Introduction
Although therapy for cardiovascular disease has led to consistent annual declines in mortality, myocardial infarction (MI) still represents an irreversible injury to the myocardium leading to the substrate for heart failure and sudden cardiac death1. Indeed, the extent of scar resulting from MI is an important predictor of mortality2. Even with timely coronary intervention, infarct size is a significant problem likely exacerbated by ischemia/reperfusion injury3. Supported by preclinical studies, cell-based therapy has emerged as an attractive treatment for minimizing/reversing the effects of MI in patients4, 5.
Stem cell therapy following acute myocardial infarction (AMI)
Stem cell (SC) mobilization from the bone marrow to acutely injured tissue significantly enhancing wound healing was first observed in a mouse skin-wound model6. Large animal models demonstrated that SC therapy produces significant improvements in AMI7,8, leading to clinical trials for SC therapy in patients with AMI and heart failure. Bone marrow-derived SC therapy for AMI is safe. Unfortunately, the hypoxic post-AMI environment is hostile to cardiomyocytes and migrating or introduced SCs, and this proapoptotic milieu may be the limiting factor clinically. However, a recent meta-analysis reported that patients with ischemic cardiomyopathy who received bone marrow-derived SCs exhibited improved left ventricular ejection fraction (LVEF), and reduced infarct size and remodeling9 (Table).
Table.
Source | Sample size |
Cell type | Number of cells |
Route of injection |
Results |
---|---|---|---|---|---|
AMI | |||||
Tendera et al. 2009 |
200 | CD34+ | NA | IC | No significant improvement |
Quyyumi et al. 2011 | 31 | CD34+ | 5–15×106 | IC | Dose dependent perfusion improvement |
Quyyumi et al. 2016 | 161 | CD34+ | 8–43×106 | IC | No significant improvement |
ICM | |||||
Patel et al. 2005 |
20 | CD34+ | NR | IM | Improved EF |
Refractory Angina | |||||
Losordo et al. 2007 |
24 | Peripheral CD34+ + G-CSF |
NR | IM | No significant improvement |
Losordo et al. 2011 | 167 | Peripheral CD34+ + G-CSF |
1–5×105/ Kg |
IM | Improved angina and exercise tolerance in low dose group |
Henry et al. 2016 |
112 | Peripheral CD34+ + G-CSF |
1–100 ×105/Kg |
IM | Early termination of study Reduced angina frequency |
NIDCM | |||||
Vrtovec et al. 2013 |
110 | Peripheral CD34+ + G-CSF |
1.13±0.26 ×108 |
IC | Improved EF, 6MWD Decreased NT-pro-BNP Lower mortality for stem cell treatment |
Vrtovec et al. 2016 |
45 | Peripheral CD34+ + G-CSF |
1.27– 2.16×108 |
IM | No response in diabetics Increased EF, decreased NT-pro-BNP in non- diabetics |
Abbreviations: AMI, acute myocardial infarction; EF, ejection fraction; G-CSF, granulocyte-colony stimulating factor; IC, intracoronary; ICM, ischemic cardiomyopathy; IM, intramuscular; NA, not available; NIDCM, non-ischemic dilated cardiomyopathy; NR, not reported; RCT, randomized controlled trial
CD34+ cells in clinical settings
Endothelial progenitor cells (EPCs) are bone marrow-derived mononuclear cells expressing both hematopoietic SC and endothelial cell markers. The prototypical EPC, selected on the basis of CD34 expression (CD34+), promotes neovascularization and regeneration10. The neovascular effects were demonstrated in a Phase I/II trial, where CD34+ EPCs administered to patients with refractory angina pectoris, decreased the frequency of events and increased exercise tolerance as compared to placebo-treated patients11. Furthermore, CD34+ EPCs mobilize from the bone-marrow post-AMI and enter the peripheral circulation; this degree of mobilization is directly correlated with improved outcomes12. However, large ischemic insults and adverse remodeling remain an extensive burden, even for efficient mobilizers. The extensive preclinical data supporting ischemic tissue repair by CD34+ cells prompted Quyyumi et al. to hypothesize that the effects of CD34+ cells were dependent on quantity and mobility following an ST Elevation MI (STEMI)13. They reported a positive dose-dependent improvement in cell mobility, cardiac perfusion, and scar size reduction following intracoronary infusion of CD34+ EPCs in subjects post-STEMI13. Those encouraging results inspired the current Phase II clinical trial (PreSERVE-AMI) to further elucidate the safety and bioactivity of autologous CD34+ marrow cells in patients post-STEMI12.
PreSERVE-AMI Trial
The PreSERVE-AMI trial: A Randomized, Double-Blind, Placebo-Controlled Clinical Trial of Intracoronary Administration of Autologous CD34+ Cells in Patients with Left Ventricular Dysfunction Post-STEMI, was novel and appropriately powered12. Subjects with successful stenting status-post-STEMI were randomized to receive autologous CD34+ cells (n=78) or placebo (n=81) via intracoronary infusion following bone marrow harvest.
The primary safety endpoints were adverse events (AEs), serious AEs (SAEs) and major adverse cardiac event (MACE). There were no differences between placebo and treated subjects in these categories. No differences were observed in survival or incidence of MACE in the treated group, regardless of dose (p≥0.05). AE and SAE incidence was similar between control and treated subjects at 12-month follow-up. The primary efficacy endpoint was improvement in resting myocardial perfusion over 6 months, which was not met, as there were no differences between groups. Furthermore, no changes in LVEF or scar size at 6 months were observed between groups. Despite primary endpoints not being met, post-hoc analyses were significant for reductions in infarct size, and changes in LVEF, after adjusting for total ischemia time. Additional testing with a larger patient population with allogeneic CD34+ cells may clarify the positive effects noted on tertiary analyses.
Impact of negative trials in regenerative medicine
While positive trials are enticing and provide a direction for the field of regenerative medicine, negative studies can be just as impactful. Negative studies move the field forward by avoiding repetition of ineffectual trials. Negative and positive trials save the field time and money, which in the end promotes higher quality study designs and conclusions.
Although the PreSERVE-AMI trial primary endpoint was not met, this failure does not negate the potential of CD34+ SCs to be an effective candidate for heart regeneration. Indeed, there are other instances in which SC trials face similar dilemmas in illustrating cell efficacy. Understanding these issues are key to interpreting the results of this study.
Factors to consider for interpretation of the PreSERVE-AMI trial results
1. Cell dose and cell source
In the study, post-hoc analysis favors a dose dependent response for improved LVEF and decreased scar. While general pharmacokinetics display a dose escalation response to a drug, such a response is not consistent for cell therapy. In fact, there are clinical trials that display higher response to lower cell doses14, possibly due to the detrimental effects of cells (pathogenic angiogenesis, obstruction). These studies illustrate that the ideal dose for SCs has yet to be elucidated.
The patient population demonstrated a wide range of harvested cells, (<20 × 106 to >60 × 106). Furthermore, autologous SCs may be encumbered with baseline comorbidities (diabetes, age-related deficiencies), and thereby are likely possess lower potency as compared to allogeneic cells. The important advantage of allogeneic cells is highlighted by the fact that 16 (8%) patients from this study did not meet release criteria after bone marrow aspiration. Allogeneic cells can be produced in a quality controlled and cost-effective manner, and represent an off-the shelf option7.
2. Timing of treatment
The time of cell infusion following stent placement was variable in this study, with a mean of 9.4 ± 1.43 and 9.3 ± 1.23 days for controls and treated patients, respectively. A meta-analysis of clinical trials utilizing adult bone marrow for the treatment of MI showed contradictory results in LVEF improvement secondary to timing of treatment, where later administration of cells proved more efficacious compared to early administration (<48 hours)9. Suppression of migration and proliferation in the SC niche is seen at times of excess inflammation (AMI). Cell therapy in the acute phase focuses on their anti-inflammatory and myocardial salvage traits; whereas, chronic treatment focuses primarily on regeneration capacities and reduction of adverse remodeling. The ideal timing post-transplantation for maximizing these effects has yet to be determined.
3. Route of administration
The optimal route of cell administration remains an area of uncertainty. Several methods are under investigation: trans-catheter endocardial, open-epicardial, intracoronary, intravenous and retrograde intra-coronary sinus. Despite these methods, the effects of SC treatment in AMI are limited. Without extracellular support (engineered tissue), evidence suggests that intramyocardial injection attains the highest number of retained cells despite their relatively low engraftment rate after an AMI15. As performed in this study, intracoronary injection is the preferred after an AMI because it avoids direct contact with the irritable myocardium thereby minimizing the risk of arrhythmia or perforation. The inherent disadvantage of intracoronary delivery is possible further occlusion of previously occluded arteries.
4. Trial size
Although the study was properly designed to power the primary efficacy endpoint, it is difficult to detect between group differences with limited sample sizes of Phase II trials. Furthermore, exploratory subgroup analyses, in this case, a dose dependent response of CD34+ cells, are likely underpowered and hypothesis generating. Utilizing LVEF as an endpoint may have also obscured between group differences. In the AMI setting, LVEF can be misleading, as the hypokinetic wall motion is a result of a heterogeneous mix of infarcted and stunned myocardium. The cell treated group had a longer total ischemic time which implies they had larger infarcts and potentially a harsher environment further depriving the tissue of potential endogenous or exogenous cell repair. These complex confounding factors cannot be simply corrected by multiple regression models. Scar size as measured by MRI is a better endpoint, since as mentioned above, the extent of the scar is an important predictor of mortality2.
Nonetheless, the number of larger Phase II and Phase III cell-based therapy trials for the treatment of heart disease trials is increasing, from 0 in 2014 and 2 in 2015, to 4 (including PreSERVE-AMI) in 2016. The Phase II CONCERT-HF (NCT02501811), Phase III DREAM HF-1 (NCT02032004) and BAMI (NCT01569178) trials are multicenter randomized trials currently enrolling an estimated 144, 600 and 3,000 patients, respectively.
Conclusion
Despite the failure to meet its endpoints, the PreSERVE-AMI trial ultimately represents an important step in the field of cardiac regeneration, by elucidating issues faced in the design of AMI trials. The hostile myocardial environment following AMI is a difficult hurdle to overcome for all progenitor cell types. This particular trial may have been affected by a number of unforeseen variables, including the use of autologous cells, which while immunotolerant, exhibit a decline in function with age and associated comorbidities (Online Figure). Autologous cell harvesting is also confounded by variable dosing, which may yield inconsistent results. Furthermore, the delay associated with the patients getting to the hospital for stenting means that time is an inherent ever-changing variable in an acute setting.
Despite the aforementioned variables, it is important to note that the use of CD34+ cells provide improvements in subjects when coronary oxygen demand exceeds its supply11. More importantly they proved to be safe in the current trial when compared to placebo. The safety of CD34+ cells will likely inspire further studies utilizing EPCs. The PreSERVE-AMI trial provides important insights regarding dosing of autologous CD34+ cells, time-to-treatment in an AMI setting, and safety. Moreover, the positive post-hoc analyses from this trial will undoubtedly lead to important new hypotheses to be tested in future trials.
Supplementary Material
Acknowledgments
Conflict of Interest Disclosures:
Dr. Hare discloses a relationship with Longeveron LLC (consulting) and Vestion Inc. (equity, board membership, and consulting). He is funded by the National Institutes of Health grants R01HL084275, R01HL107110, UM1HL113460, and R01HL110737; and grants from the Starr and Soffer Family Foundations.
References
- 1.Mozaffarian D, Benjamin EJ, Go AS, Arnett DK, Blaha MJ, Cushman M, Das SR, de Ferranti S, Despres JP, Fullerton HJ, Howard VJ, Huffman MD, Isasi CR, Jimenez MC, Judd SE, Kissela BM, Lichtman JH, Lisabeth LD, Liu S, Mackey RH, Magid DJ, McGuire DK, Mohler ER, 3rd, Moy CS, Muntner P, Mussolino ME, Nasir K, Neumar RW, Nichol G, Palaniappan L, Pandey DK, Reeves MJ, Rodriguez CJ, Rosamond W, Sorlie PD, Stein J, Towfighi A, Turan TN, Virani SS, Woo D, Yeh RW, Turner MB. Heart Disease and Stroke Statistics-2016 Update: A Report From the American Heart Association. Circulation. 2015 doi: 10.1161/CIR.0000000000000350. [DOI] [PubMed] [Google Scholar]
- 2.Wu KC, Weiss RG, Thiemann DR, Kitagawa K, Schmidt A, Dalal D, Lai S, Bluemke DA, Gerstenblith G, Marban E, Tomaselli GF, Lima JA. Late gadolinium enhancement by cardiovascular magnetic resonance heralds an adverse prognosis in nonischemic cardiomyopathy. Journal of the American College of Cardiology. 2008;51:2414–2421. doi: 10.1016/j.jacc.2008.03.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Bulluck H, Yellon DM, Hausenloy DJ. Reducing myocardial infarct size: challenges and future opportunities. Heart. 2016;102:341–348. doi: 10.1136/heartjnl-2015-307855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Simari RD, Pepine CJ, Traverse JH, Henry TD, Bolli R, Spoon DB, Yeh E, Hare JM, Schulman IH, Anderson RD, Lambert C, Sayre SL, Taylor DA, Ebert RF, Moye LA. Bone marrow mononuclear cell therapy for acute myocardial infarction: a perspective from the cardiovascular cell therapy research network. Circulation research. 2014;114:1564–1568. doi: 10.1161/CIRCRESAHA.114.303720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Hare JM, Traverse JH, Henry TD, Dib N, Strumpf RK, Schulman SP, Gerstenblith G, DeMaria AN, Denktas AE, Gammon RS, Hermiller JB, 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]
- 6.Ledney GD, Stewart DA, Gruber DF, Gelston HM, Jr, Exum ED, Sheehy PA. Hematopoietic colony-forming cells from mice after wound trauma. J Surg Res. 1985;38:55–65. doi: 10.1016/0022-4804(85)90010-1. [DOI] [PubMed] [Google Scholar]
- 7.Sanina C, Hare JM. Mesenchymal Stem Cells as a Biological Drug for Heart Disease: Where Are We With Cardiac Cell-Based Therapy? Circ Res. 2015;117:229–233. doi: 10.1161/CIRCRESAHA.117.306306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.van der Spoel TI, Jansen of Lorkeers SJ, Agostoni P, van Belle E, Gyongyosi M, Sluijter JP, Cramer MJ, Doevendans PA, Chamuleau SA. Human relevance of pre-clinical studies in stem cell therapy: systematic review and meta-analysis of large animal models of ischaemic heart disease. Cardiovasc Res. 2011;91:649–658. doi: 10.1093/cvr/cvr113. [DOI] [PubMed] [Google Scholar]
- 9.Afzal MR, Samanta A, Shah ZI, Jeevanantham V, Abdel-Latif A, Zuba-Surma EK, Dawn B. Adult Bone Marrow Cell Therapy for Ischemic Heart Disease: Evidence and Insights From Randomized Controlled Trials. Circ Res. 2015;117:558–575. doi: 10.1161/CIRCRESAHA.114.304792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Sekiguchi H, Ii M, Losordo DW. The relative potency and safety of endothelial progenitor cells and unselected mononuclear cells for recovery from myocardial infarction and ischemia. J Cell Physiol. 2009;219:235–242. doi: 10.1002/jcp.21672. [DOI] [PubMed] [Google Scholar]
- 11.Losordo DW, Henry TD, Davidson C, Sup Lee J, Costa MA, Bass T, Mendelsohn F, Fortuin FD, Pepine CJ, Traverse JH, Amrani D, Ewenstein BM, Riedel N, Story K, Barker K, Povsic TJ, Harrington RA, Schatz RA Investigators AC. Intramyocardial, autologous CD34+ cell therapy for refractory angina. Circ Res. 2011;109:428–436. doi: 10.1161/CIRCRESAHA.111.245993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Quyyumi AA, Vasquez A, Kereiakes D, Klapholz M, Schaer GL, Abdel-Latif A, Frohwein S, Henry TD, Schatz RA, Dib N, Toma C, Davidson CJ, Barsness GW, Shavelle D, Cohen M, Poole J, Moss TJ, Hyde P, Kanakaraj A, Druker V, Chung A, Junge C, Preti RA, Smith RL, Mazzo DJ, Pecora A, Losordo DW. PreSERVE-AMI: A Randomized, Double-Blind, Placebo-Controlled Clinical Trial of Intracoronary Administration of Autologous CD34+ Cells in Patients with Left Ventricular Dysfunction Post STEMI. Circ Res. 2016 doi: 10.1161/CIRCRESAHA.115.308165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Quyyumi AA, Waller EK, Murrow J, Esteves F, Galt J, Oshinski J, Lerakis S, Sher S, Vaughan D, Perin E, Willerson J, Kereiakes D, Gersh BJ, Gregory D, Werner A, Moss T, Chan WS, Preti R, Pecora AL. CD34(+) cell infusion after ST elevation myocardial infarction is associated with improved perfusion and is dose dependent. Am Heart J. 2011;161:98–105. doi: 10.1016/j.ahj.2010.09.025. [DOI] [PubMed] [Google Scholar]
- 14.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, George R, Lardo A. 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]
- 15.Terrovitis J, Lautamaki R, Bonios M, Fox J, Engles JM, Yu J, Leppo MK, Pomper MG, Wahl RL, Seidel J, Tsui BM, Bengel FM, Abraham MR, Marban E. Noninvasive quantification and optimization of acute cell retention by in vivo positron emission tomography after intramyocardial cardiac-derived stem cell delivery. J Am Coll Cardiol. 2009;54:1619–1626. doi: 10.1016/j.jacc.2009.04.097. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.