Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
ESC Heart Fail ; 7(6): 3345-3354, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33094909

ABSTRACT

AIMS: This study aims to explore long-term clinical outcomes of cardiopoiesis-guided stem cell therapy for ischaemic heart failure assessed in the Congestive Heart Failure Cardiopoietic Regenerative Therapy (CHART-1) trial. METHODS AND RESULTS: CHART-1 is a multinational, randomized, and double-blind trial conducted in 39 centres in heart failure patients (n = 315) on standard-of-care therapy. The 'active' group received cardiopoietic stem cells delivered intramyocardially using a retention-enhanced catheter. The 'control' group underwent patient-level sham procedure. Patients were followed up to 104 weeks. In the entire study population, results of the primary hierarchical composite outcome were maintained neutral at Week 52 [Mann-Whitney estimator 0.52, 95% confidence interval (CI) 0.45-0.59, P = 0.51]. Landmark analyses suggested late clinical benefit in patients with significant left ventricular enlargement receiving adequate dosing. Specifically, beyond 100 days of follow-up, patients with left ventricular end-diastolic volume of 200-370 mL treated with ≤19 injections of cardiopoietic stem cells showed reduced risk of death or cardiovascular hospitalization (hazard ratio 0.38, 95% CI 0.16-0.91, P = 0.031) and cardiovascular death or heart failure hospitalization (hazard ratio 0.28, 95% CI 0.09-0.94, P = 0.040). Cardiopoietic stem cell therapy was well tolerated long term with no difference in safety readouts compared with sham at 2 years. CONCLUSIONS: Longitudinal follow-up documents that cardiopoietic stem cell therapy is overall safe, and post hoc analyses suggest benefit in an ischaemic heart failure subpopulation defined by advanced left ventricular enlargement on tolerable stem cell dosing. The long-term clinical follow-up thus offers guidance for future targeted trials.

3.
Eur Heart J ; 39(23): 2208-2216, 2018 06 14.
Article in English | MEDLINE | ID: mdl-29315376

ABSTRACT

Aims: Autologous CD34+ (auto-CD34+) cells represent an attractive option for the treatment of refractory angina. Three double-blinded randomized trials (n = 304) compared intramyocardial (IM) auto-CD34+ cells with IM placebo injections to affect total exercise time (TET), angina frequency (AF), and major adverse cardiac events (MACE). Patient-level data were pooled from the Phase I, Phase II ACT-34, ACT-34 extension, and Phase III RENEW trials to determine the efficacy and safety of auto-CD34+ cells. Methods and results: Treatment effects for TET were analysed using an analysis of covariance mixed-effects model and for AF using Poisson regression in a log linear model with repeated measures. The Kaplan-Meier rate estimates for MACE were compared using the log-rank test. Autologous CD34+ cell therapy improved TET by 46.6 s [3 months, 95% confidence interval (CI) 13.0 s-80.3 s; P = 0.007], 49.5 s (6 months, 95% CI 9.3-89.7; P = 0.016), and 44.7 s (12 months, 95% CI - 2.7 s-92.1 s; P = 0.065). The relative frequency of angina was 0.78 (95% CI 0.63-0.98; P = 0.032), 0.66 (0.48-0.91; P = 0.012), and 0.58 (0.38-0.88; P = 0.011) at 3-, 6- and 12-months in auto-CD34+ compared with placebo patients. Results remained concordant when analysed by treatment received and when confined to the Phase III dose of 1 × 105 cells/kg. Autologous CD34 + cell therapy significantly decreased mortality (12.1% vs. 2.5%; P = 0.0025) and numerically reduced MACE (38.9% vs. 30.0; P = 0.14) at 24 months. Conclusion: Treatment with auto-CD34+ cells resulted in clinically meaningful durable improvements in TET and AF at 3-, 6- and 12-months, as well as a reduction in 24-month mortality in this patient-level meta-analysis.


Subject(s)
Angina Pectoris/therapy , Exercise Tolerance , Mortality , Stem Cell Transplantation/methods , Aged , Angina Pectoris/physiopathology , Antigens, CD34/metabolism , Female , Humans , Injections, Intramuscular , Kaplan-Meier Estimate , Male , Middle Aged , Myocardium , Randomized Controlled Trials as Topic , Transplantation, Autologous
4.
Eur J Heart Fail ; 19(4): 449-456, 2017 04.
Article in English | MEDLINE | ID: mdl-28271595

ABSTRACT

Data Monitoring Committees (DMCs) play a crucial role in the conducting of clinical trials to ensure the safety of study participants and to maintain a trial's scientific integrity. Generally accepted standards exist for DMC composition and operational conduct. However, some relevant issues are not specifically addressed in current guidance documents, resulting in uncertainties regarding optimal approaches for communication between the DMC, steering committee, and sponsors, release of information, and liability protection for DMC members. The Heart Failure Association (HFA) of the European Society of Cardiology (ESC), in collaboration with the Clinical Trials Unit of the European Heart Agency (EHA) of the ESC convened a meeting of international experts in DMCs for cardiovascular and cardiometabolic clinical trials to identify specific issues and develop steps to resolve challenges faced by DMCs.The main recommendations from the meeting relate to methodological consistency, independence, managing conflicts of interest, liability protection, and training of future DMC members. This paper summarizes the key outcomes from this expert meeting, and describes the core set of activities that might be further developed and ultimately implemented by the ESC, HFA, and other interested ESC constituent bodies. The HFA will continue to work with stakeholders in cardiovascular and cardiometabolic clinical research to promote these goals.


Subject(s)
Cardiovascular Diseases , Clinical Trials Data Monitoring Committees/organization & administration , Clinical Trials as Topic/organization & administration , Metabolic Diseases , Biomedical Research , Guidelines as Topic , Humans
5.
Eur J Heart Fail ; 19(6): 718-727, 2017 06.
Article in English | MEDLINE | ID: mdl-28345190

ABSTRACT

Despite the availability of a number of different classes of therapeutic agents with proven efficacy in heart failure, the clinical course of heart failure patients is characterized by a reduction in life expectancy, a progressive decline in health-related quality of life and functional status, as well as a high risk of hospitalization. New approaches are needed to address the unmet medical needs of this patient population. The European Medicines Agency (EMA) is undertaking a revision of its Guideline on Clinical Investigation of Medicinal Products for the Treatment of Chronic Heart Failure. The draft version of the Guideline was released for public consultation in January 2016. The Cardiovascular Round Table of the European Society of Cardiology (ESC), in partnership with the Heart Failure Association of the ESC, convened a dedicated two-day workshop to discuss three main topic areas of major interest in the field and addressed in this draft EMA guideline: (i) assessment of efficacy (i.e. endpoint selection and statistical analysis); (ii) clinical trial design (i.e. issues pertaining to patient population, optimal medical therapy, run-in period); and (iii) research approaches for testing novel therapeutic principles (i.e. cell therapy). This paper summarizes the key outputs from the workshop, reviews areas of expert consensus, and identifies gaps that require further research or discussion. Collaboration between regulators, industry, clinical trialists, cardiologists, health technology assessment bodies, payers, and patient organizations is critical to address the ongoing challenge of heart failure and to ensure the development and market access of new therapeutics in a scientifically robust, practical and safe way.


Subject(s)
Cardiovascular Agents/therapeutic use , Clinical Trials as Topic , Heart Failure/drug therapy , Outcome Assessment, Health Care , Consensus , Drug Approval , Humans
6.
Eur J Heart Fail ; 18(2): 160-8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26662998

ABSTRACT

AIMS: Cardiopoiesis is a conditioning programme that aims to upgrade the cardioregenerative aptitude of patient-derived stem cells through lineage specification. Cardiopoietic stem cells tested initially for feasibility and safety exhibited signs of clinical benefit in patients with ischaemic heart failure (HF) warranting definitive evaluation. Accordingly, CHART-1 is designed as a large randomized, sham-controlled multicentre study aimed to validate cardiopoietic stem cell therapy. METHODS: Patients (n = 240) with chronic HF secondary to ischaemic heart disease, reduced LVEF (<35%), and at high risk for recurrent HF-related events, despite optimal medical therapy, will be randomized 1:1 to receive 600 × 10(6) bone marrow-derived and lineage-directed autologous cardiopoietic stem cells administered via a retention-enhanced intramyocardial injection catheter or a sham procedure. The primary efficacy endpoint is a hierarchical composite of mortality, worsening HF, Minnesota Living with Heart Failure Questionnaire score, 6 min walk test, LV end-systolic volume, and LVEF at 9 months. The secondary efficacy endpoint is the time to cardiovascular death or worsening HF at 12 months. Safety endpoints include mortality, readmissions, aborted sudden deaths, and serious adverse events at 12 and 24 months. CONCLUSION: The CHART-1 clinical trial is powered to examine the therapeutic impact of lineage-directed stem cells as a strategy to achieve cardiac regeneration in HF populations. On completion, CHART-1 will offer a definitive evaluation of the efficacy and safety of cardiopoietic stem cells in the treatment of chronic ischaemic HF. TRIAL REGISTRATION: NCT01768702.


Subject(s)
Heart Failure/therapy , Heart/physiology , Mesenchymal Stem Cell Transplantation/methods , Regeneration , Stem Cells/physiology , Adult , Chronic Disease , Epidemiologic Research Design , Heart Failure/etiology , Humans , Myocardial Ischemia/complications
7.
J Cardiovasc Transl Res ; 8(6): 381-4, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26149227

ABSTRACT

Endomyocardial delivery in the setting of active left ventricular assist device (LVAD) support has rarely been studied. The objective was to establish a protocol for endomyocardial injections during LVAD support without compromising mechanical circulation. LVAD implantation was performed in four pigs. A curved needle catheter was percutaneously inserted into the right carotid artery and positioned into the left ventricle under fluoroscopic guidance. In the setting of increasing LVAD flows (2.3-3.1 l/min), percutaneous methylene blue dye administration into the myocardium proceeded without complications in all pigs. Transection of excised hearts revealed an anterior, lateral, inferior, and septal wall distribution of methylene blue documenting injections in all four regions of the left ventricle. Ex vivo, the catheter could be maneuvered close to the LVAD inflow cannula despite augmentation of LVAD flow up to 5 l/min. Endomyocardial injections during LVAD support was found to be feasible and safe with the curved needle catheter.


Subject(s)
Cardiac Catheterization/methods , Heart Failure/physiopathology , Heart Ventricles/physiopathology , Heart-Assist Devices , Injections , Myocardium , Radiography, Interventional , Animals , Disease Models, Animal , Fluoroscopy , Methylene Blue , Swine
9.
Stem Cells ; 32(2): 572-81, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24420906

ABSTRACT

Brown adipose tissue (BAT) plays a key role in the evolutionarily conserved mechanisms underlying energy homeostasis in mammals. It is characterized by fat vacuoles 5-10 µm in diameter and expression of uncoupling protein one, central to the regulation of thermogenesis. In the human newborn, BAT depots are typically grouped around the vasculature and solid organs. These depots maintain body temperature during cold exposure by warming the blood before its distribution to the periphery. They also ensure an optimal temperature for biochemical reactions within solid organs. BAT had been thought to involute throughout childhood and adolescence. Recent studies, however, have confirmed the presence of active BAT in adult humans with depots residing in cervical, supraclavicular, mediastinal, paravertebral, and suprarenal regions. While human pluripotent stem cells have been differentiated into functional brown adipocytes in vitro and brown adipocyte progenitor cells have been identified in murine skeletal muscle and white adipose tissue, multipotent metabolically active BAT-derived stem cells from a single depot have not been identified in adult humans to date. Here, we demonstrate a clonogenic population of metabolically active BAT stem cells residing in adult humans that can: (a) be expanded in vitro; (b) exhibit multilineage differentiation potential; and (c) functionally differentiate into metabolically active brown adipocytes. Our study defines a new target stem cell population that can be activated to restore energy homeostasis in vivo for the treatment of obesity and related metabolic disorders.


Subject(s)
Adipose Tissue, Brown/cytology , Cell Differentiation/genetics , Muscle, Skeletal/metabolism , Pluripotent Stem Cells/metabolism , Adipose Tissue, White/cytology , Animals , Biological Evolution , Energy Metabolism/genetics , Humans , Mice , Muscle, Skeletal/cytology , Obesity/genetics , Obesity/metabolism , Obesity/pathology , Pluripotent Stem Cells/cytology , Thermogenesis
10.
Am Heart J ; 165(6): 854-861.e2, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23708155

ABSTRACT

Preclinical trials indicate that CD34+ cells represent an effective angiogenic stem cell component. Early-phase clinical trials suggest that intramyocardial administration of autologous CD34+ cells may improve functional capacity and symptoms of angina. RENEW is a pivotal phase 3 trial designed to determine the efficacy of granulocyte colony-stimulating factor (G-CSF)-mobilized CD34+ stem cells for the treatment for patients with refractory angina and chronic myocardial ischemia. Patients (n = 444) receiving maximally tolerated antianginal therapies and lacking conventional revascularization options with Canadian Cardiovascular Society class III or IV angina and ischemia on stress testing will be randomized 2:1:1 to cell therapy (G-CSF-mediated stem cell mobilization, apheresis, and intramyocardial injection of 1 × 10(5) autologous CD34(+) cells/kg), active control (G-CSF-mediated stem cell mobilization, apheresis, and intramyocardial placebo injection), or open-label standard of care. The primary efficacy end point is change in exercise treadmill time in the treated vs active control patients, with 90% power to detect a 60-second difference in exercise time between cell-treated (n = 200) and active control (n = 100) patients. Key secondary end points include total number of anginal episodes per week and the incidence of independently adjudicated major adverse cardiac events and serious adverse events. RENEW will be the first adequately powered study aimed at definitively determining the efficacy of a cell therapy (intramyocardially delivered autologous CD34+ cells) for improvement of functional capacity in patients with refractory angina.


Subject(s)
Angina, Stable/surgery , Antigens, CD34/immunology , Stem Cell Transplantation/methods , Stem Cells/immunology , Adult , Aged , Aged, 80 and over , Angina, Stable/diagnosis , Angina, Stable/immunology , Double-Blind Method , Electrocardiography , Exercise Test , Female , Follow-Up Studies , Humans , Injections , Male , Middle Aged , Myocardium , Prospective Studies , Transplantation, Autologous , Treatment Outcome , Young Adult
11.
Circ Res ; 112(5): 816-25, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23429605

ABSTRACT

RATIONALE: Preclinical studies indicate that adult stem cells induce tissue repair by activating endogenous stem cells through the stromal cell-derived factor-1:chemokine receptor type 4 axis. JVS-100 is a DNA plasmid encoding human stromal cell-derived factor-1. OBJECTIVE: We tested in a phase 1, open-label, dose-escalation study with 12 months of follow-up in subjects with ischemic cardiomyopathy to see if JVS-100 improves clinical parameters. METHODS AND RESULTS: Seventeen subjects with ischemic cardiomyopathy, New York Heart Association class III heart failure, with an ejection fraction ≤40% on stable medical therapy, were enrolled to receive 5, 15, or 30 mg of JVS-100 via endomyocardial injection. The primary end points for safety and efficacy were at 1 and 4 months, respectively. The primary safety end point was a major adverse cardiac event. Efficacy end points were change in quality of life, New York Heart Association class, 6-minute walk distance, single photon emission computed tomography, N-terminal pro-brain natruretic peptide, and echocardiography at 4 and 12 months. The primary safety end point was met. At 4 months, all of the cohorts demonstrated improvements in 6-minute walk distance, quality of life, and New York Heart Association class. Subjects in the 15- and 30-mg dose groups exhibited improvements in 6-minute walk distance (15 mg: median [range]: 41 minutes [3-61 minutes]; 30 mg: 31 minutes [22-74 minutes]) and quality of life (15 mg: -16 points [+1 to -32 points]; 30 mg: -24 points [+17 to -38 points]) over baseline. At 12 months, improvements in symptoms were maintained. CONCLUSIONS: These data highlight the importance of defining the molecular mechanisms of stem cell-based tissue repair and suggest that overexpression of stromal cell-derived factor-1 via gene therapy is a strategy for improving heart failure symptoms in patients with ischemic cardiomyopathy.


Subject(s)
Chemokine CXCL12/genetics , Genetic Therapy/adverse effects , Genetic Therapy/methods , Heart Failure/therapy , Plasmids , Aged , Chemokine CXCL12/metabolism , Cohort Studies , Dose-Response Relationship, Drug , Echocardiography , Exercise Tolerance , Female , Follow-Up Studies , Heart Failure/metabolism , Heart Failure/pathology , Humans , Male , Middle Aged , Myocardium/metabolism , Myocardium/pathology , Natriuretic Peptide, Brain/blood , Peptide Fragments/blood , Positron-Emission Tomography , Quality of Life , Treatment Outcome
12.
Cell Transplant ; 22(12): 2299-309, 2013.
Article in English | MEDLINE | ID: mdl-23107489

ABSTRACT

The administration of bone marrow-derived stem cells may provide a new treatment option for patients with heart failure. Transcatheter cell injection may require multi-imaging modalities to optimize delivery. This study sought to evaluate whether endomyocardial injection of mesenchymal precursor cells (MPCs) could be guided by real-time 3D echocardiography (RT3DE) in treating chronic, postinfarction (MI) left ventricular (LV) dysfunction in sheep. Four weeks after induction of an anterior wall myocardial infarction in 39 sheep, allogeneic MPCs in doses of either 25 × 10(6) (n = 10), 75 × 10(6) (n = 9), or 225 × 10(6) (n = 10) cells or nonconditioned control media (n = 10) were administered intramyocardially into infarct and border zone areas using a catheter designed for combined fluoroscopic and RT3DE-guided injections. LV function was assessed before and after injection. Infarct dimension and vascular density were evaluated histologically. RT3DE-guided injection procedures were safe. Compared to controls, the highest dose MPC treatment led to increments in ejection fraction (3 ventricula 3% in 225M MPCs vs. -5 ± 4% in the control group, p < 0.01) and wall thickening in both infarct (4 ± 4% in 225M MPCs vs. -3 ± 6% in the control group, p = 0.02) and border zones (4 ± 6% in 225M MPCs vs. -8 ± 9% in the control group, p = 0.01). Histology analysis demonstrated significantly higher arteriole density in the infarct and border zones in the highest dose MPC-treated animals compared to the lower dose or control groups. Endomyocardial implantation of MPCs under RT3DE guidance was safe and without observed logistical obstacles. Significant increases in LV performance (ejection fraction and wall thickening) and neovascularization resulted from this technique, and so this technique has important implications for treating patients with postischemic LV dysfunction.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Myocardial Infarction/surgery , Acute Disease , Animals , Cardiac Catheterization , Chronic Disease , Coronary Vessels/pathology , Disease Models, Animal , Echocardiography, Three-Dimensional , Fluoroscopy , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/pathology , Myocardium/pathology , Sheep , Ventricular Dysfunction, Left/physiopathology
13.
Curr Opin Organ Transplant ; 17(6): 675-80, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23111647

ABSTRACT

PURPOSE OF REVIEW: Stem cell therapy for the treatment of cardiovascular disease is rapidly moving from bench to bedside. In the settings of acute and chronic myocardial injury, approaches to treatment have explored various cell populations, delivery methods, and times of administration. RECENT FINDINGS: Although initial studies in patients were performed with unfractionated bone marrow cells, further investigations in animal models of myocardial disease have elucidated mechanisms of benefit and opened doors to treatment strategies with stem cells of varied derivation. SUMMARY: Allogeneic stem cell populations have demonstrated therapeutic promise in ischemic heart disease, without a requirement for immunosuppressive drugs, and offer the potential to create large-scale, cryopreserved banks of cells for 'off the shelf' utility.


Subject(s)
Bone Marrow Transplantation , Mesenchymal Stem Cell Transplantation , Myocardial Ischemia/surgery , Animals , Bone Marrow Cells , Clinical Trials as Topic , Disease Models, Animal , Humans , Myocardial Ischemia/physiopathology , Stem Cell Transplantation/methods , Transplantation, Autologous , Transplantation, Homologous , Treatment Outcome
14.
Circ Res ; 110(2): 304-11, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-22052917

ABSTRACT

RATIONALE: MultiStem is an allogeneic bone marrow-derived adherent adult stem cell product that has shown efficacy in preclinical models of acute myocardial infarction (AMI). In this phase I clinical trial in patients with first ST-elevation-myocardial infarction (STEMI), we combine first-in-man delivery of MultiStem with a first-in-coronary adventitial delivery system to determine the effects of this system on left ventricular function at 4 months after AMI. OBJECTIVE: Test the effects of adventitial delivery of Multistem in the peri-infarct period in patients with first STEMI. METHODS AND RESULTS: This study was a phase I, open-label, dose-escalating registry control group study. Nineteen patients received MultiStem (20 million, n=6; 50 million, n=7; or 100 million, n=6) and 6 subjects were assigned to the registry control group. Two to 5 days after AMI, we delivered MultiStem to the adventitia of the infarct-related vessel in patients with first-time STEMI. All patients underwent primary percutaneous coronary intervention with resulting Thrombolysis In Myocardial Infarction grade 3 flow and with ejection fraction (EF) ≤45% as determined by echocardiogram or left ventriculogram within 12 hours of primary percutaneous coronary intervention. The cell product (20 million, 50 million, or 100 million) was well tolerated, and no serious adverse events were deemed related to MultiStem. There was no increase in creatine kinase-MB or troponin associated with the adventitial delivery of MultiStem. In patients with EF determined to be ≤45% by a core laboratory within 24 hours before the MultiStem injection, we observed a 0.9 (n=4), 3.9 (n=4), 13.5 (n=5), and 10.9 (n=2) percent absolute increases in EF in the registry, 20 million, 50 million, and 100 million dose groups, respectively. The increases in EF in the 50 million and 100 million groups were accompanied by 25.4 and 8.4 mL increases in left ventricular stroke volume. CONCLUSIONS: In this study, the delivery of MultiStem to the myocardium in patients with recent STEMI was well tolerated and safe. In patients who exhibited significant myocardial damage, the delivery of ≥50 million MultiStem resulted in improved EF and stroke volume 4 months later. These findings support further development of MultiStem in patients with AMI and they validate the potential of a system for delivery of adult stem cells at any time after primary percutaneous coronary intervention.


Subject(s)
Adult Stem Cells/transplantation , Angioplasty, Balloon, Coronary , Bone Marrow Transplantation , Myocardial Infarction/therapy , Adult Stem Cells/immunology , Aged , Bone Marrow Transplantation/adverse effects , Combined Modality Therapy , Connective Tissue , Female , Humans , Male , Microinjections , Middle Aged , Myocardial Infarction/diagnosis , Myocardial Infarction/physiopathology , Myocardial Infarction/surgery , Myocardium/pathology , Recovery of Function , Registries , Stroke Volume , Time Factors , Transplantation, Homologous , Treatment Outcome , United States , Ventricular Function, Left
15.
Am Heart J ; 162(4): 654-662.e1, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21982657

ABSTRACT

BACKGROUND: We sought to determine the safety and preliminary efficacy of transcatheter intramyocardial administration of myoblasts in patients with heart failure (HF). METHODS: MARVEL is a randomized placebo-controlled trial of image-guided, catheter-based intramyocardial injection of placebo or myoblasts (400 or 800 million) in patients with class II to IV HF and ejection fraction <35%. Primary end points were frequency of serious adverse events (safety) and changes in 6-minute walk test and Minnesota Living With HF score (efficacy). Of 330 patients intended for enrollment, 23 were randomized (MARVEL-1) before stopping the study for financial reasons. RESULTS: At 6 months, similar numbers of events occurred in each group: 8 (placebo), 7 (low dose), and 8 (high dose), without deaths. Ventricular tachycardia responsive to amiodarone was more frequent in myoblast-treated patients: 1 (placebo), 3 (low dose), and 4 (high dose). A trend toward improvement in functional capacity was noted in myoblast-treated groups (Δ6-minute walk test of -3.6 vs +95.6 vs +85.5 m [placebo vs low dose vs high dose; P = .50]) without significant changes in Minnesota Living With HF scores. CONCLUSIONS: In HF patients with chronic postinfarction cardiomyopathy, transcatheter administration of myoblasts in doses of 400 to 800 million cells is feasible and may lead to important clinical benefits. Ventricular tachycardia may be provoked by myoblast injection but appears to be a transient and treatable problem. A large-scale outcome trial of myoblast administration in HF patients with postinfarction cardiomyopathy is feasible and warranted.


Subject(s)
Heart Failure/surgery , Myoblasts, Skeletal/transplantation , Adult , Aged , Cardiac Catheterization , Cardiac Surgical Procedures/adverse effects , Cardiac Surgical Procedures/methods , Chronic Disease , Double-Blind Method , Female , Heart Failure/etiology , Humans , Male , Middle Aged , Myocardial Infarction/complications
17.
Heart Rhythm ; 7(7): 964-70, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20346417

ABSTRACT

BACKGROUND: Left ventricular pacing (LVP) to induce cardiac memory (CM) in dogs results in a decreased transient outward K current (I(to)) and reduced mRNA and protein of the I(to) channel accessory subunit, KChIP2. The KChIP2 decrease is attributed to a decrease in its transcription factor, cyclic adenosine monophosphate response element binding protein (CREB). OBJECTIVE: This study sought to determine the mechanisms responsible for the CREB decrease that is initiated by LVP. METHODS: CM was quantified as T-wave vector displacement in 18 LVP dogs. In 5 dogs, angiotensin II receptor blocker, saralasin, was infused before and during pacing. In 3 dogs, proteasomal inhibitor, lactacystin, was injected into the left anterior descending artery before LVP. Epicardial biopsy samples were taken before and after LVP. Neonatal rat cardiomyocytes (NRCM) were incubated with H(2)O(2) (50 micromol/l) for 1 hour with or without lactacystin. RESULTS: LVP significantly displaced the T-wave vector and was associated with increased lipid peroxidation and increased tissue angiotensin II levels. Saralasin prevented T-vector displacement and lipid peroxidation. CREB was significantly decreased after 2 hours of LVP and was comparably decreased in H(2)O(2)-treated NRCM. Lactacystin inhibited the CREB decrease in LVP dogs and H(2)O(2)-treated NRCM. LVP and H(2)O(2) both induced CREB ubiquitination, and the H(2)O(2)-induced CREB decrease was prevented by knocking down ubiquitin. CONCLUSION: LVP initiates myocardial angiotensin II production and reactive oxygen species synthesis, leading to CREB ubiquitination and its proteasomal degradation. This sequence of events would explain the pacing-induced reduction in KChIP2, and contribute to altered repolarization and the T-wave changes of cardiac memory.


Subject(s)
Cardiac Pacing, Artificial , Cyclic AMP Response Element-Binding Protein/metabolism , Heart Conduction System/metabolism , Kv Channel-Interacting Proteins/analysis , Ventricular Function, Left/physiology , Action Potentials/physiology , Angiotensin II/physiology , Animals , Arrhythmias, Cardiac/metabolism , Blotting, Western , Cells, Cultured , Dogs , Ion Channels/physiology , Lipid Peroxidation , Male , Models, Animal , Models, Cardiovascular , Myocardium/metabolism , Myocytes, Cardiac/cytology , Oxidative Stress/physiology , Proteasome Endopeptidase Complex/metabolism , Reactive Oxygen Species/metabolism , Ubiquitin/physiology , Ubiquitination , Ventricular Remodeling/physiology
18.
Catheter Cardiovasc Interv ; 75(5): 708-12, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20049957

ABSTRACT

OBJECTIVES: To evaluate the outcome of patients with coronary perforations who were treated with the dual catheter approach. BACKGROUND: Coronary artery perforation is a grave complication of percutaneous coronary intervention (PCI) with high mortality and morbidity. Treating a coronary artery perforation with two catheters through dual access enables a rapid delivery of covered stent or coils to the vessel, without losing control of the perforation site. METHODS: We retrospectively reviewed all patients who had a severe coronary perforation during a PCI in our center, and compared outcomes of patients treated with the dual versus the traditional single guiding catheter approach. RESULTS: Between April 2004 and October 2008, 13,466 PCI's were performed in Columbia University - New York Presbyterian Medical Center. There were 33 documented cases of coronary perforations during that period of time (0.245%), among these, 26 were angiographically severe (Ellis type 2 or 3 perforations). Eleven patients were treated acutely with a dual catheter technique whereas the other fifteen patients were treated using a single guiding catheter. In the dual catheter group one patient expired after emergent CABG (9.1%), and four patients underwent emergent paricardiocentesis (36.4%). In patients treated with single catheter, there were three deaths (20%), two surgical explorations (13.3%), eight emergent pericardiocenthesis (53.3%), and one event of severe anoxic brain damage (6.7%). CONCLUSION: The dual catheter technique is a relatively safe and reproducible approach to treat a PCI induced severe coronary artery perforation, and may improve outcome compared to historical series.


Subject(s)
Angioplasty, Balloon, Coronary/adverse effects , Balloon Occlusion/methods , Cardiac Catheterization/methods , Coronary Vessels/injuries , Iatrogenic Disease , Wounds, Penetrating/therapy , Academic Medical Centers , Aged , Balloon Occlusion/adverse effects , Balloon Occlusion/instrumentation , Cardiac Catheterization/adverse effects , Cardiac Catheterization/instrumentation , Coronary Angiography , Female , Humans , Male , New York City , Retrospective Studies , Rupture , Severity of Illness Index , Stents , Treatment Outcome , Wounds, Penetrating/diagnostic imaging , Wounds, Penetrating/etiology
19.
J Am Coll Cardiol ; 54(24): 2277-86, 2009 Dec 08.
Article in English | MEDLINE | ID: mdl-19958962

ABSTRACT

OBJECTIVES: Our aim was to investigate the safety and efficacy of intravenous allogeneic human mesenchymal stem cells (hMSCs) in patients with myocardial infarction (MI). BACKGROUND: Bone marrow-derived hMSCs may ameliorate consequences of MI, and have the advantages of preparation ease, allogeneic use due to immunoprivilege, capacity to home to injured tissue, and extensive pre-clinical support. METHODS: We performed a double-blind, placebo-controlled, dose-ranging (0.5, 1.6, and 5 million cells/kg) safety trial of intravenous allogeneic hMSCs (Prochymal, Osiris Therapeutics, Inc., Baltimore, Maryland) in reperfused MI patients (n=53). The primary end point was incidence of treatment-emergent adverse events within 6 months. Ejection fraction and left ventricular volumes determined by echocardiography and magnetic resonance imaging were exploratory efficacy end points. RESULTS: Adverse event rates were similar between the hMSC-treated (5.3 per patient) and placebo-treated (7.0 per patient) groups, and renal, hepatic, and hematologic laboratory indexes were not different. Ambulatory electrocardiogram monitoring demonstrated reduced ventricular tachycardia episodes (p=0.025), and pulmonary function testing demonstrated improved forced expiratory volume in 1 s (p=0.003) in the hMSC-treated patients. Global symptom score in all patients (p=0.027) and ejection fraction in the important subset of anterior MI patients were both significantly better in hMSCs versus placebo subjects. In the cardiac magnetic resonance imaging substudy, hMSC treatment, but not placebo, increased left ventricular ejection fraction and led to reverse remodeling. CONCLUSIONS: Intravenous allogeneic hMSCs are safe in patients after acute MI. This trial provides pivotal safety and provisional efficacy data for an allogeneic bone marrow-derived stem cell in post-infarction patients. (Safety Study of Adult Mesenchymal Stem Cells [MSC] to Treat Acute Myocardial Infarction; NCT00114452).


Subject(s)
Mesenchymal Stem Cell Transplantation , Myocardial Infarction/surgery , Double-Blind Method , Female , Humans , Infusions, Intravenous , Male , Mesenchymal Stem Cell Transplantation/methods , Middle Aged
20.
Cell Transplant ; 18(10): 1111-21, 2009.
Article in English | MEDLINE | ID: mdl-19650972

ABSTRACT

Human mesenchymal stem cells (hMSC) are being administered by direct intramyocardial (IM) injection into patients with myocardial dysfunction with an objective to improve clinical status. However, surprisingly little attention has been directed to qualifying hMSC functionality beyond simple viability. In particular, the transit of hMSCs through a small-caliber needle lumen, the final fluidic pathway for all IM injection devices, may be especially prone to inducing unwarranted effects on cell function. This study evaluated the changes in clonogenicity, gene expression, and cytokine secretion that may be induced in hMSC (20 million/ml) by injection through a 26-gauge Nitinol needle at two different flow rates compared to noninjected control samples. Results indicated that hMSC viability and colony forming unit (CFU) formation was not altered by changes in injection rate, although a trend toward lower titers was noted at the higher flow rate, for the specific batch of hMSCs studied. The gene expression and cytokine analysis data suggest that delivering a suspension of MSCs through narrow lumen needles may marginally alter certain gene expression programs, but that such in vitro effects are transient and not translated into measurable differences in protein production. Gene expression levels of four cytokines (bFGF, SDF-1, SCF, VEGF) were significantly different at 400 microl/min, and that of all cytokines were significantly different at 1600 microl/min when compared to controls (p < 0.05). These changes were less pronounced (statistically insignificant for most cases, p > 0.05) and, in certain instances directionally opposite, at 72 h. However, no differences in the amounts of secreted bFGF, VEGF, or TGF-beta were detectable at either of the two time points or flow rates. We infer that intramyocardial administration by transcatheter techniques is unlikely to interfere with the machinery required for cell replication or secretion of regulatory and other growth factors, which are the mainstays of MSC contribution to cardiac tissue repair and regeneration.


Subject(s)
Bone Marrow Cells/cytology , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Catheterization , Cell Survival , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Compressive Strength , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Humans , Injections , Mesenchymal Stem Cells/metabolism , Shear Strength , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...