 09/15/2021] [Indexed: 11/09/2022]
Abstract
In phase I KAT301 trial, intramyocardial adenovirus-mediated vascular endothelial growth factor -DΔNΔC (AdVEGF-D) gene therapy (GT) resulted in a significant improvement in myocardial perfusion reserve and relieved symptoms in refractory angina patients at 1-year follow-up without major safety concerns. We investigated the long-term safety and efficacy of AdVEGF-D GT. 30 patients (24 in VEGF-D group and 6 blinded, randomized controls) were followed for 8.2 years (range 6.3–10.4 years). Patients were interviewed for the current severity of symptoms (Canadian Cardiovascular Society class, CCS) and perceived benefit from GT. Medical records were reviewed to assess the incidence of major cardiovascular adverse event (MACE) and other predefined safety endpoints. MACE occurred in 15 patients in VEGF-D group and in five patients in control group (21.5 vs. 24.9 per 100 patient-years; hazard ratio 0.97; 95% confidence interval 0.36–2.63; P = 0.95). Mortality and new-onset comorbidity were similar between the groups. Angina symptoms (CCS) were less severe compared to baseline in VEGF-D group (1.9 vs. 2.9; P = 0.006) but not in control group (2.2 vs. 2.6; P = 0.414). Our study indicates that intramyocardial AdVEGF-D GT is safe in the long-term. In addition, the relief of symptoms remained significant during the follow-up.
Collapse
Affiliation(s)
- Aleksi J Leikas
- Heart Center, Kuopio University Hospital, Kuopio, Finland.,A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.,Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland
| | - Iiro Hassinen
- Heart Center, Kuopio University Hospital, Kuopio, Finland
| | - Antti Hedman
- Heart Center, Kuopio University Hospital, Kuopio, Finland
| | - Antti Kivelä
- Heart Center, Kuopio University Hospital, Kuopio, Finland
| | - Seppo Ylä-Herttuala
- Heart Center, Kuopio University Hospital, Kuopio, Finland.,A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.,Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland
| | - Juha E K Hartikainen
- Heart Center, Kuopio University Hospital, Kuopio, Finland. .,Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland.
| |
Collapse
|
9
|
Collins LT, Curiel DT. Synthetic Biology Approaches for Engineering Next-Generation Adenoviral Gene Therapies. ACS NANO 2021; 15:13970-13979. [PMID: 34415739 DOI: 10.1021/acsnano.1c04556] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Synthetic biology centers on the design and modular assembly of biological parts so as to construct artificial biological systems. Over the past decade, synthetic biology has blossomed into a highly productive field, yielding advances in diverse areas such as neuroscience, cell-based therapies, and chemical manufacturing. Similarly, the field of gene therapy has made enormous strides both in proof-of-concept studies and in the clinical setting. One viral vector of increasing interest for gene therapy is the adenovirus (Ad). A major part of the Ad's increasing momentum comes from synthetic biology approaches to Ad engineering. Convergence of gene therapy and synthetic biology has enhanced Ad vectors by mitigating Ad toxicity in vivo, providing precise Ad tropisms, and incorporating genetic circuits to make smart therapies which adapt to environmental stimuli. Synthetic biology engineering of Ad vectors may lead to superior gene delivery and editing platforms which could find applications in a wide range of therapeutic contexts.
Collapse
Affiliation(s)
- Logan Thrasher Collins
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri 63110, United States
| | - David T Curiel
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri 63110, United States
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri 63110, United States
| |
Collapse
|
10
|
Adini A, Adini I, Grad E, Tal Y, Danenberg HD, Kang PM, Matthews BD, D’Amato RJ. The Prominin-1-Derived Peptide Improves Cardiac Function Following Ischemia. Int J Mol Sci 2021; 22:5169. [PMID: 34068392 PMCID: PMC8153573 DOI: 10.3390/ijms22105169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/26/2021] [Accepted: 05/11/2021] [Indexed: 12/11/2022] Open
Abstract
Myocardial infarction (MI) remains the leading cause of death in the western world. Despite advancements in interventional revascularization technologies, many patients are not candidates for them due to comorbidities or lack of local resources. Non-invasive approaches to accelerate revascularization within ischemic tissues through angiogenesis by providing Vascular Endothelial Growth Factor (VEGF) in protein or gene form has been effective in animal models but not in humans likely due to its short half-life and systemic toxicity. Here, we tested the hypothesis that PR1P, a small VEGF binding peptide that we developed, which stabilizes and upregulates endogenous VEGF, could be used to improve outcome from MI in rodents. To test this hypothesis, we induced MI in mice and rats via left coronary artery ligation and then treated animals with every other day intraperitoneal PR1P or scrambled peptide for 14 days. Hemodynamic monitoring and echocardiography in mice and echocardiography in rats at 14 days showed PR1P significantly improved multiple functional markers of heart function, including stroke volume and cardiac output. Furthermore, molecular biology and histological analyses of tissue samples showed that systemic PR1P targeted, stabilized and upregulated endogenous VEGF within ischemic myocardium. We conclude that PR1P is a potential non-invasive candidate therapeutic for MI.
Collapse
Affiliation(s)
- Avner Adini
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (B.D.M.); (R.J.D.)
- Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Irit Adini
- Department of Surgery, Harvard Medical School, The Center for Engineering in Medicine, Mass General Hospital, Shriners Hospitals for Children Boston, Boston, MA 02114, USA;
| | - Etty Grad
- Interventional Cardiology, Heart Institute, Hadassah Hebrew University Medical Center, Jerusalem 91200, Israel; (E.G.); (H.D.D.)
| | - Yuval Tal
- Allergy and Clinical Immunology Unit and Department of Medicine, Hadassah University Medical Center, Jerusalem 91200, Israel;
| | - Haim D. Danenberg
- Interventional Cardiology, Heart Institute, Hadassah Hebrew University Medical Center, Jerusalem 91200, Israel; (E.G.); (H.D.D.)
| | - Peter M. Kang
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA;
| | - Benjamin D. Matthews
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (B.D.M.); (R.J.D.)
- Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Robert J. D’Amato
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (B.D.M.); (R.J.D.)
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
11
|
Hayashi SI, Rakugi H, Morishita R. Insight into the Role of Angiopoietins in Ageing-Associated Diseases. Cells 2020; 9:E2636. [PMID: 33302426 PMCID: PMC7762563 DOI: 10.3390/cells9122636] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 12/05/2020] [Accepted: 12/07/2020] [Indexed: 12/17/2022] Open
Abstract
Angiopoietin (Ang) and its receptor, TIE signaling, contribute to the development and maturation of embryonic vasculature as well as vascular remodeling and permeability in adult tissues. Targeting both this signaling pathway and the major pathway with vascular endothelial growth factor (VEGF) is expected to permit clinical applications, especially in antiangiogenic therapies against tumors. Several drugs targeting the Ang-TIE signaling pathway in cancer patients are under clinical development. Similar to how cancer increases with age, unsuitable angiogenesis or endothelial dysfunction is often seen in other ageing-associated diseases (AADs) such as atherosclerosis, Alzheimer's disease, type 2 diabetes, chronic kidney disease and cardiovascular diseases. Thus, the Ang-TIE pathway is a possible molecular target for AAD therapy. In this review, we focus on the potential role of the Ang-TIE signaling pathway in AADs, especially non-cancer-related AADs. We also suggest translational insights and future clinical applications of this pathway in those AADs.
Collapse
Affiliation(s)
- Shin-ichiro Hayashi
- Department of Clinical Gene Therapy, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Hiromi Rakugi
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan;
| | - Ryuichi Morishita
- Department of Clinical Gene Therapy, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| |
Collapse
|
12
|
Shimamura M, Nakagami H, Sanada F, Morishita R. Progress of Gene Therapy in Cardiovascular Disease. Hypertension 2020; 76:1038-1044. [DOI: 10.1161/hypertensionaha.120.14478] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Gene therapy has been extensively studied in peripheral and cardiac ischemia, heart and vein graft failure, and dyslipidemia, but most clinical trials failed to show their efficacies despite good outcomes in preclinical studies. So far, 2 gene therapies for dyslipidemia and one for critical limb ischemia in peripheral artery disease have been approved. In critical limb ischemia, gene therapy using proangiogenic factors has emerged as a novel therapeutic modality for promoting angiogenesis. Initial researches mainly focused on vascular endothelial growth factor, fibroblast growth factor, or hepatocyte growth factor. After the favorable results of basic research, several phase I and II clinical trials of these proangiogenic factors have shown promising results. However, only a phase III clinical trial of the intramuscular injection of hepatocyte growth factor plasmid DNA has shown successful outcomes, and it was recently approved in Japan for treating patients with critical limb ischemia who have ulcers and for whom no alternative therapeutic options are available. DNA vaccine is another promising modality of gene therapy. An antitumor vaccine suppressing angiogenesis through the inhibition of proangiogenic factors and an antihypertensive vaccine inhibiting the renin–angiotensin system are representative DNA vaccines. The advantage of DNA vaccine is its long-term effectiveness with a few vaccinations; however, the benefits and risks, such as adverse T-cell reaction against self-antigen or long-term side effects, of DNA vaccines should be carefully evaluated. In this review, we discuss the recent advances in proangiogenic gene therapy for critical limb ischemia and DNA vaccine for hypertension.
Collapse
Affiliation(s)
- Munehisa Shimamura
- From the Department of Health Development and Medicine, Japan (M.S., H.N.)
| | - Hironori Nakagami
- From the Department of Health Development and Medicine, Japan (M.S., H.N.)
| | - Fumihiro Sanada
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Japan (F.S., R.M.)
| | - Ryuichi Morishita
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Japan (F.S., R.M.)
| |
Collapse
|
13
|
Adini A, Wu H, Dao DT, Ko VH, Yu LJ, Pan A, Puder M, Mitiku SZ, Potla R, Chen H, Rice JM, Matthews BD. PR1P Stabilizes VEGF and Upregulates Its Signaling to Reduce Elastase-induced Murine Emphysema. Am J Respir Cell Mol Biol 2020; 63:452-463. [PMID: 32663413 PMCID: PMC7528927 DOI: 10.1165/rcmb.2019-0434oc] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 06/04/2020] [Indexed: 12/21/2022] Open
Abstract
Emphysema is a progressive and fatal lung disease with no cure that is characterized by thinning, enlargement, and destruction of alveoli, leading to impaired gas exchange. Disease progression is due in part to dysregulation of VEGF (vascular endothelial growth factor) signaling in the lungs and increased lung-cell apoptosis. Here we asked whether PR1P (Prominin-1-derived peptide), a novel short peptide we designed that increases VEGF binding to endothelial cells, could be used to improve outcome in in vitro and in vivo models of emphysema. We used computer simulation and in vitro and in vivo studies to show that PR1P upregulated endogenous VEGF receptor-2 signaling by binding VEGF and preventing its proteolytic degradation. In so doing, PR1P mitigated toxin-induced lung-cell apoptosis, including from cigarette-smoke extract in vitro and from LPS in vivo in mice. Remarkably, inhaled PR1P led to significantly increased VEGF concentrations in murine lungs within 30 minutes that remained greater than twofold above that of control animals 24 hours later. Finally, inhaled PR1P reduced acute lung injury in 4- and 21-day elastase-induced murine emphysema models. Taken together, these results highlight the potential of PR1P as a novel therapeutic agent for the treatment of emphysema or other lung diseases characterized by VEGF signaling dysregulation.
Collapse
Affiliation(s)
- Avner Adini
- Vascular Biology Program
- Department of Pathology
- Department of Surgery, and
- Department of Medicine, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Hao Wu
- Vascular Biology Program
- Department of Pathology
- Department of Surgery, and
| | - Duy T. Dao
- Vascular Biology Program
- Department of Surgery, and
| | | | - Lumeng J. Yu
- Vascular Biology Program
- Department of Surgery, and
| | - Amy Pan
- Vascular Biology Program
- Department of Surgery, and
| | - Mark Puder
- Vascular Biology Program
- Department of Surgery, and
| | - Selome Z. Mitiku
- Vascular Biology Program
- Department of Pathology
- Department of Surgery, and
- Department of Medicine, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Ratnakar Potla
- Vascular Biology Program
- Department of Pathology
- Department of Surgery, and
| | - Hong Chen
- Vascular Biology Program
- Department of Pathology
- Department of Surgery, and
| | - James M. Rice
- Vascular Biology Program
- Department of Pathology
- Department of Surgery, and
| | - Benjamin D. Matthews
- Vascular Biology Program
- Department of Pathology
- Department of Surgery, and
- Department of Medicine, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
14
|
Barć P, Antkiewicz M, Śliwa B, Frączkowska K, Guziński M, Dawiskiba T, Małodobra-Mazur M, Witkiewicz W, Kupczyńska D, Strzelec B, Janczak D, Skóra JP. Double VEGF/HGF Gene Therapy in Critical Limb Ischemia Complicated by Diabetes Mellitus. J Cardiovasc Transl Res 2020; 14:409-415. [PMID: 32875492 PMCID: PMC8219552 DOI: 10.1007/s12265-020-10066-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 08/02/2020] [Indexed: 12/21/2022]
Abstract
Critical leg ischemia (CLI) complicated by diabetes mellitus (DM), which is a very common and dangerous disease, represents the ultimate stage of peripheral arterial disease. Patients are treated with antiplatelet drugs, statins and limb revascularization, but a significant number of patients are not candidate for revascularization. Literature shows that in such cases, gene therapy could be a perfect therapeutic option. The aim of our study was to evaluate efficacy of double vascular endothelial growth factor/hepatocyte growth factor (VEGF/HGF) gene therapy in patients with CLI complicated by DM. We observed that 90 days after administration, serum level of VEGF and ankle-brachial index increased significantly (p < 0.001) and rest pain decreased significantly compared with the control group (p < 0.002). Moreover considerable improvement in vascularization was observed in computed tomography angiography (P = 0.04). Based on the results of this study, we suggest that the therapy with pIRES/VEGF165/HGF bicistronic plasmid administration is a safe and effective method of treatment of patients with both CLI and DM. Graphical abstract.
Collapse
Affiliation(s)
- Piotr Barć
- Department and Clinic of Vascular, General and Transplantation Surgery, Jan Mikulicz-Radecki Medical University Hospital, Wroclaw Medical University, Wroclaw, Poland
| | - Maciej Antkiewicz
- Department and Clinic of Vascular, General and Transplantation Surgery, Jan Mikulicz-Radecki Medical University Hospital, Wroclaw Medical University, Wroclaw, Poland.
| | - Barbara Śliwa
- Department and Clinic of Vascular, General and Transplantation Surgery, Jan Mikulicz-Radecki Medical University Hospital, Wroclaw Medical University, Wroclaw, Poland
| | - Katarzyna Frączkowska
- Department and Clinic of Vascular, General and Transplantation Surgery, Jan Mikulicz-Radecki Medical University Hospital, Wroclaw Medical University, Wroclaw, Poland
| | - Maciej Guziński
- Department of Radiology, Jan Mikulicz-Radecki Medical University Hospital, Wroclaw Medical University, Wroclaw, Poland
| | - Tomasz Dawiskiba
- Department and Clinic of Vascular, General and Transplantation Surgery, Jan Mikulicz-Radecki Medical University Hospital, Wroclaw Medical University, Wroclaw, Poland
| | | | - Wojciech Witkiewicz
- Regional Specialized Hospital in Wroclaw, Research and Development Center, Wroclaw, Poland
| | - Diana Kupczyńska
- Department and Clinic of Vascular, General and Transplantation Surgery, Jan Mikulicz-Radecki Medical University Hospital, Wroclaw Medical University, Wroclaw, Poland
| | - Bartłomiej Strzelec
- Department and Clinic of Vascular, General and Transplantation Surgery, Jan Mikulicz-Radecki Medical University Hospital, Wroclaw Medical University, Wroclaw, Poland
| | - Dariusz Janczak
- Department and Clinic of Vascular, General and Transplantation Surgery, Jan Mikulicz-Radecki Medical University Hospital, Wroclaw Medical University, Wroclaw, Poland
| | - Jan Paweł Skóra
- Department and Clinic of Vascular, General and Transplantation Surgery, Jan Mikulicz-Radecki Medical University Hospital, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
15
|
Abstract
While clinical gene therapy celebrates its first successes, with several products already approved for clinical use and several hundreds in the final stages of the clinical approval pipeline, there is not a single gene therapy approach that has worked for the heart. Here, we review the past experience gained in the several cardiac gene therapy clinical trials that had the goal of inducing therapeutic angiogenesis in the ischemic heart and in the attempts at modulating cardiac function in heart failure. Critical assessment of the results so far achieved indicates that the efficiency of cardiac gene delivery remains a major hurdle preventing success but also that improvements need to be sought in establishing more reliable large animal models, choosing more effective therapeutic genes, better designing clinical trials, and more deeply understanding cardiac biology. We also emphasize a few areas of cardiac gene therapy development that hold great promise for the future. In particular, the transition from gene addition studies using protein-coding cDNAs to the modulation of gene expression using small RNA therapeutics and the improvement of precise gene editing now pave the way to applications such as cardiac regeneration after myocardial infarction and gene correction for inherited cardiomyopathies that were unapproachable until a decade ago.
Collapse
Affiliation(s)
- Antonio Cannatà
- From the King's College London, British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, United Kingdom (A.C., H.A., M.G.).,Department of Medical, Surgical and Health Sciences, University of Trieste, Italy (A.C., G.S., M.G.)
| | - Hashim Ali
- From the King's College London, British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, United Kingdom (A.C., H.A., M.G.).,Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy (H.A., M.G.)
| | - Gianfranco Sinagra
- Department of Medical, Surgical and Health Sciences, University of Trieste, Italy (A.C., G.S., M.G.)
| | - Mauro Giacca
- From the King's College London, British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, United Kingdom (A.C., H.A., M.G.).,Department of Medical, Surgical and Health Sciences, University of Trieste, Italy (A.C., G.S., M.G.).,Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy (H.A., M.G.)
| |
Collapse
|
16
|
TRPC Channels: Dysregulation and Ca 2+ Mishandling in Ischemic Heart Disease. Cells 2020; 9:cells9010173. [PMID: 31936700 PMCID: PMC7017417 DOI: 10.3390/cells9010173] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/03/2020] [Accepted: 01/08/2020] [Indexed: 12/17/2022] Open
Abstract
Transient receptor potential canonical (TRPC) channels are ubiquitously expressed in excitable and non-excitable cardiac cells where they sense and respond to a wide variety of physical and chemical stimuli. As other TRP channels, TRPC channels may form homo or heterotetrameric ion channels, and they can associate with other membrane receptors and ion channels to regulate intracellular calcium concentration. Dysfunctions of TRPC channels are involved in many types of cardiovascular diseases. Significant increase in the expression of different TRPC isoforms was observed in different animal models of heart infarcts and in vitro experimental models of ischemia and reperfusion. TRPC channel-mediated increase of the intracellular Ca2+ concentration seems to be required for the activation of the signaling pathway that plays minor roles in the healthy heart, but they are more relevant for cardiac responses to ischemia, such as the activation of different factors of transcription and cardiac hypertrophy, fibrosis, and angiogenesis. In this review, we highlight the current knowledge regarding TRPC implication in different cellular processes related to ischemia and reperfusion and to heart infarction.
Collapse
|
17
|
Besnier M, Shantikumar S, Anwar M, Dixit P, Chamorro-Jorganes A, Sweaad W, Sala-Newby G, Madeddu P, Thomas AC, Howard L, Mushtaq S, Petretto E, Caporali A, Emanueli C. miR-15a/-16 Inhibit Angiogenesis by Targeting the Tie2 Coding Sequence: Therapeutic Potential of a miR-15a/16 Decoy System in Limb Ischemia. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 17:49-62. [PMID: 31220779 PMCID: PMC6586592 DOI: 10.1016/j.omtn.2019.05.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 04/09/2019] [Accepted: 05/05/2019] [Indexed: 12/13/2022]
Abstract
MicroRNA-15a (miR-15a) and miR-16, which are transcribed from the miR-15a/miR-16-1 cluster, inhibit post-ischemic angiogenesis. MicroRNA (miRNA) binding to mRNA coding sequences (CDSs) is a newly emerging mechanism of gene expression regulation. We aimed to (1) identify new mediators of the anti-angiogenic action of miR-15a and -16, (2) develop an adenovirus (Ad)-based miR-15a/16 decoy system carrying a luciferase reporter (Luc) to both sense and inhibit miR-15a/16 activity, and (3) investigate Ad.Luc-Decoy-15a/16 therapeutic potential in a mouse limb ischemia (LI) model. LI increased miR-15a and -16 expression in mouse muscular endothelial cells (ECs). The miRNAs also increased in cultured human umbilical vein ECs (HUVECs) exposed to serum starvation, but not hypoxia. Using bioinformatic tools and luciferase activity assays, we characterized miR-15a and -16 binding to Tie2 CDS. In HUVECs, miR-15a or -16 overexpression reduced Tie2 at the protein, but not the mRNA, level. Conversely, miR-15a or -16 inhibition improved angiogenesis in a Tie2-dependent manner. Local Ad.Luc-Decoy-15a/16 delivery increased Tie2 levels in ischemic skeletal muscle and improved post-LI angiogenesis and perfusion recovery, with reduced toe necrosis. Bioluminescent imaging (in vivo imaging system [IVIS]) provided evidence that the Ad.Luc-Decoy-15a/16 system responds to miR-15a/16 increases. In conclusion, we have provided novel mechanistic evidence of the therapeutic potential of local miR-15a/16 inhibition in LI.
Collapse
Affiliation(s)
- Marie Besnier
- Bristol Heart Institute, University of Bristol, Bristol, UK
| | | | - Maryam Anwar
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Parul Dixit
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Walid Sweaad
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Paolo Madeddu
- Bristol Heart Institute, University of Bristol, Bristol, UK
| | - Anita C Thomas
- Bristol Heart Institute, University of Bristol, Bristol, UK
| | - Lynsey Howard
- Bristol Heart Institute, University of Bristol, Bristol, UK
| | - Sobia Mushtaq
- Bristol Heart Institute, University of Bristol, Bristol, UK
| | - Enrico Petretto
- Institute of Clinical Sciences, Imperial College London, London, UK; Cardiovascular & Metabolic Disorders Programme, Centre for Computational Biology, Duke NUS Medical School, Singapore, Singapore
| | - Andrea Caporali
- Bristol Heart Institute, University of Bristol, Bristol, UK; BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Costanza Emanueli
- Bristol Heart Institute, University of Bristol, Bristol, UK; National Heart and Lung Institute, Imperial College London, London, UK.
| |
Collapse
|
18
|
Kukuła K, Urbanowicz A, Kłopotowski M, Dąbrowski M, Pręgowski J, Kądziela J, Chmielak Z, Witkowski A, Rużyłło W. Long-term follow-up and safety assessment of angiogenic gene therapy trial VIF-CAD: Transcatheter intramyocardial administration of a bicistronic plasmid expressing VEGF-A165/bFGF cDNA for the treatment of refractory coronary artery disease. Am Heart J 2019; 215:78-82. [PMID: 31288177 DOI: 10.1016/j.ahj.2019.06.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 06/07/2019] [Indexed: 10/26/2022]
Abstract
There have been a number of angiogenic gene therapy trials, yielding mixed results as to efficacy, but demonstrating uniform short-term treatment safety. Data regarding long-term safety of angiogenic gene therapy are limited. Double-blind VIF-CAD trial (NCT00620217) assessed myocardial perfusion and clinical data in 52 refractory coronary artery disease (CAD) patients randomized into treatment (VIF; n = 33) and Placebo (n = 19) arms. VIF group received electromechanical system NOGA-guided intramyocardial injections of VEGF-A165/bFGF plasmid (VIF) into ischemic regions, while the Placebo group-placebo plasmid injections. Full 1-year follow-up data have been published. This study presents the results of over 10-year (median 133 months, range 95-149) safety follow-up of VIF-CAD patients. Overall, 12 (36.4%) patients died in VIF and 8 (42.1%) in Placebo group (P = .68). Cardiovascular mortality was 12/33 (36.4%) in the VIF group and 6/19 (31.6%) in Placebo group (P = .73). Two Placebo patients died due to malignancies, but no VIF patients (P = .17). The Kaplan-Meier curves of combined endpoint: cardiovascular mortality, myocardial infarction and stroke were similar for both patient groups (P = .71). Odds ratio of Placebo group increasing (reaching a worse) their CCS class versus VIF was non-significant (OR 1.28, 95% CI = 0.66-2.45; P = .47). However, CCS class improved in time irrespectively of treatment-OR of reaching a less favorable CCS class per each year of follow-up was 0.74 (95% CI 0.685-0.792; P < .0001, pooled data). There were no differences in readmission rates. Intramyocardial VEGF-A165/bFGF plasmid administration appears safe, with no evidence of an increase in the incidence of death, malignancy, myocardial infarction or stroke during 10-year follow-up in this limited patient population.
Collapse
|
19
|
Shabunin AV, Matveev DV, Kuznetsov MR, Matveev AD. [Conservative treatment of chronic lower limb ischemia in ambulatory practice (in Russian only)]. Khirurgiia (Mosk) 2019:98-104. [PMID: 30938364 DOI: 10.17116/hirurgia201903198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Peripheral artery disease is still one of the most important surgical problems. General surgeons treat the majority of patients with chronic lower limb ischemia due to the lack of specialized surgical care. Current methods for risk factors adjustment, exercise therapy and the most common drugs for intermittent claudication management are reviewed in the article. The effect of these medicines on subjective (pain-free walking distance, maximal walking distance, etc.) and objective (ankle-brachial index) parameters and the incidence of complications are analyzed.
Collapse
Affiliation(s)
- A V Shabunin
- Russian Medical Academy of Postgraduate Education, Moscow, Russia
| | - D V Matveev
- Russian Medical Academy of Postgraduate Education, Moscow, Russia
| | - M R Kuznetsov
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - A D Matveev
- Russian Medical Academy of Postgraduate Education, Moscow, Russia
| |
Collapse
|
20
|
Imaging the Proangiogenic Effects of Cardiovascular Drugs in a Diabetic Model of Limb Ischemia. CONTRAST MEDIA & MOLECULAR IMAGING 2019; 2019:2538909. [PMID: 30863219 PMCID: PMC6378011 DOI: 10.1155/2019/2538909] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 10/03/2018] [Accepted: 10/18/2018] [Indexed: 02/02/2023]
Abstract
Purpose Peripheral artery disease (PAD) causes narrowing of arteries in the limbs, leading to tissue ischemia, gangrene, and eventually limb amputation. The presence of diabetes greatly exacerbates the course of PAD, accounting for the majority of lower limb amputations. Therapeutic strategies focussing on macrovascular repair are less effective in diabetic patients where smaller vessels are affected, and proangiogenic therapies offer a viable adjunct to improve vascularisation in these at risk individuals. The purpose of the current study was to assess the proangiogenic effects of drugs routinely used to treat cardiovascular disease in a diabetic murine model of hind limb ischemia longitudinally using multimodal imaging. Procedures Diabetic mice underwent surgical intervention to induce hind limb ischemia and were treated with simvastatin, metformin, or a combination orally for 28 days and compared to diabetic and nondiabetic mice. Neovascularisation was assessed using [18F]FtRGD PET imaging, and macrovascular volume was assessed by quantitative time of flight MRI. At each imaging time point, VEGF expression and capillary vessel density were quantified using immunohistochemical analysis, and functional recovery and disease progression were assessed. Results Combined use of simvastatin and metformin significantly increased neovascularisation above levels measured with either treatment alone. Early angiogenic events were accurately assessed using PET [18F]FtRGD, showing maximal retention in the ischemic hind limb by day 8, which translated to a sustained increase in vascular volume at later time points. Immunohistochemical analysis shows that combined therapy significantly increased VEGF expression and capillary density (CD31+) in a similar time course and also slowed disease progression while simultaneously improving functional foot use. Conclusions Combined treatment with simvastatin and metformin led to a significant improvement in limb angiogenesis, vascular volume, and sustained functional recovery in a diabetic murine model of HLI. PET imaging with [18F]FtRGD provides a robust method for early detection of these proangiogenic effects preclinically and may be useful for the assessment of proangiogenic therapies used clinically to treat diabetic PAD patients.
Collapse
|
21
|
Zhu Y, Gao M, Zhou T, Xie M, Mao A, Feng L, Yao X, Wong WT, Ma X. The TRPC5 channel regulates angiogenesis and promotes recovery from ischemic injury in mice. J Biol Chem 2018; 294:28-37. [PMID: 30413532 DOI: 10.1074/jbc.ra118.005392] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 11/06/2018] [Indexed: 11/06/2022] Open
Abstract
Ischemia-related diseases are a leading cause of death worldwide, and promoting therapeutic angiogenesis is key for effective recovery from hypoxia-ischemia. Given the limited success of angiogenic factors, such as vascular endothelial growth factor, in clinical trials, it is important to find more promising angiogenic targets. Here, using both cell- and tissue-based assays and a mouse model of injury-induced ischemia, we investigated the involvement of the transient receptor potential canonical 5 (TRPC5) ion channel in angiogenesis and the effects of a TRPC5 activator, the Food and Drug Administration-approved drug riluzole, on recovery from ischemic injury. We demonstrate that TRPC5 is involved in endothelial cell sprouting, angiogenesis, and blood perfusion in an oxygen-induced retinopathy model and a hind limb ischemia model. We found a potential regulatory link between nuclear factor of activated T cell isoform c3 and angiopoietin-1 that could provide the mechanistic basis for the angiogenic function of TRPC5. Importantly, treatment with riluzole, which can activate TRPC5 in endothelial cells, improved recovery from ischemia in mice. Our study reveals TRPC5 as a potential angiogenic target and suggests riluzole as a promising drug for managing ischemic diseases.
Collapse
Affiliation(s)
- Yifei Zhu
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214000, China; School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong 999077, China
| | - Mengru Gao
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214000, China
| | - Tingting Zhou
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214000, China
| | - Mingxu Xie
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214000, China; School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong 999077, China
| | - Aiqin Mao
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214000, China
| | - Lei Feng
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214000, China
| | - Xiaoqiang Yao
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong 999077, China
| | - Wing Tak Wong
- State Key Laboratory of Agrobiotechnology (CUHK), School of Life Sciences, Chinese University of Hong Kong, Hong Kong 999077, China
| | - Xin Ma
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214000, China.
| |
Collapse
|
22
|
Abstract
BACKGROUND Peripheral arterial disease (PAD), caused by narrowing of the arteries in the limbs, is increasing in incidence and prevalence as our population is ageing and as diabetes is becoming more prevalent. PAD can cause pain in the limbs while walking, known as intermittent claudication, or can be more severe and cause pain while at rest, ulceration, and ultimately gangrene and limb loss. This more severe stage of PAD is known as 'critical limb ischaemia'. Treatments for PAD include medications that help to reduce the increased risk of cardiovascular events and help improve blood flow, as well as endovascular or surgical repair or bypass of the blocked arteries. However, many people are unresponsive to medications and are not suited to surgical or endovascular treatment, leaving amputation as the last option. Gene therapy is a novel approach in which genetic material encoding for proteins that may help increase revascularisation is injected into the affected limbs of patients. This type of treatment has been shown to be safe, but its efficacy, especially regarding ulcer healing, effects on quality of life, and other symptomatic outcomes remain unknown. OBJECTIVES To assess the effects of gene therapy for symptomatic peripheral arterial disease. SEARCH METHODS The Cochrane Vascular Information Specialist searched Cochrane CENTRAL, the Cochrane Vascular Specialised Register, MEDLINE Ovid, Embase Ovid, CINAHL, and AMED, along with trials registries (all searched 27 November 2017). We also checked reference lists of included studies and systematic reviews for further studies. SELECTION CRITERIA We included randomised and quasi-randomised studies that evaluated gene therapy versus no gene therapy in people with PAD. We excluded studies that evaluated direct growth hormone treatment or cell-based treatments. DATA COLLECTION AND ANALYSIS Two review authors independently selected studies, performed quality assessment, and extracted data from the included studies. We collected pertinent information on each study, as well as data for the outcomes of amputation-free survival, ulcer healing, quality of life, amputation, all-cause mortality, ankle brachial index, symptom scores, and claudication distance. MAIN RESULTS We included in this review a total of 17 studies with 1988 participants (evidence current until November 2017). Three studies limited their inclusion to people with intermittent claudication, 12 limited inclusion to people with varying levels of critical limb ischaemia, and two included people with either condition. Study investigators evaluated many different types of gene therapies, using different protocols. Most studies evaluated growth factor-encoding gene therapy, with six studies using vascular endothelial growth factor (VEGF)-encoding genes, four using hepatocyte growth factor (HGF)-encoding genes, and three using fibroblast growth factor (FGF)-encoded genes. Two studies evaluated hypoxia-inducible factor 1-alpha (HIF-1α) gene therapy, one study used a developmental endothelial locus-1 gene therapy, and the final study evaluated a stromal cell-derived factor-1 (SDF-1) gene therapy. Most studies reported outcomes after 12 months of follow-up, but follow-up ranged from three months to two years.Overall risk of bias varied between studies, with many studies not providing sufficient detail for adequate determination of low risk of bias for many domains. Two studies did not utilise a placebo control, leading to risk of performance bias. Several studies reported in previous protocols or in their Methods sections that they would report on certain outcomes for which no data were then reported, increasing risk of reporting bias. All included studies reported sponsorships from corporate entities that led to unclear risk of other bias. The overall quality of evidence ranged from moderate to very low, generally as the result of heterogeneity and imprecision, with few or no studies reporting on outcomes.Evidence suggests no clear differences for the outcomes of amputation-free survival, major amputation, and all-cause mortality between those treated with gene therapy and those not receiving this treatment (all moderate-quality evidence). Low-quality evidence suggests improvement in complete ulcer healing with gene therapy (odds ratio (OR) 2.16, 95% confidence interval (CI) 1.02 to 4.59; P = 0.04). We could not combine data on quality of life and can draw no conclusions at this time regarding this outcome (very low-quality evidence). We included one study in the meta-analysis for ankle brachial index, which showed no clear differences between treatments, but we can draw no overall association (low-quality evidence). We combined in a meta-analysis pain symptom scores as assessed by visual analogue scales from two studies and found no clear differences between treatment groups (very low-quality evidence). We carried out extensive subgroup analyses by PAD classification, dosage schedule, vector type, and gene used but identified no substantial differences. AUTHORS' CONCLUSIONS Moderate-quality evidence shows no clear differences in amputation-free survival, major amputation, and all-cause mortality between those treated with gene therapy and those not receiving gene therapy. Some evidence suggests that gene therapy may lead to improved complete ulcer healing, but this outcome needs to be explored with improved reporting of the measure, such as decreased ulcer area in cm², and better description of ulcer types and healing. Further standardised data that are amenable to meta-analysis are needed to evaluate other outcomes such as quality of life, ankle brachial index, symptom scores, and claudication distance.
Collapse
Affiliation(s)
- Rachel Forster
- University of EdinburghUsher Institute of Population Health Sciences and InformaticsEdinburghUKEH8 9AG
| | - Aaron Liew
- Newcastle UniversityInstitute of Cellular Medicine4th Floor, William Leech BuildingFramlington PlaceNewcastle upon TyneUKNE2 4HH
- National University of Ireland Galway (NUIG), Portiuncula University Hospital & Galway University Hospital, Saolta University Health Care GroupGalwayIreland
| | - Vish Bhattacharya
- Queen Elizabeth HospitalDepartment of General and Vascular SurgeryQueen Elizabeth AvenueSheriff HillGatesheadTyne and WearUKNE9 6SX
| | - James Shaw
- Newcastle UniversityInstitute of Cellular Medicine4th Floor, William Leech BuildingFramlington PlaceNewcastle upon TyneUKNE2 4HH
| | - Gerard Stansby
- Freeman HospitalNorthern Vascular CentreNewcastle upon TyneUKNE7 7DN
| | | |
Collapse
|
23
|
Vascular Endothelial Growth Factor Enhances Compensatory Lung Growth in Piglets. Surgery 2018; 164:1279-1286. [PMID: 30193736 DOI: 10.1016/j.surg.2018.07.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 06/25/2018] [Accepted: 07/05/2018] [Indexed: 01/12/2023]
Abstract
BACKGROUND Vascular endothelial growth factor has been found to accelerate compensatory lung growth after left pneumonectomy in mice. The aim of this study was to determine the natural history and the effects of vascular endothelial growth factor on compensatory lung growth in a large animal model. METHODS To determine the natural history of compensatory lung growth, female Yorkshire piglets underwent a left pneumonectomy on days of life 10-11. Tissue harvest and volume measurement of the right lung were performed at baseline (n = 5) and on postoperative days 7 (n = 5), 14 (n = 4), and 21 (n = 5). For pharmacokinetic studies, vascular endothelial growth factor was infused via a central venous catheter, with plasma vascular endothelial growth factor levels measured at various time points. To test the effect of vascular endothelial growth factor on compensatory lung growth, 26 female Yorkshire piglets underwent a left pneumonectomy followed by daily infusion of vascular endothelial growth factor at 200 µg/kg or isovolumetric 0.9% NaCl (saline control). Lungs were harvested on postoperative day 7 for volume measurement and morphometric analyses. RESULTS Compared with baseline, right lung volume after left pneumonectomy increased by factors of 2.1 ± 0.6, 3.3 ± 0.6, and 3.6 ± 0.4 on postoperative days 7, 14, and 21, respectively. The half-life of VEGF ranged from 89 to 144 minutes. Lesser doses of vascular endothelial growth factor resulted in better tolerance, volume of distribution, and clearance. Compared with the control group, piglets treated with vascular endothelial growth factor had greater lung volume (P < 0.0001), alveolar volume (P = 0.001), septal surface area (P = 0.007) and total alveolar count (P = 0.01). CONCLUSION Vascular endothelial growth factor enhanced alveolar growth in neonatal piglets after unilateral pneumonectomy.
Collapse
|
24
|
Deev R, Plaksa I, Bozo I, Mzhavanadze N, Suchkov I, Chervyakov Y, Staroverov I, Kalinin R, Isaev A. Results of 5-year follow-up study in patients with peripheral artery disease treated with PL-VEGF165 for intermittent claudication. Ther Adv Cardiovasc Dis 2018; 12:237-246. [PMID: 29996720 PMCID: PMC6116753 DOI: 10.1177/1753944718786926] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 05/04/2018] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND The effective treatment of chronic lower limb ischemia is one of the most challenging issues confronting vascular surgeons. Current pharmacological therapies play an auxiliary role and cannot prevent disease progression, and new treatment methods are needed. In 2011, a plasmid VEGF65-gene therapy drug was approved in Russia for the treatment of chronic lower limb ischemia ( ClinicalTrials.gov identifier: NCT03068585). The objective of this follow-up study was to evaluate the long-term safety and efficacy of gene therapy in patients with limb ischemia of atherosclerotic genesis. AIMS To evaluate the long-term safety and efficacy of the therapeutic angiogenesis, 36 patients in the treatment group (pl- VEGF165) and 12 patients in the control group participated in a 5-year follow-up study. Planned examinations were carried out annually for 5 years after pl- VEGF165 administration. RESULTS Differences in the frequency of major cardiovascular events (pl- VEGF165 5/36 versus control 2/12; p = 0.85), malignancies (pl- VEGF165 1/36 versus control 0/12; p = 0.38) and impaired vision (there was none in either group) over the 5-year follow-up period did not achieve statistical significance. The target limb salvage was 95% ( n = 36) and 67% ( n = 12) in the pl- VEGF165 and control groups, respectively. The pain-free walking distance value increased by 288% from 105.7 ± 16.5 m to 384 ± 39 m in the treatment group by the end of the fifth year, with a peak of 410.6 ± 86.1 m achieved by the end of the third year. The ankle-brachial index (ABI) increased from 0.47 ± 0.01 to 0.56 ± 0.02 by the end of the first year, with a subsequent slight decrease to 0.51 ± 0.02 by the fifth year. The maximum increment of transcutaneous oximetry test (tcoO2) by 36%, from 66.6 ± 3.7 mm Hg to 90.7 ± 4.9 mm Hg, was observed by the end of the second year. CONCLUSION The therapeutic effect of angiogenesis induction by gene therapy persists for 5 years.
Collapse
Affiliation(s)
- Roman Deev
- Human Stem Cells Institute, Moscow, Russia Ryazan I.P. Pavlov State Medical University, Ryazan, Russia
| | - Igor Plaksa
- Human Stem Cells Institute, Moscow, Russia Moscow City Oncology Hospital No 62, Moscow, Russia
| | - Ilia Bozo
- Human Stem Cells Institute, Moscow, Russia A.I. Evdokimov Moscow State University of Medicine and Dentistry, Moscow, Russia
| | | | - Igor Suchkov
- Ryazan I.P. Pavlov State Medical University, Ryazan, Russia
| | | | | | - Roman Kalinin
- Ryazan I.P. Pavlov State Medical University, Ryazan, Russia
| | | |
Collapse
|
25
|
David AL. Maternal uterine artery VEGF gene therapy for treatment of intrauterine growth restriction. Placenta 2017; 59 Suppl 1:S44-S50. [DOI: 10.1016/j.placenta.2017.09.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 09/18/2017] [Accepted: 09/25/2017] [Indexed: 11/24/2022]
|
26
|
Formulation of the bivalent prostate cancer vaccine with surgifoam elicits antigen-specific effector T cells in PSA-transgenic mice. Vaccine 2017; 35:5794-5798. [PMID: 28939158 DOI: 10.1016/j.vaccine.2017.09.037] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 08/07/2017] [Accepted: 09/12/2017] [Indexed: 11/24/2022]
Abstract
We previously developed and characterized an adenoviral-based prostate cancer vaccine for simultaneous targeting of prostate-specific antigen (PSA) and prostate stem cell antigen (PSCA). We also demonstrated that immunization of mice with the bivalent vaccine (Ad5-PSA+PSCA) inhibited the growth of established prostate tumors. However, there are multiple challenges hindering the success of immunological therapies in the clinic. One of the prime concerns has been to overcome the immunological tolerance and maintenance of long-term effector T cells. In this study, we further characterized the use of the bivalent vaccine (Ad5-PSA+PSCA) in a transgenic mouse model expressing human PSA in the mouse prostate. We demonstrated the expression of PSA analyzed at the mRNA level (by RT-PCR) and protein level (by immunohistochemistry) in the prostate lobes harvested from the PSA-transgenic (PSA-Tg) mice. We established that the administration of the bivalent vaccine in surgifoam to the PSA-Tg mice induces strong PSA-specific effector CD8+ T cells as measured by IFN-γ secretion and in vitro cytotoxic T-cell assay. Furthermore, the use of surgifoam with Ad5-PSA+PSCA vaccine allows multiple boosting vaccinations with a significant increase in antigen-specific CD8+ T cells. These observations suggest that the formulation of the bivalent prostate cancer vaccine (Ad5-PSA+PSCA) with surgifoam bypasses the neutralizing antibody response, thus allowing multiple boosting. This formulation is also helpful for inducing an antigen-specific immune response in the presence of self-antigen, and maintains long-term effector CD8+ T cells.
Collapse
|
27
|
Kim J, Mirando AC, Popel AS, Green JJ. Gene delivery nanoparticles to modulate angiogenesis. Adv Drug Deliv Rev 2017; 119:20-43. [PMID: 27913120 PMCID: PMC5449271 DOI: 10.1016/j.addr.2016.11.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Revised: 10/01/2016] [Accepted: 11/24/2016] [Indexed: 01/19/2023]
Abstract
Angiogenesis is naturally balanced by many pro- and anti-angiogenic factors while an imbalance of these factors leads to aberrant angiogenesis, which is closely associated with many diseases. Gene therapy has become a promising strategy for the treatment of such a disordered state through the introduction of exogenous nucleic acids that express or silence the target agents, thereby engineering neovascularization in both directions. Numerous non-viral gene delivery nanoparticles have been investigated towards this goal, but their clinical translation has been hampered by issues associated with safety, delivery efficiency, and therapeutic effect. This review summarizes key factors targeted for therapeutic angiogenesis and anti-angiogenesis gene therapy, non-viral nanoparticle-mediated approaches to gene delivery, and recent gene therapy applications in pre-clinical and clinical trials for ischemia, tissue regeneration, cancer, and wet age-related macular degeneration. Enhanced nanoparticle design strategies are also proposed to further improve the efficacy of gene delivery nanoparticles to modulate angiogenesis.
Collapse
Affiliation(s)
- Jayoung Kim
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Translational Tissue Engineering Center and Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Adam C Mirando
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Jordan J Green
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Translational Tissue Engineering Center and Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Departments of Ophthalmology, Neurosurgery, and Materials Science & Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| |
Collapse
|
28
|
Lähteenvuo J, Ylä-Herttuala S. Advances and Challenges in Cardiovascular Gene Therapy. Hum Gene Ther 2017; 28:1024-1032. [PMID: 28810808 DOI: 10.1089/hum.2017.129] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Many promising cardiovascular gene therapy approaches have failed to fulfill expectations in clinical trials. However, 20 years of research and method development has laid a solid groundwork for future therapies, and the need for new treatment options still exists. The safety of gene therapy has been established with various viral vectors, transgenes and delivery methods. Improving success in clinical settings requires careful consideration of the translational process. This requires both improving animal models and preclinical end points, and new approach in patient recruitment and selection of clinical end points. This review focuses on bidirectional translationality from bench to bedside and back and proposes ways to improve the process. Developing a highly complex new therapy has taken an enormous amount of work and resources, but perhaps now after the hard lessons cardiovascular gene therapy is ready become a clinical reality.
Collapse
Affiliation(s)
- Johanna Lähteenvuo
- 1 A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Seppo Ylä-Herttuala
- 1 A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland .,2 Heart Center and Gene Therapy Unit, Kuopio University Hospital , Kuopio, Finland
| |
Collapse
|
29
|
Abstract
BACKGROUND Peripheral artery disease (PAD) is associated with a high clinical and socioeconomic burden. Treatments to alleviate the symptoms of PAD and decrease the risks of amputation and death are a high societal priority. A number of growth factors have shown a potential to stimulate angiogenesis. Growth factors delivered directly (as recombinant proteins), or indirectly (e.g. by viral vectors or DNA plasmids encoding these factors), have emerged as a promising strategy to treat patients with PAD. OBJECTIVES To assess the effects of growth factors that promote angiogenesis for treating people with PAD of the lower extremities. SEARCH METHODS The Cochrane Vascular Information Specialist searched the Specialised Register (June 2016) and CENTRAL (2016, Issue 5). We searched trial registries for details of ongoing or unpublished studies. We also checked the reference lists of relevant publications and, if necessary, tried to contact the trialists for details of the studies. SELECTION CRITERIA We included randomised controlled trials comparing growth factors (delivered directly or indirectly) with no intervention, placebo or any other intervention not based on the growth factor's action in patients with PAD of the lower extremities. The primary outcomes were limb amputation, death and adverse events. The secondary outcomes comprised walking ability, haemodynamic measures, ulceration and rest pain. DATA COLLECTION AND ANALYSIS Two review authors independently selected trials and assessed the risk of bias. We used outcomes of the studies at low risk of bias for the main analysis and of all studies in the sensitivity analyses. We calculated odds ratios (OR) for dichotomous outcomes and mean differences for continuous outcomes with 95% confidence intervals (CI). We evaluated statistical heterogeneity using the I2 statistic and Cochrane's Q test. We conducted meta-analysis for the overall effect and for each growth factor as a subgroup analysis using OR in a fixed-effect model. We evaluated the robustness of the results in a sensitivity analysis using risk ratio (RR) and/or a random-effects model. We also assessed the quality of the evidence for each outcome. MAIN RESULTS We included 20 trials in the review and used 14 studies (on approximately 1400 participants) with published results in the analyses. Six published studies compared fibroblast growth factors (FGF), four studies hepatocyte growth factors (HGF) and another four studies vascular endothelial growth factors (VEGF), versus placebo or no therapy. Six of these studies exclusively or mainly investigated participants with intermittent claudication and eight studies exclusively participants with critical limb ischaemia. Follow-up generally ranged from three months to one year. Two small studies provided some data at 2 years and one of them also at 10 years.The direction and size of effects for growth factors on major limb amputations (OR 0.99, 95% CI 0.71 to 1.38; 10 studies, N = 1075) and death (OR 0.99, 95% CI 0.69 to 1.41; 12 studies, N = 1371) at up to two years are uncertain. The quality of the evidence is low due to risk of bias and imprecision (at one year, moderate-quality evidence due to imprecision). However, growth factors may decrease the rate of any limb amputations (OR 0.56, 95% CI 0.31 to 0.99; 6 studies, N = 415). The quality of the evidence is low due to risk of bias and selective reporting.The direction and size of effects for growth factors on serious adverse events (OR 1.09, 95% CI 0.79 to 1.50; 13 studies, N = 1411) and on any adverse events (OR 1.10, 95% CI 0.73 to 1.64; 4 studies, N = 709) at up to two years are also uncertain. The quality of the evidence is low due to risk of bias and imprecision (for serious adverse events at one year, moderate-quality evidence due to imprecision).Growth factors may improve haemodynamic measures (low-quality evidence), ulceration (very low-quality evidence) and rest pain (very low-quality evidence) up to one year, but they have little or no effect on walking ability (low-quality evidence). We did not identify any relevant differences in effects between growth factors (FGF, HGF and VEGF). AUTHORS' CONCLUSIONS The results of this review do not support the use of therapy with the growth factors FGF, HGF or VEGF in people with PAD of the lower extremities to prevent death or major limb amputation or to improve walking ability. However, the use of these growth factors may improve haemodynamic measures and decrease the rate of any limb amputations (probably due to preventing minor amputations) with an uncertain effect on adverse events; an improvement of ulceration and rest pain is very uncertain. New trials at low risk of bias are needed to generate evidence with more certainty.
Collapse
Affiliation(s)
- Vitali Gorenoi
- Hannover Medical SchoolEvidence based Medicine & Health Technology Assessment Working Group, Institute for Epidemiology, Social Medicine and Health Systems ResearchCarl‐Neuberg‐Str. 1HannoverGermany30625
| | - Michael U Brehm
- Hannover Medical SchoolDepartment for Cardiology and AngiologyCarl‐Neuberg‐Str. 1HannoverGermany30265
| | - Armin Koch
- Institute for Biometry, Hannover Medical SchoolCarl‐Neuberg‐Str. 1HannoverGermany30625
| | - Anja Hagen
- Hannover Medical SchoolEvidence based Medicine & Health Technology Assessment Working Group, Institute for Epidemiology, Social Medicine and Health Systems ResearchCarl‐Neuberg‐Str. 1HannoverGermany30625
| | | |
Collapse
|
30
|
Ylä-Herttuala S, Bridges C, Katz MG, Korpisalo P. Angiogenic gene therapy in cardiovascular diseases: dream or vision? Eur Heart J 2017; 38:1365-1371. [PMID: 28073865 PMCID: PMC5837788 DOI: 10.1093/eurheartj/ehw547] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 06/10/2016] [Accepted: 10/24/2016] [Indexed: 12/17/2022] Open
Abstract
Chronic cardiovascular diseases are significant health problems. Although current treatment strategies have tremendously improved disease management, up to 30% of these patients cannot be successfully treated with current treatment approaches and new treatment strategies are clearly needed. Gene therapy and therapeutic vascular growth may provide a new treatment option for these patients. Several growth factors, like vascular endothelial growth factors, fibroblast growth factors and hepatocyte growth factor have been tested in clinical trials. However, apart from demonstration of increased vascularity, very few results with clinical significance have been obtained. Problems with gene transfer efficiency, short duration of transgene expression, selection of endpoints, and suboptimal patients for gene therapy have been recognized. Ongoing gene therapy trials have included improvements in study protocols, vector delivery and endpoints, addressing the identified problems. Better, targeted delivery systems and new, more optimal growth factors have been taken to clinical testing. Recent advances in these areas will be discussed and the concept of angiogenic therapy as a sole treatment is re-evaluated. A combination with regenerative therapies or standard revascularization operations might be needed to improve tissue function and clinical benefits.
Collapse
Affiliation(s)
- Seppo Ylä-Herttuala
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, Kuopio 70211, Finland
- Heart Center, Kuopio University Hospital, Puijonlaaksontie 2, 70210 Kuopio, Finland
| | - Charles Bridges
- Cardiovascular Research Center, Mount Sinai School of Medicine, 1470 Madison Avenue, New York, New York 10029, USA
| | - Michael G. Katz
- Cardiovascular Research Center, Mount Sinai School of Medicine, 1470 Madison Avenue, New York, New York 10029, USA
| | - Petra Korpisalo
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, Kuopio 70211, Finland
| |
Collapse
|
31
|
Ylä-Herttuala S, Baker AH. Cardiovascular Gene Therapy: Past, Present, and Future. Mol Ther 2017; 25:1095-1106. [PMID: 28389321 PMCID: PMC5417840 DOI: 10.1016/j.ymthe.2017.03.027] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 03/21/2017] [Accepted: 03/21/2017] [Indexed: 12/12/2022] Open
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.
Collapse
Affiliation(s)
- Seppo Ylä-Herttuala
- A.I. Virtanen Institute, University of Eastern Finland, Yliopistonranta 1, 70211 Kuopio, Finland; Heart Center and Gene Therapy Unit, Kuopio University Hospital, PO Box 100, 70029 KYS Kuopio, Finland.
| | - Andrew H Baker
- Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| |
Collapse
|
32
|
Late responses to adenoviral-mediated transfer of the aquaporin-1 gene for radiation-induced salivary hypofunction. Gene Ther 2016; 24:176-186. [PMID: 27996967 PMCID: PMC5373995 DOI: 10.1038/gt.2016.87] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 12/09/2016] [Accepted: 12/14/2016] [Indexed: 12/14/2022]
Abstract
We evaluated late effects of AdhAQP1 administration in five subjects in a clinical trial for radiation-induced salivary hypofunction (http://www.clinicaltrials.gov/ct/show/NCT00372320?order=). All were identified as initially responding to human aquaporin-1 (hAQP1) gene transfer (Baum et al, 2012). They were followed for 3-4 years after AdhAQP1 delivery to one parotid gland. At intervals we examined salivary flow, xerostomic symptoms, saliva composition, vector presence and efficacy in the targeted gland, clinical laboratory data, and adverse events. All displayed marked increases (71-500% above baseline) in parotid flow 3-4.7 years after treatment, with improved symptoms for ~ 2-3 years. There were some changes in [Na+] and [Cl−] consistent with elevated salivary flow, but no uniform changes in secretion of key parotid proteins. There were no clinically significant adverse events, nor consistent negative changes in laboratory parameters. One subject underwent a core needle biopsy of the targeted parotid gland 3.1 years post treatment and displayed evidence of hAQP1 protein in acinar, but not duct, cell membranes. All subjects responding to hAQP1 gene transfer initially had benefits for much longer times. First generation adenoviral vectors typically yield transit effects, but these data show beneficial effects can continue years after parotid gland delivery.
Collapse
|
33
|
Niyaz M, Gürpınar ÖA, Oktar GL, Günaydın S, Onur MA, Özsin KK, Yener A. Effects of VEGF and MSCs on vascular regeneration in a trauma model in rats. Wound Repair Regen 2016; 23:262-7. [PMID: 25754793 DOI: 10.1111/wrr.12278] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Accepted: 02/25/2015] [Indexed: 12/26/2022]
Abstract
In the human body, vascular injuries that are caused by trauma, vessel lumen stenosis, and occlusions are often irreversible and can lead to sequelae formation as the vessels cannot reproduce fast enough. To solve this problem, the blood flow must be returned to the region as fast as possible. The adipose tissue contains progenitor cells with angiogenic potential and can be used to resolve the issue. In the present study, mesenchymal stem cells (MSCs) derived from rat adipose tissue, vascular endothelial growth factor (VEGF), and their mixture were applied on the dorsum of a rat, which was traumatized and its contribution to vascular regeneration was reviewed. No application was made to the control group. The results showed that the percentage of necrotic area was significantly lower in the MSC group than that of all the other groups. When the VEGF group was compared to the VEGF + MSCs, the percentage of necrotic area was observed to be similiar. However, VEGF showed effects only when a large quantites of VEGF was applied to the flap area. VEGF could not fully respond to the needs, whereas MSCs can produce VEGF according to the needs of tissue. This makes them superior to stem cells.
Collapse
Affiliation(s)
- Mehmet Niyaz
- Department of Cardiovascular Surgery, Muş State Hospital, Muş, Turkey
| | - Özer Aylin Gürpınar
- Faculty of Science, Department of Biology, Hacettepe University, Ankara, Turkey
| | - Gürsel Levent Oktar
- Faculty of Medicine, Department of Cardiovascular Surgery, Gazi University, Ankara, Turkey
| | - Serdar Günaydın
- Department of Cardiovascular Surgery, Medline Eskişehir Hospital, Eskişehir, Turkey
| | - Mehmet Ali Onur
- Faculty of Science, Department of Biology, Hacettepe University, Ankara, Turkey
| | - Kadir Kaan Özsin
- Department of Cardiovascular Surgery, Bursa Training and Reseach Hospital, Bursa, Turkey
| | - Ali Yener
- Faculty of Medicine, Department of Cardiovascular Surgery, Gazi University, Ankara, Turkey
| |
Collapse
|
34
|
Efficacy and safety of myocardial gene transfer of adenovirus, adeno-associated virus and lentivirus vectors in the mouse heart. Gene Ther 2015; 23:296-305. [PMID: 26704723 DOI: 10.1038/gt.2015.114] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 10/20/2015] [Accepted: 12/21/2015] [Indexed: 01/18/2023]
Abstract
Gene therapy is a promising new treatment option for cardiac diseases. For finding the most suitable and safe vector for cardiac gene transfer, we delivered adenovirus (AdV), adeno-associated virus (AAV) and lentivirus (LeV) vectors into the mouse heart with sophisticated closed-chest echocardiography-guided intramyocardial injection method for comparing them with regards to transduction efficiency, myocardial damage, effects on the left ventricular function and electrocardiography (ECG). AdV had the highest transduction efficiency in cardiomyocytes followed by AAV2 and AAV9, and the lowest efficiency was seen with LeV. The local myocardial inflammation and fibrosis in the left ventricle (LV) was proportional to transduction efficiency. AdV caused LV dilatation and systolic dysfunction. Neither of the locally injected AAV serotypes impaired the LV systolic function, but AAV9 caused diastolic dysfunction to some extent. LeV did not affect the cardiac function. We also studied systemic delivery of AAV9, which led to transduction of cardiomyocytes throughout the myocardium. However, also diffuse fibrosis was present leading to significantly impaired LV systolic and diastolic function and pathological ECG changes. Compared with widely used AdV vector, AAV2, AAV9 and LeV were less effective in transducing cardiomyocytes but also less harmful. Local administration of AAV9 was safer and more efficient compared with systemic administration.
Collapse
|
35
|
Current therapies and investigational drugs for peripheral arterial disease. Hypertens Res 2015; 39:183-91. [PMID: 26631852 DOI: 10.1038/hr.2015.134] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 10/18/2015] [Accepted: 10/19/2015] [Indexed: 12/17/2022]
Abstract
Peripheral artery disease (PAD) is associated with elevated morbidity and mortality with cardiovascular (CV) disease. The guideline recommends smoking cessation and antiplatelet/antithrombotic drugs for asymptomatic and symptomatic PAD patients. It also recommends that PAD patients with critical limb ischemia (CLI) should be considered to receive endovascular and open surgical treatment for limb salvage. Although PAD patients with CLI receive these treatments, they are sometimes unable to deliver sufficient blood flow to eliminate their symptoms. Thus specific strategies are needed to promote enough blood flow. To establish the effective method, many investigations have been performed using cell-based therapy. Endothelial progenitor cells, mononuclear cells and mesenchymal stem cells have been well investigated in clinical settings. To induce angiogenesis, vascular endothelial growth factor, fibroblast growth factor and hepatocyte growth factor (HGF) have also been transfected in PAD patients. Among them, HGF is the most promising factor because it can induce angiogenesis without the induction of vascular inflammation and increased permeability. In this review article, we summarize current treatments and investigational drugs of PAD.
Collapse
|
36
|
Rincon MY, VandenDriessche T, Chuah MK. Gene therapy for cardiovascular disease: advances in vector development, targeting, and delivery for clinical translation. Cardiovasc Res 2015; 108:4-20. [PMID: 26239654 PMCID: PMC4571836 DOI: 10.1093/cvr/cvv205] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 07/22/2015] [Indexed: 01/06/2023] Open
Abstract
Gene therapy is a promising modality for the treatment of inherited and acquired cardiovascular diseases. The identification of the molecular pathways involved in the pathophysiology of heart failure and other associated cardiac diseases led to encouraging preclinical gene therapy studies in small and large animal models. However, the initial clinical results yielded only modest or no improvement in clinical endpoints. The presence of neutralizing antibodies and cellular immune responses directed against the viral vector and/or the gene-modified cells, the insufficient gene expression levels, and the limited gene transduction efficiencies accounted for the overall limited clinical improvements. Nevertheless, further improvements of the gene delivery technology and a better understanding of the underlying biology fostered renewed interest in gene therapy for heart failure. In particular, improved vectors based on emerging cardiotropic serotypes of the adeno-associated viral vector (AAV) are particularly well suited to coax expression of therapeutic genes in the heart. This led to new clinical trials based on the delivery of the sarcoplasmic reticulum Ca2+-ATPase protein (SERCA2a). Though the first clinical results were encouraging, a recent Phase IIb trial did not confirm the beneficial clinical outcomes that were initially reported. New approaches based on S100A1 and adenylate cyclase 6 are also being considered for clinical applications. Emerging paradigms based on the use of miRNA regulation or CRISPR/Cas9-based genome engineering open new therapeutic perspectives for treating cardiovascular diseases by gene therapy. Nevertheless, the continuous improvement of cardiac gene delivery is needed to allow the use of safer and more effective vector doses, ultimately bringing gene therapy for heart failure one step closer to reality.
Collapse
Affiliation(s)
- Melvin Y Rincon
- Department of Gene Therapy and Regenerative Medicine, Free University of Brussels (VUB), Building D, room D306, Laarbeeklaan 103, Brussels, Belgium Centro de Investigaciones, Fundacion Cardiovascular de Colombia, Floridablanca, Colombia
| | - Thierry VandenDriessche
- Department of Gene Therapy and Regenerative Medicine, Free University of Brussels (VUB), Building D, room D306, Laarbeeklaan 103, Brussels, Belgium Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Marinee K Chuah
- Department of Gene Therapy and Regenerative Medicine, Free University of Brussels (VUB), Building D, room D306, Laarbeeklaan 103, Brussels, Belgium Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| |
Collapse
|
37
|
Vemulapalli S, Patel MR, Jones WS. Limb Ischemia: Cardiovascular Diagnosis and Management from Head to Toe. Curr Cardiol Rep 2015; 17:611. [DOI: 10.1007/s11886-015-0611-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
38
|
Skóra J, Pupka A, Janczak D, Barć P, Dawiskiba T, Korta K, Baczyńska D, Mastalerz-Migas A, Garcarek J. Combined autologous bone marrow mononuclear cell and gene therapy as the last resort for patients with critical limb ischemia. Arch Med Sci 2015; 11:325-31. [PMID: 25995748 PMCID: PMC4424239 DOI: 10.5114/aoms.2013.39935] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 04/02/2013] [Accepted: 05/17/2013] [Indexed: 11/29/2022] Open
Abstract
INTRODUCTION Our study was designed to investigate the safety and efficacy of combined autologous bone marrow mononuclear cell (MNC) and gene therapy in comparison to conventional drug therapy in patients with critical limb ischemia (CLI). MATERIAL AND METHODS Thirty-two patients with CLI persisting for 12-48 months (average time 27.5 months) were randomized into 2 groups, each group consisting of 16 patients. In the first group, administration of autologous bone marrow MNC and vascular endothelial growth factor (VEGF) plasmid was performed. The patients from the second group were treated pharmacologically with pentoxifylline. Ankle-brachial index (ABI) was measured and angiography was performed before and finally 3 months after treatment. The pain was evaluated using the Visual Analog Scale (VAS) before and after 3 months. RESULTS Ankle-brachial index improved significantly from 0.29 ±0.21 to 0.52 ±0.23 (p < 0.001) in 12 patients (75.0%) 3 months after the experimental therapy in group 1. In this group angiography showed the development of collateral vessels. Ischemic ulcers healed completely in 11 patients (68.75%). In group 2 the ABI did not improve in any patient; moreover the complete healing of skin ulcers was not found in any of the patients of this group. Amputation was performed in 4 (25.0%) patients in group 1, and in 8 patients (50%) from group 2. CONCLUSIONS These data after 3-month follow-up indicate that intramuscular injection of MNC combined with gene therapy in patients with chronic CLI is safe, and a more feasible and effective method of treatment than the conventional therapy. However, both therapies are limited by the degree of microcirculation damage.
Collapse
Affiliation(s)
- Jan Skóra
- Department of Vascular, General and Transplantation Surgery, Wroclaw Medical University, Wroclaw, Poland
| | - Artur Pupka
- Department of Vascular, General and Transplantation Surgery, Wroclaw Medical University, Wroclaw, Poland
| | - Dariusz Janczak
- Department of Clinical Procedures, Wroclaw Medical University, Wroclaw, Poland
| | - Piotr Barć
- Department of Vascular, General and Transplantation Surgery, Wroclaw Medical University, Wroclaw, Poland
| | - Tomasz Dawiskiba
- Department of Vascular, General and Transplantation Surgery, Wroclaw Medical University, Wroclaw, Poland
| | - Krzysztof Korta
- Department of Vascular, General and Transplantation Surgery, Wroclaw Medical University, Wroclaw, Poland
| | - Dagmara Baczyńska
- Department of Forensic Medicine, Molecular Techniques Unit, Wroclaw Medical University, Wroclaw, Poland
| | | | - Jerzy Garcarek
- Department of General Radiology, Interventional Radiology and Neuroradiology, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
39
|
Birzniece V. Doping in sport: effects, harm and misconceptions. Intern Med J 2015; 45:239-48. [DOI: 10.1111/imj.12629] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 10/26/2014] [Indexed: 11/28/2022]
Affiliation(s)
- V. Birzniece
- School of Medicine; University of Western Sydney; Sydney New South Wales Australia
| |
Collapse
|
40
|
Rowe GC, Raghuram S, Jang C, Nagy JA, Patten IS, Goyal A, Chan MC, Liu LX, Jiang A, Spokes KC, Beeler D, Dvorak H, Aird WC, Arany Z. PGC-1α induces SPP1 to activate macrophages and orchestrate functional angiogenesis in skeletal muscle. Circ Res 2014; 115:504-17. [PMID: 25009290 DOI: 10.1161/circresaha.115.303829] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
RATIONALE Mechanisms of angiogenesis in skeletal muscle remain poorly understood. Efforts to induce physiological angiogenesis hold promise for the treatment of diabetic microvascular disease and peripheral artery disease but are hindered by the complexity of physiological angiogenesis and by the poor angiogenic response of aged and patients with diabetes mellitus. To date, the best therapy for diabetic vascular disease remains exercise, often a challenging option for patients with leg pain. Peroxisome proliferation activator receptor-γ coactivator-1α (PGC-1α), a powerful regulator of metabolism, mediates exercise-induced angiogenesis in skeletal muscle. OBJECTIVE To test whether, and how, PGC-1α can induce functional angiogenesis in adult skeletal muscle. METHODS AND RESULTS Here, we show that muscle PGC-1α robustly induces functional angiogenesis in adult, aged, and diabetic mice. The process involves the orchestration of numerous cell types and leads to patent, nonleaky, properly organized, and functional nascent vessels. These findings contrast sharply with the disorganized vasculature elicited by induction of vascular endothelial growth factor alone. Bioinformatic analyses revealed that PGC-1α induces the secretion of secreted phosphoprotein 1 and the recruitment of macrophages. Secreted phosphoprotein 1 stimulates macrophages to secrete monocyte chemoattractant protein-1, which then activates adjacent endothelial cells, pericytes, and smooth muscle cells. In contrast, induction of PGC-1α in secreted phosphoprotein 1(-/-) mice leads to immature capillarization and blunted arteriolarization. Finally, adenoviral delivery of PGC-1α into skeletal muscle of either young or old and diabetic mice improved the recovery of blood flow in the murine hindlimb ischemia model of peripheral artery disease. CONCLUSIONS PGC-1α drives functional angiogenesis in skeletal muscle and likely recapitulates the complex physiological angiogenesis elicited by exercise.
Collapse
Affiliation(s)
- Glenn C Rowe
- From the Department of Medicine, Cardiovascular Institute (G.C.R., S.R., C.J., I.S.P., A.G., M.C.C., L.X.L., A.J., Z.A.), Center for Vascular Biology Research (J.A.N., K.C.S., D.B., H.D., W.C.A., Z.A.), and Department of Pathology (J.A.N., H.D.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - Srilatha Raghuram
- From the Department of Medicine, Cardiovascular Institute (G.C.R., S.R., C.J., I.S.P., A.G., M.C.C., L.X.L., A.J., Z.A.), Center for Vascular Biology Research (J.A.N., K.C.S., D.B., H.D., W.C.A., Z.A.), and Department of Pathology (J.A.N., H.D.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - Cholsoon Jang
- From the Department of Medicine, Cardiovascular Institute (G.C.R., S.R., C.J., I.S.P., A.G., M.C.C., L.X.L., A.J., Z.A.), Center for Vascular Biology Research (J.A.N., K.C.S., D.B., H.D., W.C.A., Z.A.), and Department of Pathology (J.A.N., H.D.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - Janice A Nagy
- From the Department of Medicine, Cardiovascular Institute (G.C.R., S.R., C.J., I.S.P., A.G., M.C.C., L.X.L., A.J., Z.A.), Center for Vascular Biology Research (J.A.N., K.C.S., D.B., H.D., W.C.A., Z.A.), and Department of Pathology (J.A.N., H.D.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - Ian S Patten
- From the Department of Medicine, Cardiovascular Institute (G.C.R., S.R., C.J., I.S.P., A.G., M.C.C., L.X.L., A.J., Z.A.), Center for Vascular Biology Research (J.A.N., K.C.S., D.B., H.D., W.C.A., Z.A.), and Department of Pathology (J.A.N., H.D.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - Amrita Goyal
- From the Department of Medicine, Cardiovascular Institute (G.C.R., S.R., C.J., I.S.P., A.G., M.C.C., L.X.L., A.J., Z.A.), Center for Vascular Biology Research (J.A.N., K.C.S., D.B., H.D., W.C.A., Z.A.), and Department of Pathology (J.A.N., H.D.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - Mun Chun Chan
- From the Department of Medicine, Cardiovascular Institute (G.C.R., S.R., C.J., I.S.P., A.G., M.C.C., L.X.L., A.J., Z.A.), Center for Vascular Biology Research (J.A.N., K.C.S., D.B., H.D., W.C.A., Z.A.), and Department of Pathology (J.A.N., H.D.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - Laura X Liu
- From the Department of Medicine, Cardiovascular Institute (G.C.R., S.R., C.J., I.S.P., A.G., M.C.C., L.X.L., A.J., Z.A.), Center for Vascular Biology Research (J.A.N., K.C.S., D.B., H.D., W.C.A., Z.A.), and Department of Pathology (J.A.N., H.D.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - Aihua Jiang
- From the Department of Medicine, Cardiovascular Institute (G.C.R., S.R., C.J., I.S.P., A.G., M.C.C., L.X.L., A.J., Z.A.), Center for Vascular Biology Research (J.A.N., K.C.S., D.B., H.D., W.C.A., Z.A.), and Department of Pathology (J.A.N., H.D.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - Katherine C Spokes
- From the Department of Medicine, Cardiovascular Institute (G.C.R., S.R., C.J., I.S.P., A.G., M.C.C., L.X.L., A.J., Z.A.), Center for Vascular Biology Research (J.A.N., K.C.S., D.B., H.D., W.C.A., Z.A.), and Department of Pathology (J.A.N., H.D.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - David Beeler
- From the Department of Medicine, Cardiovascular Institute (G.C.R., S.R., C.J., I.S.P., A.G., M.C.C., L.X.L., A.J., Z.A.), Center for Vascular Biology Research (J.A.N., K.C.S., D.B., H.D., W.C.A., Z.A.), and Department of Pathology (J.A.N., H.D.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - Harold Dvorak
- From the Department of Medicine, Cardiovascular Institute (G.C.R., S.R., C.J., I.S.P., A.G., M.C.C., L.X.L., A.J., Z.A.), Center for Vascular Biology Research (J.A.N., K.C.S., D.B., H.D., W.C.A., Z.A.), and Department of Pathology (J.A.N., H.D.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - William C Aird
- From the Department of Medicine, Cardiovascular Institute (G.C.R., S.R., C.J., I.S.P., A.G., M.C.C., L.X.L., A.J., Z.A.), Center for Vascular Biology Research (J.A.N., K.C.S., D.B., H.D., W.C.A., Z.A.), and Department of Pathology (J.A.N., H.D.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - Zolt Arany
- From the Department of Medicine, Cardiovascular Institute (G.C.R., S.R., C.J., I.S.P., A.G., M.C.C., L.X.L., A.J., Z.A.), Center for Vascular Biology Research (J.A.N., K.C.S., D.B., H.D., W.C.A., Z.A.), and Department of Pathology (J.A.N., H.D.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA.
| |
Collapse
|
41
|
Shimamura M, Nakagami H, Taniyama Y, Morishita R. Gene therapy for peripheral arterial disease. Expert Opin Biol Ther 2014; 14:1175-84. [PMID: 24766232 DOI: 10.1517/14712598.2014.912272] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Gene therapy has emerged as a novel therapy to promote angiogenesis in patients with critical limb ischemia (CLI) caused by peripheral artery disease. Researchers working in this area have focused on pro-angiogenic factors, such as VEGF, fibroblast growth factor (FGF) and hepatocyte growth factor (HGF). Based on the elaborate studies and favorable results of basic research using naked plasmid DNA (pDNA) encoding these growth factors, some clinical Phase I and Phase II trials have been performed. The results of these studies demonstrate the safety of these approaches and their potential for symptomatic improvement in CLI patients. However, the Phase III clinical trials have so far been limited to HGF gene therapy. Because one pitfall of the Phase III trials has been the limited transgene expression achieved using naked pDNA alone, the development of more efficient gene transfer systems, such as ultrasound microbubbles and the needleless injector, as well as the addition of other genes will make these novel therapies more effective and ease the symptoms of CLI. AREAS COVERED This study reviews the previously published basic research and clinical trials that have studied VEGF, FGF and HGF gene therapies for the treatment of CLI. Adjunctive therapies, such as the addition of prostacyclin synthase genes and the development of more efficient gene transfer techniques for pDNA, are also reviewed. EXPERT OPINION To date, clinical studies have demonstrated the safety of gene therapy in limb ischemia but the effectiveness of this treatment has not been determined. Larger clinical studies, as well as the development of more effective gene therapy, are needed to achieve and confirm beneficial effects.
Collapse
Affiliation(s)
- Munehisa Shimamura
- Osaka University, Kanazawa University and Hamamatsu University School of Medicine, United Graduate School of Child Development, Division of Vascular Medicine and Epigenetics, Department of Child Development , Suita , Japan
| | | | | | | |
Collapse
|
42
|
Lijkwan MA, Hellingman AA, Bos EJ, van der Bogt KEA, Huang M, Kooreman NG, de Vries MR, Peters HAB, Robbins RC, Hamming JF, Quax PHA, Wu JC. Short hairpin RNA gene silencing of prolyl hydroxylase-2 with a minicircle vector improves neovascularization of hindlimb ischemia. Hum Gene Ther 2014; 25:41-9. [PMID: 24090375 DOI: 10.1089/hum.2013.110] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
In this study, we target the hypoxia inducible factor-1 alpha (HIF-1-alpha) pathway by short hairpin RNA interference therapy targeting prolyl hydroxylase-2 (shPHD2). We use the minicircle (MC) vector technology as an alternative for conventional nonviral plasmid (PL) vectors in order to improve neovascularization after unilateral hindlimb ischemia in a murine model. Gene expression and transfection efficiency of MC and PL, both in vitro and in vivo, were assessed using bioluminescence imaging (BLI) and firefly luciferase (Luc) reporter gene. C57Bl6 mice underwent unilateral electrocoagulation of the femoral artery and gastrocnemic muscle injection with MC-shPHD2, PL-shPHD2, or phosphate-buffered saline (PBS) as control. Blood flow recovery was monitored using laser Doppler perfusion imaging, and collaterals were visualized by immunohistochemistry and angiography. MC-Luc showed a 4.6-fold higher in vitro BLI signal compared with PL-Luc. BLI signals in vivo were 4.3×10(5)±3.3×10(5) (MC-Luc) versus 0.4×10(5)±0.3×10(5) (PL-Luc) at day 28 (p=0.016). Compared with PL-shPHD2 or PBS, MC-shPHD2 significantly improved blood flow recovery, up to 50% from day 3 until day 14 after ischemia induction. MC-shPHD2 significantly increased collateral density and capillary density, as monitored by alpha-smooth muscle actin expression and CD31(+) expression, respectively. Angiography data confirmed the histological findings. Significant downregulation of PHD2 mRNA levels by MC-shPHD2 was confirmed by quantitative polymerase chain reaction. Finally, Western blot analysis confirmed significantly higher levels of HIF-1-alpha protein by MC-shPHD2, compared with PL-shPHD2 and PBS. This study provides initial evidence of a new potential therapeutic approach for peripheral artery disease. The combination of HIF-1-alpha pathway targeting by shPHD2 with the robust nonviral MC plasmid improved postischemic neovascularization, making this approach a promising potential treatment option for critical limb ischemia.
Collapse
Affiliation(s)
- Maarten A Lijkwan
- 1 Department of Medicine and Radiology, Stanford University School of Medicine , Stanford, CA 94305
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Halonen PJ, Nurro J, Kuivanen A, Ylä-Herttuala S. Current gene therapy trials for vascular diseases. Expert Opin Biol Ther 2014; 14:327-36. [PMID: 24387602 DOI: 10.1517/14712598.2014.872237] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION In the previous gene therapy trials for vascular diseases, safety of the therapies has been demonstrated with some evidence for clinical benefits. In the future, it will be important to also test the potential clinical benefits of the treatments in randomized and controlled trials with sufficient numbers of patients. AREAS COVERED This review covers 15 currently ongoing cardiovascular gene therapy trials that aim to treat coronary artery disease, heart failure and peripheral artery disease. This review summarizes current trials and their main features in the cardiovascular field. EXPERT OPINION In the gene therapy trials for vascular diseases, some limiting factors are still present. The trials have enrolled mainly elderly and severely affected patients who might not have the capacity to respond optimally to the therapies. Also, major cardiac adverse events, major amputations, mortality and other very demanding hard clinical end points have been used in relatively small patient populations. Therefore, there is an urgent need to enroll less severely affected patients and to use more informative surrogate end points in the forthcoming clinical trials.
Collapse
Affiliation(s)
- Paavo J Halonen
- University of Eastern Finland, A. I. Virtanen Institute, Department of Biotechnology and Molecular Medicine , P.O. Box 1627, FIN-70211, Kuopio , Finland +358 40 355 2076 ;
| | | | | | | |
Collapse
|
44
|
Shimamura M, Nakagami H, Koriyama H, Morishita R. Gene therapy and cell-based therapies for therapeutic angiogenesis in peripheral artery disease. BIOMED RESEARCH INTERNATIONAL 2013; 2013:186215. [PMID: 24294599 PMCID: PMC3835886 DOI: 10.1155/2013/186215] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 09/09/2013] [Indexed: 02/02/2023]
Abstract
Gene therapy and cell-based therapy have emerged as novel therapies to promote therapeutic angiogenesis in critical limb ischemia (CLI) caused by peripheral artery disease (PAD). Although researchers initially focused on gene therapy using proangiogenic factors, such as vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF), and hepatocyte growth factors (HGF), cell therapy using bone marrow mononuclear cells (BMMNCs), mesenchymal stem cells (BMMSCs), G-CSF-mobilized peripheral blood mononuclear cells (M-PBMNCs), and endothelial progenitor cells (EPCs) have also been extensively studied. Based on the elaborate studies and favorable results of basic research, some clinical phase I/II trials have been performed, and the results demonstrate the safety of these approaches and their potential for symptomatic improvement in CLI. However, the phase 3 clinical trials have thus far been limited to gene therapy using the HGF gene. Further studies using well-designed larger placebo-controlled and long-term randomized control trials (RCTs) will clarify the effectiveness of gene therapy and cell-based therapy for the treatment of CLI. Furthermore, the development of efficient gene transfer systems and effective methods for keeping transplanted cells healthy will make these novel therapies more effective and ease the symptoms of CLI.
Collapse
Affiliation(s)
- Munehisa Shimamura
- Division of Vascular Medicine and Epigenetics, Department of Child Development, United Graduate School of Child Development, Osaka University, Kanazawa University, and Hamamatsu University School of Medicine, 2-1 Yamadaoka, Suita 565-0817, Osaka, Japan
| | - Hironori Nakagami
- Division of Vascular Medicine and Epigenetics, Department of Child Development, United Graduate School of Child Development, Osaka University, Kanazawa University, and Hamamatsu University School of Medicine, 2-1 Yamadaoka, Suita 565-0817, Osaka, Japan
| | - Hiroshi Koriyama
- Division of Vascular Medicine and Epigenetics, Department of Child Development, United Graduate School of Child Development, Osaka University, Kanazawa University, and Hamamatsu University School of Medicine, 2-1 Yamadaoka, Suita 565-0817, Osaka, Japan
| | - Ryuichi Morishita
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita 565-0871, Osaka, Japan
| |
Collapse
|
45
|
Abstract
Critical limb ischaemia (CLI) is a severe form of peripheral arterial disease (PAD). CLI often causes disabling symptoms of pain and can lead to loss of the affected limb. It is also associated with increased risk of myocardial infarction, stroke and death from cardiovascular disease. The aims of management in patients with CLI are to relieve ischaemic pain, heal ulcers, prevent limb loss, improve function and quality of life and prolong survival. Here, current evidence regarding the medical management of CLI is reviewed. Cardiovascular risk factors should be assessed in all patients with CLI; smoking cessation and treatment of hypertension, hyperlipidaemia and diabetes all reduce the mortality rate in those with PAD. Antiplatelet agents (either aspirin or clopidogrel) are recommended to reduce both the incidence of cardiovascular events and risk of arterial occlusion. By contrast, routine use of anticoagulation (either warfarin or heparin) is not recommended. Treatment of the limbs themselves is often more challenging. Prostanoids may have some efficacy for treating rest pain and for ulcer healing, and iloprost shows favourable results in reducing the risk of major amputations, but long-term follow-up data regarding disease progression are lacking. There is insufficient evidence to support the use of naftidrofuryl or cilostazol, and pentoxifylline is not beneficial. Furthermore, there is no evidence of proven benefit of hyperbaric oxygen. A number of angiogenic growth factors have been studied in Phase I studies and randomized controlled trials (RCTs). They appear to be safe, but efficacy results have been mixed. Treatment with stem cells also shows some potential from early trials, but further larger RCTs are needed to demonstrate clear benefit. Thrombolysis may be an alternative for patients who develop acute limb ischaemia and are unsuitable for surgical intervention. However, newer endovascular techniques are likely to have a greater role in the future.
Collapse
Affiliation(s)
- M A Lambert
- Division of Cardiovascular and Diabetes Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | | |
Collapse
|
46
|
Ylä-Herttuala S. Cardiovascular gene therapy with vascular endothelial growth factors. Gene 2013; 525:217-9. [DOI: 10.1016/j.gene.2013.03.051] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 03/12/2013] [Indexed: 10/26/2022]
|
47
|
Wirth T, Parker N, Ylä-Herttuala S. History of gene therapy. Gene 2013; 525:162-9. [PMID: 23618815 DOI: 10.1016/j.gene.2013.03.137] [Citation(s) in RCA: 356] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Revised: 03/26/2013] [Accepted: 03/27/2013] [Indexed: 12/24/2022]
Abstract
Two decades after the initial gene therapy trials and more than 1700 approved clinical trials worldwide we not only have gained much new information and knowledge regarding gene therapy in general, but also learned to understand the concern that has persisted in society. Despite the setbacks gene therapy has faced, success stories have increasingly emerged. Examples for these are the positive recommendation for a gene therapy product (Glybera) by the EMA for approval in the European Union and the positive trials for the treatment of ADA deficiency, SCID-X1 and adrenoleukodystrophy. Nevertheless, our knowledge continues to grow and during the course of time more safety data has become available that helps us to develop better gene therapy approaches. Also, with the increased understanding of molecular medicine, we have been able to develop more specific and efficient gene transfer vectors which are now producing clinical results. In this review, we will take a historical view and highlight some of the milestones that had an important impact on the development of gene therapy. We will also discuss briefly the safety and ethical aspects of gene therapy and address some concerns that have been connected with gene therapy as an important therapeutic modality.
Collapse
Affiliation(s)
- Thomas Wirth
- A.I. Virtanen Institute, Biotechnology and Molecular Medicine Unit, Univ. of Eastern Finland, Kuopio, Finland
| | | | | |
Collapse
|
48
|
Mäkinen PI, Ylä-Herttuala S. Therapeutic gene targeting approaches for the treatment of dyslipidemias and atherosclerosis. Curr Opin Lipidol 2013; 24:116-22. [PMID: 23314926 DOI: 10.1097/mol.0b013e32835da13c] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PURPOSE OF REVIEW Despite improved therapies, cardiovascular diseases are the leading cause of morbidity and mortality worldwide. Therefore, new therapeutic approaches are still needed. In the gene therapy field, RNA interference (RNAi) and regulation of microRNAs (miRNAs) have gained a lot of attention in addition to traditional overexpression based strategies. Here, recent findings in therapeutic gene silencing and modulation of small RNA expression related to atherogenesis and dyslipidemia are summarized. RECENT FINDINGS Novel gene therapy approaches for the treatment of hyperlipidemia have been addressed. Antisense oligonucleotide and RNAi-based therapies against apolipoprotein B100 and proprotein convertase subtilisin/kexin type 9 have shown already efficacy in preclinical and clinical trials. In addition, several miRNAs dysregulated in atherosclerotic lesions and regulating cholesterol homeostasis have been found, which may represent novel targets for future therapies. SUMMARY New therapies for lowering lipid levels are now being tested in clinical trials, and both antisense oligonucleotide and RNAi-based therapies have shown promising results in lowering cholesterol levels. However, the modulation of inflammatory component in atherosclerosis by gene therapy and targeting of the effects to plaques are still difficult challenges.
Collapse
MESH Headings
- Atherosclerosis/genetics
- Atherosclerosis/therapy
- Cholesterol, HDL/genetics
- Cholesterol, HDL/metabolism
- Cholesterol, LDL/genetics
- Cholesterol, LDL/metabolism
- Clinical Trials as Topic
- Dyslipidemias/genetics
- Dyslipidemias/therapy
- Epigenesis, Genetic
- Genetic Therapy
- Humans
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Plaque, Atherosclerotic/genetics
- Plaque, Atherosclerotic/metabolism
- RNA Interference
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
Collapse
Affiliation(s)
- Petri I Mäkinen
- Department of Biotechnology and Molecular Medicine, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland
| | | |
Collapse
|
49
|
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; 6:312-22. [PMID: 23471910 PMCID: PMC3597014 DOI: 10.1242/dmm.010413] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Angiogenic therapy, which involves the use of an exogenous stimulus to promote blood vessel growth, is an attractive approach for the treatment of ischemic diseases. It has been shown in animal models that the stimulation of blood vessel growth leads to the growth of the whole vascular tree, improvement of ischemic tissue perfusion and improved muscle aerobic energy metabolism. However, very few positive results have been gained from Phase 2 and 3 clinical angiogenesis trials. Many reasons have been given for the failures of clinical trials, including poor transgene expression (in gene-therapy trials) and instability of the vessels induced by therapy. In this Review, we discuss the selection of preclinical models as one of the main reasons why clinical translation has been unsuccessful thus far. This issue has received little attention, but could have had dramatic implications on the expectations of clinical trials. We highlight crucial differences between human patients and animal models with regards to blood flow and pressure, as well as issues concerning the chronic nature of ischemic diseases in humans. We use these as examples to demonstrate why the results from preclinical trials might have overestimated the efficacy of angiogenic therapies developed to date. We also suggest ways in which currently available animal models of ischemic disease could be improved to better mimic human disease conditions, and offer advice on how to work with existing models to avoid overestimating the efficacy of new angiogenic therapies.
Collapse
Affiliation(s)
- Galina Dragneva
- Department of Biotechnology and Molecular Medicine, A. I. Virtanen Institute, University of Eastern Finland, FI-70211 Kuopio, Finland
| | | | | |
Collapse
|
50
|
Su CH, Wu YJ, Wang HH, Yeh HI. Nonviral gene therapy targeting cardiovascular system. Am J Physiol Heart Circ Physiol 2012; 303:H629-38. [PMID: 22821991 DOI: 10.1152/ajpheart.00126.2012] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The goal of gene therapy is either to introduce a therapeutic gene into or replace a defective gene in an individual's cells and tissues. Gene therapy has been urged as a potential method to induce therapeutic angiogenesis in ischemic myocardium and peripheral tissues after extensive investigation in recent preclinical and clinical studies. A successful gene therapy mainly relies on the development of the gene delivery vector. Developments in viral and nonviral vector technology including cell-based gene transfer will further improve transgene delivery and expression efficiency. Nonviral approaches as alternative gene delivery vehicles to viral vectors have received significant attention. Recently, a simple and safe approach of gene delivery into target cells using naked DNA has been improved by combining several techniques. Among the physical approaches, ultrasonic microbubble gene delivery, with its high safety profile, low costs, and repeatable applicability, can increase the permeability of cell membrane to macromolecules such as plasmid DNA by its bioeffects and can provide as a feasible tool in gene delivery. On the other hand, among the promising areas for gene therapy in acquired diseases, ischemic cardiovascular diseases have been widely studied. As a result, gene therapy using advanced technology may play an important role in this regard. The aims of this review focus on understanding the cellular and in vivo barriers in gene transfer and provide an overview of currently used chemical vectors and physical tools that are applied in nonviral cardiovascular gene transfer.
Collapse
Affiliation(s)
- Cheng-Huang Su
- Departments of Internal Medicine and Medical Research, Mackay Memorial Hospital, New Taipei City, Taiwan
| | | | | | | |
Collapse
|