Abstract
Rationale
Autologous bone marrow (BM) mesenchymal stem cells (MSCs) and c-kit+ cardiac progenitor cells (CPCs) are two promising cell types being evaluated for patients with heart failure (HF) secondary to ischemic cardiomyopathy. No information is available in humans regarding the relative efficacy of MSCs and CPCs and whether their combination is more efficacious than either cell type alone.
Objective
CONCERT-HF (Combination Of meseNchymal and c-kit+ Cardiac stEm cells as Regenerative Therapy for Heart Failure) is a Phase II trial aimed at elucidating these issues by assessing the feasibility, safety, and efficacy of transendocardial administration of autologous MSCs and CPCs, alone and in combination, in patients with HF caused by chronic ischemic cardiomyopathy (coronary artery disease and old myocardial infarction).
Methods and Results
Using a randomized, double-blinded, placebo-controlled, multi-center, multi-treatment, and adaptive design, CONCERT-HF examines whether administration of MSCs alone, CPCs alone, or MSCs + CPCs in this population alleviates left ventricular (LV) remodeling and dysfunction, reduces scar size, improves quality of life, or augments functional capacity. The four-arm design enables comparisons of MSCs alone with CPCs alone and with their combination. CONCERT-HF consists of 162 patients, 18 in a safety lead-in phase (Stage 1) and 144 in the main trial (Stage 2). Stage 1 is complete and Stage 2 is currently randomizing patients from seven centers across the US.
Conclusions
CONCERT-HF will provide important insights into the potential therapeutic utility of MSCs and CPCs, given alone and in combination, for patients with HF secondary to ischemic cardiomyopathy.
Keywords: Cell therapy, trial design, mesenchymal stem cells, c-kit+ cardiac progenitor cells, heart failure
Subject Terms: Chronic Ischemic Heart Disease, Clinical Trials, Stem Cells
INTRODUCTION
Cell therapy has recently emerged as a potential novel approach to the treatment of heart failure (HF) caused by chronic ischemic cardiomyopathy (coronary artery disease and old myocardial infarction), hereby referred to as “ischemic HF”. Many studies have explored the use of various types of stem or progenitor cells in patients with chronic ischemic HF1, with encouraging results1–4. However, since the studies performed to date have been relatively small, definitive demonstration of therapeutic efficacy awaits the results of large, well-designed Phase III trials.
Two of the most promising types of cells being considered for patients with ischemic HF are bone marrow (BM) mesenchymal stem cells (MSCs)4 and c-kit+ cardiac progenitor cells (CPCs)1, 5. No information is available regarding the relative efficacy of MSCs and CPCs in humans. MSCs have been shown to ameliorate post-myocardial infarction (MI) left ventricular (LV) dysfunction and reduce scar size in preclinical models of ischemic cardiomyopathy (ICM), including rodents6 and pigs7–14. Several clinical trials also have suggested that MSCs are safe and therapeutically beneficial in patients with chronic ischemic HF4, 15–23. Similarly, CPCs have been found to attenuate LV dysfunction and remodeling in many preclinical studies from several independent laboratories, both in rodent and in porcine models of chronic ICM24–29. A Phase I trial in patients with ischemic HF has provided evidence for the safety and feasibility of administering autologous CPCs after bypass surgery and has suggested benefit, although the study was not designed to establish therapeutic efficacy30, 31.
Another important question is whether the combination of MSCs and CPCs is more efficacious than either cell type alone. A beneficial interaction between these cells types is suggested by preclinical evidence that transplantation of MSCs promotes proliferation of endogenous CPCs, suggesting that this may be an important mechanism for the salutary effects of MSC administration7. Indeed, when MSCs and CPCs were combined in preclinical models, the therapeutic effects were additive, and thus superior to those of either cell type alone24, 25. The concept of combinatorial stem cell therapy is conceptually attractive but has never been tested in humans.
CONCERT-HF (Combination Of meseNchymal and c-kit+ Cardiac stEm cells as Regenerative Therapy for Heart Failure) (clinicaltrials.gov Identifier: NCT02501811) is a Phase II trial aimed at addressing the aforementioned issues by assessing the feasibility, safety, and efficacy of MSCs and CPCs, alone and in combination, in patients with ischemic HF. Using a randomized, double-blinded, placebo-controlled, multi-center, adaptive design, CONCERT-HF addresses several important questions: Is combined treatment with autologous MSCs and CPCs feasible and safe in patients with ischemic HF? Do MSCs and CPCs, given alone or in combination, alleviate LV dysfunction, reduce scar size, improve quality of life, and/or augment functional capacity in this population? Is either cell type more effective than the other? Is the combination of MSCs and CPCs superior to MSCs alone or CPCs alone in terms of therapeutic efficacy?
METHODS
Study population
Upon completion of the CONCERT-HF trial, results will be publicly available at www.clinicaltrials.gov and data will be available by request to the corresponding author of the final results paper. Inclusion and exclusion criteria are summarized in Tables 1 and 2, respectively. Candidates for enrollment include men and women, aged 21 to 79, with HF of ischemic etiology. Participants must have documented coronary artery disease, evidence of myocardial scar involving ≥5% of the LV mass by magnetic resonance imaging (MRI), LV systolic dysfunction (LV ejection fraction [LVEF] ≤40% by MRI), and symptoms of HF (NYHA class II-III), and must be receiving guideline-driven medical therapy (e.g., beta blockers, ACE inhibitors/ARBs/ARNIs, and/or aldosterone antagonists) at stable and tolerated doses for ≥1 month before consent. At least 3 months must have elapsed from a percutaneous coronary intervention or implantation of a CTR device.
Table 1.
Inclusion Criteria
To participate, a patient MUST:
|
Table 2.
Exclusion Criteria
To participate, a patient MUST NOT HAVE:
|
Study design
The primary objectives are to assess: i) whether autologous MSCs and CPCs, alone or in combination, can be manufactured and delivered to patients with ischemic HF (feasibility); ii) whether the administration of MSCs and CPCs, alone or in combination, is well-tolerated (safety); and iii) whether MSCs and CPCs, delivered alone or in combination, improve LV function, quality of life, and functional capacity, and/or reduce scar size (efficacy).
The planned total sample size is 160 patients, enrolled in two stages. In Stage 1 (open label, lead-in study) 16 patients (expanded to 18) were randomized 1:1 to either a standard-of-care control group (i.e., they did not undergo harvest, mapping, or injection procedures) or combination therapy (MSCs + CPCs, as described below for Stage 2). Stage 1 participants were followed for three months to complete safety and functional assessments. After review of the three-month data by an NHLBI Data and Safety Monitoring Board (DSMB), approval was granted to proceed with Stage 2 (vide infra). Patients randomized to MSCs + CPCs in Stage 1 were followed for 12 months for safety. Those randomized to the standard-of-care control group had the option to be evaluated for enrollment in Stage 2.
Stage 2 is a Phase II, double-blind, randomized, placebo-controlled trial designed to evaluate the feasibility, safety, and efficacy of MSCs alone, CPCs alone, and their combination compared with placebo as well as each other in patients with ischemic HF (Figure 1). Using an adaptive design, Stage 2 patients (n=144) are randomized (1:1:1:1) to one of four treatments: placebo (cell-free PlasmaLyte-A), autologous MSCs (“MSCs alone”; target dose, 150 × 106 cells), autologous CPCs (“CPCs alone”; target dose, 5 × 106 cells), or a combination of autologous MSCs and CPCs (“MSCs + CPCs;” target doses, 150 × 106 MSCs plus 5 × 106 CSCs).
Figure 1.
Protocol Overview for Stage 2.
Baseline testing, randomization, and tissue harvesting
Within 60 days of written informed consent, patients undergo baseline testing (Table 3). Any eligible patients are randomized via an online database created and maintained by the Data Coordinating Center (DCC) and undergo, on the same day, bone marrow aspiration (BMA) and right heart catheterization (RHC) (all groups) and right ventricular endocardial biopsy (EMB) (only in the CPCs alone and MSCs + CPCs groups).
Table 3.
Baseline Evaluation
|
NT-Pro BNP= N-Terminal pro-Brain Natriuretic Peptide; HbA1c= Glycated Hemoglobin (A1c); PT/INR/PTT= Prothrombin Time/International Normalized Ratio/ Partial Thromboplastin Time; HIV=Human Immunodeficiency Virus; HBsAg=Hepatitis B surface antigen; Anti-HBs=Hepatitis B surface antibody; Anti-HBc=Hepatitis B core antibody; Anti-HCV= Hepatitis C Virus
A total of ~90 ml of BM is harvested from the posterior iliac crest. Upon arrival at the local cell processing laboratory, BM samples undergo quality control (QC) tests including sterility (aerobic, anaerobic, and fungal cultures), nucleated cell count, and viability (Trypan Blue). Fresh BM samples are shipped to both the Central Cell Manufacturing Facility (CCMF) for production of MSCs and, with appropriate consent, to the CCTRN Biorepository (BRC)32.
In patients assigned to the CPCs alone and MSCs + CPCs groups, EMB is performed during RHC. Up to six endomyocardial samples are obtained to achieve approximately 20 mg total tissue. Fresh EMB samples are shipped to the CCMF for isolation and expansion of CPCs. Two transthoracic 2-D echoes without contrast are done on the day of harvest for all study patients: 1) a pre-RHC procedure echo to assess for pre-existing pericardial effusion and 2) a post-RHC procedure performed within 6 hours post-procedure. To minimize the risk of serious adverse events, RHC procedures are canceled or terminated if the patient’s NYHA class deteriorates to class IV, if the right ventricular or pulmonary artery systolic pressure is ≥60 mmHg, or if the pulmonary capillary wedge pressure is ≥35 mmHg. Furthermore, the RHC procedure is temporarily halted and the suitability of the patient for continuation re-evaluated if systemic blood pressure is <80 mmHg (change from baseline), heart rate is >100 beats/min (change from baseline), either right ventricular or pulmonary artery systolic pressures are 50-59 mmHg, or pulmonary capillary wedge pressure is 30-34 mmHg.
Cell manufacturing
MSCs and CPCs are manufactured by the CCMF at the University of Miami Miller School of Medicine’s Interdisciplinary Stem Cell Institute.
MSCs are manufactured using each patient’s own BM. The mononuclear cells (MNCs) are isolated using a density gradient with Lymphocyte Separation Medium. The low-density cells are collected from the gradient and plated for further expansion. After 14 days of culture, passage zero (P0) cells are harvested by enzymatic treatment and expanded into 60 flasks. These flasks are incubated for a week and the MSCs are harvested again (P1 cells). The P1 cells are then washed and total viable cell counts are determined for cell dosing. Samples of MSC products are characterized to assure that they meet predetermined specifications with regard to cell count, phenotype, potency, mycoplasma testing, and other QC tests (Table 4). MNCs of participants randomized to CPCs alone or to placebo are cryopreserved and shipped to the BRC. Media from the final MSC passage prior to product cryopreservation is likewise cryopreserved and sent to BRC.
Table 4.
Quality Control Testing of Cell Product Prior to Cryopreservation at the CCMF
Assay | Test Method | Specification |
---|---|---|
Mycoplasma PCR | VenorGeM® (Cells in Conditioned Medium prior to cryopreservation) | Negative |
Viability | Trypan Blue | ≥70% |
Aerobic, Anaerobic, and Fungal | 14 day Bactec/BacT/ALERT assay or equivalent | No Growth |
MSC Product only: | ||
CFU-F | Colony Formation, 14 Days | Growth |
Phenotype/Cell Characterization CD 105+ CD45+ |
Flow Cytometry | CD105+ >80% CD45+ <2% |
Cell Count | Hemocytometer | >75×106 |
CPC Product only: | ||
Phenotype/Cell Characterization CD 117+ CD45+ |
Flow Cytometry | CD117+ ≥70% CD45+<2% |
Cell Count | Manual | >0.8 ×106 |
PCR: polymerase chain reaction; CFU-F: colony forming unit fibroblasts.
In patients randomized to the CPCs alone or the MSCs + CPCs groups, CPCs are manufactured using each patient’s own EMB samples. The methods for culture and subsequent production of CPCs are a modified version of the protocol previously used for SCIPIO30. Briefly, EMBs are enzymatically digested with warm Collagenase Type II (Gibco # 17101) solution (final concentration 329 U/ml diluted in Ham’s F12) at 37°C. The digested solution is collected, washed, and plated in culture media to expand CPCs. Cells are expanded in culture for approximately 3-4 weeks; when cell confluence >80% is attained, the culture is enriched for c-kit+ cells using immunomagnetic beads (CD117 MicroBead Kit, Miltenyi Biotec). The c-kit+ CPCs are further expanded to generate the CPC product. Samples of the cell suspension taken from the last passage are characterized to assure that they meet predetermined specifications, including cell count, phenotype (CD117 positivity), mycoplasma testing, and other QC tests, as indicated in Table 4. Isolated CPC products are then suspended in cryoprotectant at 0.5-1.0×106 cells/ml and cryopreserved using a control rate freezer. The frozen vials are placed into the vapor phase of liquid nitrogen freezers and stored for at least three weeks. These products are not released until the required sterility testing has been completed on samples obtained before and after adding cryopreservation medium as outlined in Tables 4 and 5. Media from the final CPC passage prior to product cryopreservation is likewise cryopreserved and sent to BRC.
Table 5.
Quality Control Tests on Thawed, Washed Cell Product at Center Cell Processing Labs
Assay | Test Method | Specification |
---|---|---|
Rapid Sterility* | Gram Stain | No organisms seen (negative) |
Viability* | Trypan Blue | ≥70% |
Endotoxin* | EndoSafe PTS | ≤ 5EU/kg† |
Aerobic, Anaerobic, and Fungal | 14 day Bactec/BacT/ALERT assay or equivalent | No Growth |
Cell Count – CPCs* | Manual | Minimum 0.8×106 final dose Maximum 5×106 final dose |
Cell Count – MSCs* | Manual | Minimum 75×106 final dose Maximum 150×106 final dose |
Tests done for release criteria specification prior to injection; for cell counts, the product must meet the minimum count for release
Based on recipient weight and product volume
MSC and CPC products are cryopreserved at the CCMF and stored in liquid nitrogen until ready to be shipped to the local cell processing labs. Cell products are transported to each clinical site via validated liquid nitrogen dry shipper within one week prior to the scheduled injection procedure. Local cell processors receive and prepare all products for administration. Placebo product consists of cell-free PlasmaLyte-A. All study products (MSCs alone, CPCs alone, MSCs + CPCs, and placebo) are provided to the treatment teams in a packaging with identical appearance.
Biorepository core (BRC)
An optional central BRC is utilized for participants who provide additional informed consent. The goal is three-fold: i) to provide storage of critical biomaterials (i.e., participant peripheral blood, BM, MSC product, and CPC product), ii) to provide long-term integrity (up to 10 years) of these biospecimens and products, and iii) to provide management of immunologic, immunohistochemical, cellular, and molecular analyses of collected samples, as well as phenotypic and functional analyses of cells and plasma samples. The aim of the BRC is to identify factors that modulate disease progression and/or predict successful intervention and to gain insights into potential mechanisms of action of MSCs and CPCs as well as the relationship of MSC and CPC phenotypes to clinical outcomes. Samples collected and available for further analyses via ancillary studies are specified in Table 6.
Table 6.
Biorepository
Biologic | Sample # | Sample details | Potential ancillary analyses on stored (frozen) samples† |
---|---|---|---|
Bone marrow | n=144 (all patients) |
Fresh BM, flash frozen buffy coat, cryopreserved buffy coat, cryopreserved BM supernatant | In vivo and in vitro studies, RNA, DNA, protein |
BM-MNCs | n=72, (placebo & CPC patients) |
Cryopreserved MNCs | In vivo and in vitro studies, RNA, DNA, protein |
Excess cell product | n=144 (3 active groups)* |
10-30 million MSCs, 0.5-1 million CPCs | In vivo and in vitro studies, RNA, DNA, protein |
Cell culture medium | n=144, (3 active groups)* |
up to 10 12-ml aliquots | Secretome (microparticles/ exosomes, miRNAs, cyto/chemokines, GFs) |
Peripheral Blood | n=144 (all patients) |
Flash frozen buffy coat and plasma, cryopreserved buffy coat and plasma | In vivo and in vitro studies, RNA, DNA, protein, secretome (microparticles/ exosomes, miRNAs, cyto/chemokines, GFs) |
Explanted Heart (as available) |
Unknown | 10 cardiac regions, formalin fixed or flash frozen | RNA, DNA, protein, histology, |
One product/MSC patient, 1 product/CPC patient; 2 products/MSC + CPC patient
Analyses require ancillary study application and funding source outside scope; samples are stored in such a way as to realize these potential future analyses
Study product delivery
Approximately 14 weeks after harvest, participants return to the cardiac catheterization laboratory for cardiac mapping and to receive the study product. The product undergoes cell counts and QC testing before injection, as indicated in Table 5. Each patient receives 15 intramyocardial, electromechanically-guided injections using the NOGA® XP Mapping and Navigation System (Biologic Delivery Systems, McKees Rocks, PA). Intervention groups receive one of three treatments: MSCs in 6 ml of PlasmaLyte-A (target dose, 150×106 cells; minimum dose, 75×106 cells), CPCs in 6 ml of PlasmaLyte-A (target dose, 5×106 cells; minimum dose, 0.8×106), or MSCs + CPCs (same doses as above) in 6 ml of PlasmaLyte-A. The placebo group receives 6 ml of cell-free PlasmaLyte-A. In order to maintain study blinding, if a patient randomized to MSCs + CPCs does not meet the minimum dose of CPCs, he/she will receive MSCs alone and conversely, if a patient does not meet the minimum dose of MSCs, he/she will receive CPCs alone; should both products fail to meet release criteria, the patient would receive placebo. If a patient in either the MSCs or CPCs alone groups does not meet the minimum dose, he/she will receive placebo. For statistical analysis purposes, all participants will be analyzed in the group to which they were randomized in accordance with the “intention-to-treat” principle.
Injections are targeted to the border zones (5-10 mm regions adjacent to scar) identified by electromechanical mapping. Injections are placed so as to encircle the scar, with injection sites in both i) the viable border zone surrounding the area of scar and ii) the scar adjacent to the viable border zone. Priority is given to viable zones with a unipolar voltage ≥7 mV and a premature ventricular contraction on extension of the needle; some injections are made in the border zone with a voltage ≥4mV without the requirement of a premature ventricular contraction. In addition, injection sites should satisfy the following two criteria: i) perpendicular position of the catheter to the LV wall, and ii) loop stability <4 mm. After proper placement of the catheter is confirmed, patients in the treatment groups receive a needle injection of 0.4 ml of solution (5-10 × 106 MSCs and/or 0.05-0.33 × 106 CPCs per injection) delivered over 60 s; those in the placebo group receive vehicle (0.4 ml of PlasmaLyte-A per injection). All patients are monitored overnight. A transthoracic 2-D echo without contrast is performed within 6 h after the procedure to evaluate for pericardial effusion, and blood samples for troponin measurements are collected ~ 8 h following the procedure and prior to discharge.
Outcomes
Visits occur at 1 day, 1 week, and 1, 3, 6, and 12 months after study product injection. A telephone contact takes place at 24 months to assess the patient’s current medications, as well as morbidity and mortality. The primary endpoints assess feasibility, safety, and efficacy (Table 7). All adverse events grade 2 and higher, based on the Common Terminology Criteria for Adverse Events, are captured. These events include major adverse cardiac events related to HF (HF-MACE) (death, hospitalization for worsening HF, and/or exacerbation of HF not requiring hospitalization) and other significant clinical events. The feasibility of harvests, preparing and delivering the intended number of cells, and collecting cardiac MRI variables in patients with cardiac devices, is assessed.
Table 7.
Study Endpoints
Feasibility assessment (the following measures will be reported) |
---|
Number and percent of patients who have:
|
Safety assessment (assessed from baseline to 6 months and from baseline to 12 months) |
Major Adverse Cardiac Events (HF MACE)
|
Other Significant Clinical Events
|
All adverse events at least grade 2 in severity (Common Terminology Criteria for Adverse Events) |
Prospectively declared efficacy endpoint measures: (assessed from baseline to 6 months and from baseline to 12 months) |
Myocardial Function and Structure (cardiac MRI)
|
Functional Capacity
|
Clinical Outcomes
|
Biomarkers
|
Efficacy measures are obtained at baseline and at 6 and 12 months after study product injection, followed by a 24-month phone call. To comprehensively assess the efficacy of cell therapy, multiple endpoints have been selected from different categories of effects (domains) (Table 7). MRI evaluations of LV function, LV structure (including LV volumes), and scar burden provide a comprehensive assessment of myocardial performance and morphology. MRI evaluations are performed at the time of screening, within 30 days prior to study product injection (baseline), and at 6 and 12 months after injection. The presence of a pacemaker or implanted defibrillator device is not a contraindication to MRI33, 34; in these patients, the procedures recommended by the American Heart Association33 are followed. All MRI analyses are performed in a core laboratory by investigators masked to treatment assignment and other patient data. Functional capacity encompasses maximal oxygen consumption (VO2 max, treadmill test) (also assessed by a core laboratory blinded to treatment assignment), 6-min walk test, and quality of life assessment (Minnesota Living with Heart Failure Questionnaire [MLHFQ]). We also assess clinical outcomes (measured by HF-MACE and cumulative days alive and out of the hospital) and biomarkers of heart failure (NT-proBNP).
Statistical methods
The principal analysis will be based on an “intention-to-treat” assessment of the three cell therapies. The reasons for non-treatment of the anticipated small number of participants who are randomized but do not receive any treatment, or do not receive the randomly assigned treatment, will be tabulated. An “as-treated” evaluation placing patients in treatment groups based on what they received also will be conducted. Safety and feasibility data will be reported for participants in both the Stage 1 lead-in study (n=18) and the Stage 2 main cohort study (n=144). Exact testing for categorical variables and analysis of variance for continuous variables will be used to evaluate the differences in baseline variables between treatment groups.
The change in every endpoint variable will be assessed using a repeated-measures general linear model for i) the 6-month follow-up and ii) the 12-month follow-up. The dependent variable in this model will be the change in the treatment group; the independent variable will be the therapy group assignment. In addition, an interaction term consisting of the product of the CPCs and MSCs alone treatment arms will be included to assess the presence of cell therapy synergism. These analyses will provide the effect of each treatment for each assessment time period for each prospectively declared endpoint. Because the effects of MSCs and CPCs in these patients are unknown, and because this is not a pivotal trial, no adjustment for multiplicity will be used. This comprehensive analysis is based on the concept that early phase studies should provide the most complete and elaborative exposition and synthesis of data in order to guide the direction of future cell therapy research35. Sample sizes were chosen assuming a 20% attrition rate. Safety and feasibility data from Stage 1 will be included in the formal analysis of the study.
Underestimation of the sample sizes needed to achieve the desired power would weaken the conclusions of the study, whereas overestimation would cause unnecessary expense of time and resources. Consequently, CONCERT-HF uses an adaptive design for finalizing the sample size. Specifically, the final sample size will be decided on the basis of the standard deviation (SD) observed for each of the change in EF and of the change in infarct size between the baseline and 6-month data points in the control group. Two interim analyses will be performed when 34% and 66% of the control group have completed the 6-month data collection and the data will be submitted to the DSMB for review and recommendation.
RESULTS
Study design
CONCERT-HF is a Phase II trial that assesses the effects of MSCs and CPCs, given alone or in combination, in patients with HF of ischemic etiology. The four-arm design enables this trial to address multiple questions: i) Does administration of MSCs alone have beneficial effects on LV dysfunction and remodeling, scar size, functional capacity, quality of life, and/or clinical outcome? ii) Does administration of CPCs alone have beneficial effects on these variables? iii) Does administration of MSCs + CPCs have beneficial effects on these variables? iv) Is the combination of MSCs and CPCs superior to either cell type alone? v) When compared to each other, is either cell type alone superior? Each of these questions has obvious clinical significance. Considerable new information will be provided by CONCERT-HF. The effects of CPCs in ischemic HF have been evaluated in a Phase I, single-center trial30, 31 but not in multicenter, randomized, double-blind studies powered for efficacy. The relative effects of MSCs and CPCs in patients have not been compared in the same trial. Further, no previous study has evaluated a combination of two different cell types.
Changes in study design resulting from Stage 1 (lead-in study)
Stage 1 was conducted to assess the feasibility and safety of the study procedures as well as the bioactivity of the products. One patient enrolled in Stage 1 died during the EMB procedure because of right ventricular perforation resulting in hemopericardium and tamponade. In a study of patients with severe HF who undergo invasive procedures, adverse events are expected36. However, as a result of the unfortunate death during Stage 1, several important changes to the design were made prior to the initiation of Stage 2 to further reduce risk to study participants. The study protocol was modified as follows: i) the EMB procedure was replaced with a sham procedure (RHC without EMB) in patients allocated to the control and MSCs alone groups; ii) stopping and/or halting thresholds were added on the day of harvest based on RHC pressures, heart rate, and NHYA class; iii) more safety guidance was added for EMB harvesters, and harvesters are required to participate in ongoing best practices sessions aimed at enhancing the safety of the procedure; iv) pre- and post-EMB echocardiograms were added to detect new pericardial effusion; and v) a post-consent case review by the Steering Committee was implemented for each potential participant prior to final eligibility determination. In addition, several procedural changes were made to maximize the ability to consistently reach the target CPC dose of 5×106 cells.
Blinding of investigators
As noted above, Stage 2 includes sham EMB procedures for half of the study population (patients assigned to placebo or MSCs alone groups). In our judgement it is unlikely that the EMB has any impact on patient outcomes, particularly because the study product is administered ~ 3.5 months later. All patients undergo BMA, RHC, and NOGA-guided injections. A sham protocol is followed for patients undergoing RHC without EMB, so that they are not aware of whether an EMB is performed or not. Double-blinding is maintained by having separate blinded and unblinded study teams at each center; the team members directly involved in RHC/EMB procedures are not involved in any other part of the protocol, and all other members of the team are blinded to the therapy assignment. Similarly, cardiac MRI and VO2 max endpoints are determined by core laboratories that are blinded to therapy assignment.
Management of variability in MRI endpoints
The power of CONCERT-HF is very sensitive to the variability of its endpoints. For example, the power to detect an increase of 6 absolute EF units is 81% if the standard deviation (SD) is 8, but drops to 72% if the SD increases by only 1 unit to 9. To minimize variability of MRI-based endpoints, the DCC and the MRI Core Lab monitor the SDs of individual centers (using only data in the control group). If center outliers are identified with respect to the SD of MRI endpoints, an effort will be made to reduce such variability, e.g., by assessing technicians’ ability to obtain images and providing additional training when needed, by evaluating changes in equipment at each center and/or need for software/hardware updates, by monitoring the rate of staff turnover and its impact, etc.
Group sizes and adaptive design
One of the most innovative features of CONCERT-HF is the use of an adaptive design with regard to sample sizes (see Statistical methods). The sample sizes described herein are tentative, and were selected to ensure sufficient power to detect meaningful changes in endpoint measures. Power calculations were based on the variability encountered in previous trials in similar patient populations, including SCIPIO30, 31 and TIME37, LateTIME38, and SWISS-AMI39. According to these estimates, CONCERT-HF is adequately powered to detect significant changes in the following endpoints: a five unit increase in LVEF (power, 89%), a 4.5% decrease in myocardial scar size (93%), a 12 ml decrease in LV end systolic volume (LVESV) (86%), and a 0.08 change in sphericity index (86%). Power for our other prospectively declared endpoints (i.e., change in LV end diastolic volume [LVEDV], VO2 max, 6-minute walk test, and MLHFQ score) is marginal at 70-80%, but given the anticipated interest in these outcomes, we are committed to reporting findings on these variables to the research community. In accordance with the adaptive design of the trial, final group sizes will be determined on the basis of two interim analyses (see Statistical methods).
DISCUSSION
Study design
Because data on the safety and feasibility of MSCs and CPCs have been reported previously16, 18, 30, 31, CONCERT-HF was designed as a Phase II trial to assess not only feasibility and safety but also efficacy, and to allow comparisons of each of the four treatment arms to the other three. Selecting an appropriate primary endpoint, however, is difficult because the effects of MSCs and CPCs and their mechanism of action in humans remain unclear. Although LVEF has been used as the primary endpoint in many trials, failure to improve LVEF may not necessarily signify absence of benefit, as exemplified by recent studies in which cell therapy reduced MACE21, 23 or improved the 6-min walking distance and MLHFQ score16, 18 without improving LVEF. Accordingly, CONCERT-HF assesses multiple primary endpoints in various domains, including LV structure (e.g., volumes), LV function, LV morphology (e.g., scar volume), functional capacity, quality of life, clinical outcome, and biomarkers (Table 7).
If the combination of MSCs and CPCs proves to be beneficial, the question will arise as to which cell type(s) is/are responsible for this effect. Answering this question requires a four-arm design, which makes it possible not only to ascribe to MSCs, CPCs, or both, any favorable changes observed in the MSCs + CPCs group, but also to determine whether the actions of MSCs and CPCs are additive or synergistic. An additional reason for including the CPCs alone group is to continue the evaluation of CPCs that was started in SCIPIO30, 31. Although in that trial patients receiving CPCs exhibited an improvement in LV function, scar size, functional capacity, and quality of life30, 31, SCIPIO was a small, open-label, first-in-human trial designed to assess feasibility and safety, not efficacy, in patients treated 3-4 months after bypass surgery. Therefore, it is important that the encouraging results of SCIPIO be rigorously tested in a Phase II study using a randomized, double-blind, placebo-controlled, multicenter design and without limiting the analysis to the post-bypass surgery setting. In addition, whereas in SCIPIO the route of administration was intracoronary30, 31, CONCERT-HF will provide information regarding the efficacy of CPCs administered transendocardially.
Preclinical and clinical studies of MSCs in chronic ischemic HF
MSCs are a particularly promising cell type for cardiac reparative therapy because of their availability, immunomodulatory properties, and track-record of safety and efficacy4. In rodent and swine models of acute MI, administration of MSCs (both autologous8, 9 and allogeneic6, 7, 10, 11) enhances recovery of LV function. In swine models of chronic MI, MSCs (both autologous12 and allogeneic13), delivered either via epicardial injection12 or transendocardial injection13), augment LV function12–14. In both settings, MSCs have been found to promote angiogenesis, decrease collagen deposition, and reduce myocyte apoptosis4. Importantly, MSCs interact with endogenous precursor cells (i.e., CPCs) that may play an important role in cardiac repair7 (vide infra).
In the clinical arena, several studies have assessed the effects of MSCs, delivered via the transendocardial route, in patients with chronic ischemic HF4, 16, 18, 20–23, 40, 41. The POSEIDON trial16 assessed the safety and efficacy of autologous and allogeneic MSCs (no placebo control group was studied). Compared with baseline, at 1 year both autologous and allogeneic MSCs reduced scar size, but only autologous MSCs improved the 6-min walk distance and MLHFQ score. Neither cell type increased LVEF. The TAC-HFT trial18 evaluated autologous MSCs and unfractionated mononuclear BM cells (BMCs). Compared with placebo at 1 year, both MSC and BMC administration improved the MLHFQ score, but only MSCs improved regional LV function and 6-min walk distance and reduced scar size (although they did not affect LVEF). Autologous MSCs incubated in vitro with a “cardiogenic” cocktail were found to have beneficial effects in the C-CURE trial40 but not in the subsequent larger CHART-1 trial41, although in the latter a subgroup analysis suggested benefit in patients with LVEDV >200 ml. A dose-escalating study of allogeneic STRO-1bright mesenchymal progenitor cells found a reduction in HF-related MACE with the highest dose (150×106 cells) at 3 years, although there was no improvement in LVEF or LV volumes21. In all of these studies16, 18, 21, 40, 41, administration of MSCs was safe, and allogeneic cells did not elicit a detectable immune reaction.
Preclinical and clinical studies of CPCs in chronic ischemic HF
The ability of CPCs to alleviate LV dysfunction has been repeatedly demonstrated by several independent laboratories in various preclinical animal models of acute MI1. CPCs have also been shown to alleviate LV dysfunction and remodeling in rat26, 27, 29, 42 and porcine28 models of chronic MI, where they reduced not only scar size but also collagen deposition in the noninfarcted region26, 27, 29, 42. Because of the poor survival of transplanted cells, these effects have been ascribed to paracrine actions5, 43. The feasibility and safety of CPC therapy in the clinical setting are supported by the SCIPIO trial, a Phase I, open-label trial of autologous CPCs in patients with chronic ischemic HF30, 31. In SCIPIO, CPCs were isolated and expanded in all 20 treated patients, and their intracoronary (i.c.) infusion did not result in detectable adverse effects. At 2 years, CPC-treated patients exhibited improved LV function, functional status (NYHA class), and MLHFQ score as well as a decrease in scar size30, 31. These results provide the rationale for studying the CPCs alone group in CONCERT-HF.
Preclinical studies of the MSCs + CPCs combination
The rationale for examining the combination of MSCs and CPCs stems from studies in porcine models in which administration of MSCs 3 days after MI induced proliferation and differentiation of endogenous CPCs7, suggesting that an MSC-CPC interaction could be an important mechanism for the beneficial effects of MSCs. A subsequent study demonstrated that the combination of both cell types (i.e., human MSCs and CPCs in immunosuppressed pigs), given transendocardially 14 days after MI, produced a greater reduction in scar size and improvement in LV function than each cell type alone, restoring diastolic and systolic LV function toward normal24. Additive beneficial effects of MSCs and CPCs have also been observed in rats25. Importantly, an additional porcine study was conducted, employing autologous porcine cell combination therapy in a model of chronic MI, with cells delivered by NOGA catheter44. In this study, designed to recapitulate the delivery of cells that would occur in humans, the combination of MSCs and CPCs produced synergistic improvements in global and regional LV function relative to either cell type alone44. Together, these findings illustrate potentially important biological interactions between MSCs and CPCs that enhance cell therapeutic responses.
Rationale for dose selection
The target dose of MSCs in CONCERT-HF (150×106) was chosen on the basis of a dose-escalating trial of STRO-1bright MPCs21 (cells similar to MSCs), in which increasing doses of MPCs were associated with decreasing LVEDV and LVESV; the benefits were maximal at a dose of 150×106 cells, and no serious adverse events were reported. The safety of this dose is further supported by the fact that transendocardial injection of up to 200×106 MSCs was safe in preclinical porcine models12, 13 and in the TAC-HFT18 and POSEIDON trials16, and that intravenous doses of MSCs >200×106 cells have been used with no significant adverse effects15.
The choice of the target dose of CPCs (5×106) reflects a number of considerations. Pig studies have shown that post-MI LV dysfunction is effectively ameliorated by 5×105 CPCs (a dose equivalent to ~1×106 CPCs in humans) given by the i.c. route28 or 1×106 CPCs (equivalent to ~2×106 CPCs in humans) given transendocardially24. In the only clinical trial of CPCs to date, SCIPIO30, 31, 1×106 CPCs were given by the i.c. route, with encouraging results regarding LV function, scar size, functional status, and quality of life30. Furthermore, i.c. infusion of higher CPC doses in pigs (up to 20×106, equivalent to ~40×106 CPCs in humans) is well tolerated and produces no discernible adverse effects, as assessed by cardiac enzymes, myocardial function, and histology45. Thus, both preclinical and clinical evidence supports the notion that 5×106 CPCs should be effective in humans. Although higher doses could conceivably have greater efficacy, it would be challenging to reproducibly generate >5×106 CPCs from small amounts of cardiac tissue (10-20 mg). In conclusion, the target cell doses for CONCERT-HF (150×106 MSCs and 5×106 CPCs) have a strong safety profile and there is considerable evidence for their potential to improve LV function and functional status in patients with HF.
Rationale for using the transendocardial route of administration
In previous studies in porcine models7, 10, 11, 13, 24 and in humans16, 18, 21, 40, 41, transendocardial delivery of MSCs improved post-MI LV dysfunction and remodeling and (in clinical trials) functional capacity and MLHFQ score. The efficacy of intramyocardial injections of CPCs has been well documented in rodents25, 46 and in pigs1, 24. Although the only previous clinical trial of CPCs used i.c. infusion30, 31, we chose the transendocardial route for CPCs so that MSCs and CPCs could be delivered together. Transendocardial delivery was the route used for both cell types in the porcine studies that support the superiority of the combination over either cell type alone7, 24; presumably, the juxtaposition of MSCs and CPCs promotes their interactions and may be the basis for their additive therapeutic effects4. Additional advantages of transendocardial delivery are that it is associated with greater myocardial retention47 and makes it possible to reach regions distal to occluded coronary arteries.
Use of MRI in patients with cardiac devices
MRI was selected over other imaging modalities, such as cardiac CT and echocardiography, because it enables accurate, high-resolution assessment of myocardial scar48-50, is considered to be more reproducible for measuring LV function and volumes51, 52, and does not require any radiation exposure. One of the innovative aspects of CONCERT-HF is that MRI is used in patients with pacemakers and/or implanted defibrillator devices. Traditionally, these patients have been excluded because of the concern that the MRI exam may interfere with the function of the device and also because of the suboptimal image quality53, 54. However, studies have shown that with appropriate study candidate selection as well as software, equipment, and positioning optimization, MRI scanning can be performed safely in the presence of most cardiac devices without precluding acquisition of meaningful data33, 34. In this trial, extensive in-person and webinar-based training procedures were put in place at the local Radiology and Cardiology departments at each site under the supervision of the MRI Core lab to ensure patient safety and proper execution of the MRI protocol. The initial experience garnered at the MRI Core at Johns Hopkins University shows that, with the use of the exclusion criteria listed in Table 2 and appropriate software55-57, MRI scans can be obtained safely and the images can be analyzed for LV volumes and scar size in the vast majority of the cases. The experience of CONCERT-HF will generate new insights into the feasibility of using of MRI in patients with cardiac devices, which may have an important impact on the design of future studies not only of cell therapy but also of other interventions in HF. This information also may help obviate the problems stemming from the fact that patients may acquire devices over the course of a trial, which results in significant drop-off of patients when serial MRI imaging is utilized to assess changes in LV function over time58.
Limitations
Because of the innovative nature of CONCERT-HF, the feasibility of certain aspects of the study is unclear. EMBs may not be sufficient to generate the target dose of CPCs (5×106) in all patients. The long interval between EMB and product injection (necessitated by the time needed for expansion) may cause some patient drop-out. Since this is the first trial to use MRI in patients with devices, the quality and reproducibility of images are not known. CONCERT-HF will furnish important feasibility information regarding these issues. An additional possible limitation is that, depending on variability, the trial may not have enough power to assess some of its endpoints.
In conclusion, CONCERT-HF is the first cell therapy clinical trial to evaluate the combination of two cell types. This study will provide important insights into the potential therapeutic utility of MSCs and CPCs, given alone and in combination, for patients with ischemic HF. In addition, the collection and storage of HF patient-specific biomaterials may offer important tools for future ancillary studies of regenerative medicine.
NOVELTY AND SIGNIFICANCE.
What Is Known?
Bone marrow mesenchymal stem cells (MSCs) ameliorate left ventricular (LV) dysfunction and reduce scar size in preclinical models of chronic ischemic cardiomyopathy (ICM), and several clinical trials suggest that MSCs are safe and beneficial in patients with chronic heart failure (HF) of ischemic etiology.
c-kit+ cardiac progenitor cells (CPCs) also attenuate LV dysfunction and remodeling in animal models of chronic ICM. The SCIPIO trial has suggested benefit in patients with ischemic HF, but the study was not designed to establish efficacy.
When MSCs and CPCs are combined in preclinical models, the therapeutic effects are additive, and thus superior to those of either cell type alone. However, neither the concept of combinatorial stem cell therapy nor the relative efficacy of MSCs vs. CPCs has been tested in humans.
What New Information Does This Article Provide?
We describe CONCERT-HF, a Phase II trial that assesses the feasibility, safety, and efficacy of autologous MSCs and CPCs, given alone or in combination, in patients with ischemic HF.
CONCERT-HF is the first trial to evaluate the effects of CPCs in ischemic HF with a multicenter, randomized, double-blind protocol powered for efficacy.
CONCERT-HF is the first trial to compare the relative effects of MSCs and CPCs in patients in the same study.
CONCERT-HF is the first trial to evaluate a combination of two different cell types.
Other innovative features of CONCERT-HF include the adaptive design and the use of magnetic resonance imaging (MRI) in patients with devices.
The four-arm design (MSCs alone, CPCs alone, MSCs + CPCs, and vehicle) enables CONCERT-HF to address multiple questions: i) Is combined treatment with autologous MSCs and CPCs feasible and safe in patients with ischemic HF? ii) Does administration of MSCs alone have beneficial effects on LV dysfunction and remodeling, scar size, functional capacity, quality of life, and/or clinical outcome? iii) Does administration of CPCs alone have beneficial effects on these variables? iv) Does administration of MSCs + CPCs have beneficial effects on these variables? v) Is the combination of MSCs and CPCs superior to either cell type alone? vi) When compared to each other, is either cell type alone superior? Each of these questions has obvious significance for the development of cell-based therapies in patients with ischemic HF. In addition, the collection and storage of HF patient-specific biomaterials may offer important tools for future ancillary studies of regenerative medicine.
Acknowledgments
SOURCES OF FUNDING
This work is supported by National Institutes of Health, Bethesda, MD: 5 UM1 HL087318.
DISCLOSURES
All investigators received funding from the NIH National Heart, Lung, and Blood Institute (NHLBI) for conduct of the Cardiovascular Cell Therapy Research Network (CCTRN). Additional disclosures: Hare has a relationship with Vestion Inc that includes equity, board membership, and consulting. Dr. Ebert is a staff member of the NHLBI, the source of funding for the CONCERT-HF Trial. The views expressed in this article are those of the authors and do not necessarily represent the views of the National Heart, Lung, and Blood Institute, National Institutes of Health, or the United States Department of Health and Human Services.
Nonstandard Abbreviations and Acronyms
- BM
bone marrow
- BMA
bone marrow aspiration
- BMCs
bone marrow cells
- BRC
CCTRN Biorepository
- CCMF
Central Cell Manufacturing Facility
- CPCs
c-kit+ cardiac progenitor cells
- DCC
Data Coordinating Center
- EMB
endocardial biopsy
- HF
heart failure
- HF-MACE
major adverse cardiac events related to heart failure i.c. intracoronary
- ICM
ischemic cardiomyopathy
- LV
left ventricular
- LVEDV
left ventricular end diastolic volume
- LVEF
left ventricular ejection fractions
- LVESV
left ventricular end systolic volume
- MI
myocardial infarction
- MLHFQ
Minnesota Living with Heart Failure Questionnaire
- MNCs
mononuclear cells
- MRI
magnetic resonance imaging
- MSCs
mesenchymal stem cells
- QC
quality control
- RHC
right heart catheterization
- SD
standard deviation
- VO2 max
maximal oxygen consumption
Footnotes
References
- 1.Sanganalmath SK, Bolli R. Cell therapy for heart failure: A comprehensive overview of experimental and clinical studies, current challenges, and future directions. Circ Res. 2013;113:810–834. doi: 10.1161/CIRCRESAHA.113.300219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Keith MC, Bolli R. "String theory" of c-kit(pos) cardiac cells: A new paradigm regarding the nature of these cells that may reconcile apparently discrepant results. Circ Res. 2015;116:1216–1230. doi: 10.1161/CIRCRESAHA.116.305557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Mangi AA, Noiseux N, Kong D, He H, Rezvani M, Ingwall JS, Dzau VJ. Mesenchymal stem cells modified with akt prevent remodeling and restore performance of infarcted hearts. Nat Med. 2003;9:1195–1201. doi: 10.1038/nm912. [DOI] [PubMed] [Google Scholar]
- 4.Hatzistergos KE, Quevedo H, Oskouei BN, Hu Q, Feigenbaum GS, Margitich IS, Mazhari R, Boyle AJ, Zambrano JP, Rodriguez JE, Dulce R, Pattany PM, Valdes D, Revilla C, Heldman AW, McNiece I, Hare JM. Bone marrow mesenchymal stem cells stimulate cardiac stem cell proliferation and differentiation. Circ Res. 2010;107:913–922. doi: 10.1161/CIRCRESAHA.110.222703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Shake JG, Gruber PJ, Baumgartner WA, Senechal G, Meyers J, Redmond JM, Pittenger MF, Martin BJ. Mesenchymal stem cell implantation in a swine myocardial infarct model: Engraftment and functional effects. Ann Thorac Surg. 2002;73:1919–25. doi: 10.1016/s0003-4975(02)03517-8. discussion 1926. [DOI] [PubMed] [Google Scholar]
- 6.Schuleri KH, Amado LC, Boyle AJ, Centola M, Saliaris AP, Gutman MR, Hatzistergos KE, Oskouei BN, Zimmet JM, Young RG, Heldman AW, Lardo AC, Hare JM. Early improvement in cardiac tissue perfusion due to mesenchymal stem cells. Am J Physiol Heart Circ Physiol. 2008;294:H2002–11. doi: 10.1152/ajpheart.00762.2007. [DOI] [PubMed] [Google Scholar]
- 7.Amado LC, Saliaris AP, Schuleri KH, St John M, Xie JS, Cattaneo S, Durand DJ, Fitton T, Kuang JQ, Stewart G, Lehrke S, Baumgartner WW, Martin BJ, Heldman AW, Hare JM. Cardiac repair with intramyocardial injection of allogeneic mesenchymal stem cells after myocardial infarction. Proc Natl Acad Sci U S A. 2005;102:11474–11479. doi: 10.1073/pnas.0504388102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Amado LC, Schuleri KH, Saliaris AP, Boyle AJ, Helm R, Oskouei B, Centola M, Eneboe V, Young R, Lima JA, Lardo AC, Heldman AW, Hare JM. Multimodality noninvasive imaging demonstrates in vivo cardiac regeneration after mesenchymal stem cell therapy. J Am Coll Cardiol. 2006;48:2116–2124. doi: 10.1016/j.jacc.2006.06.073. [DOI] [PubMed] [Google Scholar]
- 9.Schuleri KH, Feigenbaum GS, Centola M, Weiss ES, Zimmet JM, Turney J, Kellner J, Zviman MM, Hatzistergos KE, Detrick B, Conte JV, McNiece I, Steenbergen C, Lardo AC, Hare JM. Autologous mesenchymal stem cells produce reverse remodelling in chronic ischaemic cardiomyopathy. Eur Heart J. 2009;30:2722–2732. doi: 10.1093/eurheartj/ehp265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Quevedo HC, Hatzistergos KE, Oskouei BN, Feigenbaum GS, Rodriguez JE, Valdes D, Pattany PM, Zambrano JP, Hu Q, McNiece I, Heldman AW, Hare JM. Allogeneic mesenchymal stem cells restore cardiac function in chronic ischemic cardiomyopathy via trilineage differentiating capacity. Proc Natl Acad Sci U S A. 2009;106:14022–14027. doi: 10.1073/pnas.0903201106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Schuleri KH, Boyle AJ, Centola M, Amado LC, Evers R, Zimmet JM, Evers KS, Ostbye KM, Scorpio DG, Hare JM, Lardo AC. The adult gottingen minipig as a model for chronic heart failure after myocardial infarction: Focus on cardiovascular imaging and regenerative therapies. Comp Med. 2008;58:568–579. [PMC free article] [PubMed] [Google Scholar]
- 12.Hare JM, Traverse JH, Henry TD, Dib N, Strumpf RK, Schulman SP, Gerstenblith G, DeMaria AN, Denktas AE, Gammon RS, Hermiller JB, Jr, Reisman MA, Schaer GL, Sherman W. A randomized, double-blind, placebo-controlled, dose-escalation study of intravenous adult human mesenchymal stem cells (prochymal) after acute myocardial infarction. J Am Coll Cardiol. 2009;54:2277–2286. doi: 10.1016/j.jacc.2009.06.055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Hare JM, Fishman JE, Gerstenblith G, Difede Velazquez DL, Zambrano JP, Suncion VY, Tracy M, Ghersin E, Johnston PV, Brinker JA, Breton E, Davis-Sproul J, Schulman IH, Byrnes J, Mendizabal AM, Lowery MH, Rouy D, Altman P, Wong Po Foo C, Ruiz P, Amador A, Da Silva J, McNiece IK, Heldman AW. Comparison of allogeneic vs autologous bone marrow-derived mesenchymal stem cells delivered by transendocardial injection in patients with ischemic cardiomyopathy: The POSEIDON randomized trial. JAMA. 2012;308:2369–2379. doi: 10.1001/jama.2012.25321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Karantalis V, Difede DL, Gerstenblith G, Pham S, Symes J, Zambrano JP, Fishman J, Pattany P, McNiece I, Conte J, Schulman S, Wu K, Shah A, Breton E, Davis-Sproul J, Schwarz R, Feigenbaum G, Mushtaq M, Suncion VY, Lardo AC, Borrello I, Mendizabal A, Karas TZ, Byrnes J, Lowery M, Heldman AW, Hare JM. Autologous mesenchymal stem cells produce concordant improvements in regional function, tissue perfusion, and fibrotic burden when administered to patients undergoing coronary artery bypass grafting: The prospective randomized study of mesenchymal stem cell therapy in patients undergoing cardiac surgery (PROMETHEUS) trial. Circ Res. 2014;114:1302–1310. doi: 10.1161/CIRCRESAHA.114.303180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Heldman AW, DiFede DL, Fishman JE, Zambrano JP, Trachtenberg BH, Karantalis V, Mushtaq M, Williams AR, Suncion VY, McNiece IK, Ghersin E, Soto V, Lopera G, Miki R, Willens H, Hendel R, Mitrani R, Pattany P, Feigenbaum G, Oskouei B, Byrnes J, Lowery MH, Sierra J, Pujol MV, Delgado C, Gonzalez PJ, Rodriguez JE, Bagno LL, Rouy D, Altman P, Foo CW, da Silva J, Anderson E, Schwarz R, Mendizabal A, Hare JM. Transendocardial mesenchymal stem cells and mononuclear bone marrow cells for ischemic cardiomyopathy: The TAC-HFT randomized trial. JAMA. 2014;311:62–73. doi: 10.1001/jama.2013.282909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Williams AR, Trachtenberg B, Velazquez DL, McNiece I, Altman P, Rouy D, Mendizabal AM, Pattany PM, Lopera GA, Fishman J, Zambrano JP, Heldman AW, Hare JM. Intramyocardial stem cell injection in patients with ischemic cardiomyopathy: Functional recovery and reverse remodeling. Circ Res. 2011;108:792–796. doi: 10.1161/CIRCRESAHA.111.242610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Mathiasen AB, Qayyum AA, Jorgensen E, Helqvist S, Fischer-Nielsen A, Kofoed KF, Haack-Sorensen M, Ekblond A, Kastrup J. Bone marrow-derived mesenchymal stromal cell treatment in patients with severe ischaemic heart failure: A randomized placebo-controlled trial (MSC-HF trial) Eur Heart J. 2015;36:1744–1753. doi: 10.1093/eurheartj/ehv136. [DOI] [PubMed] [Google Scholar]
- 18.Perin EC, Borow KM, Silva GV, DeMaria AN, Marroquin OC, Huang PP, Traverse JH, Krum H, Skerrett D, Zheng Y, Willerson JT, Itescu S, Henry TD. A phase II dose-escalation study of allogeneic mesenchymal precursor cells in patients with ischemic or nonischemic heart failure. Circ Res. 2015;117:576–584. doi: 10.1161/CIRCRESAHA.115.306332. [DOI] [PubMed] [Google Scholar]
- 19.Williams AR, Hatzistergos KE, Addicott B, McCall F, Carvalho D, Suncion V, Morales AR, Da Silva J, Sussman MA, Heldman AW, Hare JM. Enhanced effect of human cardiac stem cells and bone marrow mesenchymal stem cells to reduce infarct size and restore cardiac function after myocardial infarction. Circulation. 2013;127:213–223. doi: 10.1161/CIRCULATIONAHA.112.131110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Tang XL, Rokosh G, Sanganalmath SK, Yuan F, Sato H, Mu J, Dai S, Li C, Chen N, Peng Y, Dawn B, Hunt G, Leri A, Kajstura J, Tiwari S, Shirk G, Anversa P, Bolli R. Intracoronary administration of cardiac progenitor cells alleviates left ventricular dysfunction in rats with a 30-day-old infarction. Circulation. 2010;121:293–305. doi: 10.1161/CIRCULATIONAHA.109.871905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Bolli R, Tang XL, Sanganalmath SK, Rimoldi O, Mosna F, Abdel-Latif A, Jneid H, Rota M, Leri A, Kajstura J. Intracoronary delivery of autologous cardiac stem cells improves cardiac function in a porcine model of chronic ischemic cardiomyopathy. Circulation. 2013;128:122–131. doi: 10.1161/CIRCULATIONAHA.112.001075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Bolli R, Chugh AR, D’Amario D, Loughran JH, Stoddard MF, Ikram S, Beache GM, Wagner SG, Leri A, Hosoda T, Sanada F, Elmore JB, Goichberg P, Cappetta D, Solankhi NK, Fahsah I, Rokosh DG, Slaughter MS, Kajstura J, Anversa P. Cardiac stem cells in patients with ischaemic cardiomyopathy (SCIPIO): Initial results of a randomised phase 1 trial. Lancet. 2011;378:1847–1857. doi: 10.1016/S0140-6736(11)61590-0. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 23.Chugh AR, Beache GM, Loughran JH, Mewton N, Elmore JB, Kajstura J, Pappas P, Tatooles A, Stoddard MF, Lima JA, Slaughter MS, Anversa P, Bolli R. Administration of cardiac stem cells in patients with ischemic cardiomyopathy: The SCIPIO trial: Surgical aspects and interim analysis of myocardial function and viability by magnetic resonance. Circulation. 2012;126:S54–64. doi: 10.1161/CIRCULATIONAHA.112.092627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Zierold C, Carlson MA, Obodo UC, Wise E, Piazza VA, Meeks MW, Vojvodic RW, Baraniuk S, Henry TD, Gee AP, Ellis SG, Moye LA, Pepine CJ, Cogle CR, Taylor DA. Developing mechanistic insights into cardiovascular cell therapy: Cardiovascular cell therapy research network biorepository core laboratory rationale. Am Heart J. 2011;162:973–980. doi: 10.1016/j.ahj.2011.05.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Levine GN, Gomes AS, Arai AE, Bluemke DA, Flamm SD, Kanal E, Manning WJ, Martin ET, Smith JM, Wilke N, Shellock FS, American Heart Association Committee on Diagnostic and Interventional Cardiac Catheterization, American Heart Association Council on Clinical Cardiology, American Heart Association Council on Cardiovascular Radiology and Intervention Safety of magnetic resonance imaging in patients with cardiovascular devices: An american heart association scientific statement from the committee on diagnostic and interventional cardiac catheterization, council on clinical cardiology, and the council on cardiovascular radiology and intervention: Endorsed by the american college of cardiology foundation, the north american society for cardiac imaging, and the society for cardiovascular magnetic resonance. Circulation. 2007;116:2878–2891. doi: 10.1161/CIRCULATIONAHA.107.187256. [DOI] [PubMed] [Google Scholar]
- 26.Junttila MJ, Fishman JE, Lopera GA, Pattany PM, Velazquez DL, Williams AR, Trachtenberg BH, Sanina C, Mather J, Hare JM. Safety of serial MRI in patients with implantable cardioverter defibrillators. Heart. 2011;97:1852–1856. doi: 10.1136/heartjnl-2011-300153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Hare JM, Bolli R, Cooke JP, Gordon DJ, Henry TD, Perin EC, March KL, Murphy MP, Pepine CJ, Simari RD, Skarlatos SI, Traverse JH, Willerson JT, Szady AD, Taylor DA, Vojvodic RW, Yang PC, Moye LA, Cardiovascular Cell Therapy Research Network (CCTRN) Phase II clinical research design in cardiology: Learning the right lessons too well: Observations and recommendations from the cardiovascular cell therapy research network (CCTRN) Circulation. 2013;127:1630–1635. doi: 10.1161/CIRCULATIONAHA.112.000779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Deckers JW, Hare JM, Baughman KL. Complications of transvenous right ventricular endomyocardial biopsy in adult patients with cardiomyopathy: A seven-year survey of 546 consecutive diagnostic procedures in a tertiary referral center. J Am Coll Cardiol. 1992;19:43–47. doi: 10.1016/0735-1097(92)90049-s. [DOI] [PubMed] [Google Scholar]
- 29.Traverse JH, Henry TD, Pepine CJ, Willerson JT, Zhao DX, Ellis SG, Forder JR, Anderson RD, Hatzopoulos AK, Penn MS, Perin EC, Chambers J, Baran KW, Raveendran G, Lambert C, Lerman A, Simon DI, Vaughan DE, Lai D, Gee AP, Taylor DA, Cogle CR, Thomas JD, Olson RE, Bowman S, Francescon J, Geither C, Handberg E, Kappenman C, Westbrook L, Piller LB, Simpson LM, Baraniuk S, Loghin C, Aguilar D, Richman S, Zierold C, Spoon DB, Bettencourt J, Sayre SL, Vojvodic RW, Skarlatos SI, Gordon DJ, Ebert RF, Kwak M, Moye LA, Simari RD, Cardiovascular Cell Therapy Research Network (CCTRN) Effect of the use and timing of bone marrow mononuclear cell delivery on left ventricular function after acute myocardial infarction: The TIME randomized trial. JAMA. 2012;308:2380–2389. doi: 10.1001/jama.2012.28726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Traverse JH, Henry TD, Ellis SG, Pepine CJ, Willerson JT, Zhao DX, Forder JR, Byrne BJ, Hatzopoulos AK, Penn MS, Perin EC, Baran KW, Chambers J, Lambert C, Raveendran G, Simon DI, Vaughan DE, Simpson LM, Gee AP, Taylor DA, Cogle CR, Thomas JD, Silva GV, Jorgenson BC, Olson RE, Bowman S, Francescon J, Geither C, Handberg E, Smith DX, Baraniuk S, Piller LB, Loghin C, Aguilar D, Richman S, Zierold C, Bettencourt J, Sayre SL, Vojvodic RW, Skarlatos SI, Gordon DJ, Ebert RF, Kwak M, Moye LA, Simari RD, Cardiovascular Cell Therapy Research Network Effect of intracoronary delivery of autologous bone marrow mononuclear cells 2 to 3 weeks following acute myocardial infarction on left ventricular function: The LateTIME randomized trial. JAMA. 2011;306:2110–2119. doi: 10.1001/jama.2011.1670. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Surder D, Manka R, Moccetti T, Lo Cicero V, Emmert MY, Klersy C, Soncin S, Turchetto L, Radrizzani M, Zuber M, Windecker S, Moschovitis A, Buhler I, Kozerke S, Erne P, Luscher TF, Corti R. Effect of bone marrow-derived mononuclear cell treatment, early or late after acute myocardial infarction: Twelve months CMR and long-term clinical results. Circ Res. 2016;119:481–490. doi: 10.1161/CIRCRESAHA.116.308639. [DOI] [PubMed] [Google Scholar]
- 32.Bartunek J, Behfar A, Dolatabadi D, Vanderheyden M, Ostojic M, Dens J, El Nakadi B, Banovic M, Beleslin B, Vrolix M, Legrand V, Vrints C, Vanoverschelde JL, Crespo-Diaz R, Homsy C, Tendera M, Waldman S, Wijns W, Terzic A. Cardiopoietic stem cell therapy in heart failure: The C-CURE (cardiopoietic stem cell therapy in heart failURE) multicenter randomized trial with lineage-specified biologics. J Am Coll Cardiol. 2013;61:2329–2338. doi: 10.1016/j.jacc.2013.02.071. [DOI] [PubMed] [Google Scholar]
- 33.Tang XL, Rokosh G, Sanganalmath SK, Tokita Y, Keith MC, Shirk G, Stowers H, Hunt GN, Wu W, Dawn B, Bolli R. Effects of intracoronary infusion of escalating doses of cardiac stem cells in rats with acute myocardial infarction. Circ Heart Fail. 2015;8:757–765. doi: 10.1161/CIRCHEARTFAILURE.115.002210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Karantalis V, Suncion-Loescher VY, Bagno L, Golpanian S, Wolf A, Sanina C, Premer C, Kanelidis AJ, McCall F, Wang B, Balkan W, Rodriguez J, Rosado M, Morales A, Hatzistergos K, Natsumeda M, Margitich I, Schulman IH, Gomes SA, Mushtaq M, DiFede DL, Fishman JE, Pattany P, Zambrano JP, Heldman AW, Hare JM. Synergistic effects of combined cell therapy for chronic ischemic cardiomyopathy. J Am Coll Cardiol. 2015;66:1990–1999. doi: 10.1016/j.jacc.2015.08.879. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Keith MC, Tang XL, Tokita Y, Li QH, Ghafghazi S, Moore Iv J, Hong KU, Elmore B, Amraotkar A, Ganzel BL, Grubb KJ, Flaherty MP, Hunt G, Vajravelu B, Wysoczynski M, Bolli R. Safety of intracoronary infusion of 20 million C-kit positive human cardiac stem cells in pigs. PLoS One. 2015;10:e0124227. doi: 10.1371/journal.pone.0124227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Hou D, Youssef EA, Brinton TJ, Zhang P, Rogers P, Price ET, Yeung AC, Johnstone BH, Yock PG, March KL. Radiolabeled cell distribution after intramyocardial, intracoronary, and interstitial retrograde coronary venous delivery: Implications for current clinical trials. Circulation. 2005;112:I150–6. doi: 10.1161/CIRCULATIONAHA.104.526749. [DOI] [PubMed] [Google Scholar]
- 37.Kim RJ, Albert TS, Wible JH, Elliott MD, Allen JC, Lee JC, Parker M, Napoli A, Judd RM, Gadoversetamide Myocardial Infarction Imaging Investigators Performance of delayed-enhancement magnetic resonance imaging with gadoversetamide contrast for the detection and assessment of myocardial infarction: An international, multicenter, double-blinded, randomized trial. Circulation. 2008;117:629–637. doi: 10.1161/CIRCULATIONAHA.107.723262. [DOI] [PubMed] [Google Scholar]
- 38.Amado LC, Gerber BL, Gupta SN, Rettmann DW, Szarf G, Schock R, Nasir K, Kraitchman DL, Lima JA. Accurate and objective infarct sizing by contrast-enhanced magnetic resonance imaging in a canine myocardial infarction model. J Am Coll Cardiol. 2004;44:2383–2389. doi: 10.1016/j.jacc.2004.09.020. [DOI] [PubMed] [Google Scholar]
- 39.Ambale-Venkatesh B, Lima JA. Cardiac MRI: A central prognostic tool in myocardial fibrosis. Nat Rev Cardiol. 2015;12:18–29. doi: 10.1038/nrcardio.2014.159. [DOI] [PubMed] [Google Scholar]
- 40.Grothues F, Smith GC, Moon JC, Bellenger NG, Collins P, Klein HU, Pennell DJ. Comparison of interstudy reproducibility of cardiovascular magnetic resonance with two-dimensional echocardiography in normal subjects and in patients with heart failure or left ventricular hypertrophy. Am J Cardiol. 2002;90:29–34. doi: 10.1016/s0002-9149(02)02381-0. [DOI] [PubMed] [Google Scholar]
- 41.Schulz-Menger J, Bluemke DA, Bremerich J, Flamm SD, Fogel MA, Friedrich MG, Kim RJ, von Knobelsdorff-Brenkenhoff F, Kramer CM, Pennell DJ, Plein S, Nagel E. Standardized image interpretation and post processing in cardiovascular magnetic resonance: Society for cardiovascular magnetic resonance (SCMR) board of trustees task force on standardized post processing. J Cardiovasc Magn Reson. 2013;15:35–429X-15-35. doi: 10.1186/1532-429X-15-35. [DOI] [PMC free article] [PubMed] [Google Scholar]