Regenerative Cell-Based Therapy for the Treatment of Cardiac Disease


Heart failure (HF) is a burgeoning disease. Recently published American Heart Association projections indicate that its prevalence will increase by 46% from 2012 to 2030 resulting in approximately 8 million people with HF in the United States alone. Ischemic heart disease is the major contributor to HF and despite best practices and management strategies, many patients are left with significant disability. Events such as myocardial infarction can result in the irreparable loss of viable cardiac mass and in this and many other clinical scenarios, save for transplantation, no therapies currently exist to remedy lost myocardium. Considering the shortage of donor hearts, a rationale to develop regenerative stem cell based therapies is provided.

Thousands of clinicians and investigators worldwide are working toward harnessing the properties of adult stem or progenitor cells to treat injury in tissues and organs that have limited intrinsic capacity for repair and regeneration, such as the brain and the heart. Yet, achieving cellular cardiomyoplasty remains elusive. As a basic definition, cell therapy for regeneration is the therapeutic use of a cell population to promote growth and repair of diseased and / or damaged tissue to restore anatomy and function. The focus tends to be, typically, in settings where this does not occur appreciably such as myocardial infarction and traumatic brain or spinal cord injury. In the setting of ischemic cardiomyopathies, the goal of stem cell therapy would, preferably, be a combination of promoting the growth of new or existing blood vessels to improve tissue perfusion, and the growth of new cardiomyocytes within the damaged myocardium. Since its inception, cell therapy has achieved the most success in facilitating new blood vessel formation via either angiogenesis or vasculogenesis. Unfortunately, it has been somewhat more difficult to restore lost cardiomyocytes. Although we are likely still decades away from completely regenerating the diseased myocardium, recent evidence indicates that we are at least a little farther ahead. The aim of this chapter is to familiarize readers with the field of cell therapy for cardiac regeneration. Despite many deserving preclinical and clinical studies, unfortunately this review will not be comprehensive, because of length restrictions. Instead, only those cell types with the most promise and best available evidence will be discussed. The clinical focus will be acute MI where the loss of cardiomyocytes is a hallmark of this disorder; however, stem cell use in other clinical entities will also be briefly touched upon. Methods of administration will be covered, as well as the principal mechanisms of action that have been identified. Finally, a brief section will outline some of the current challenges facing the field and possible future directions. At the end of this chapter readers will be familiar with the field of cell therapy for cardiac repair and for those interested references are provided.

Delivery Methods for Cell Transplantation

Stem cell products for the injured heart can be administered through several methods—some more invasive than others—and each presenting with unique advantages and disadvantages ( Table 101-1 ). Generally, stem cells can be infused intravenously or via patent coronary arteries supplying the ischemic region of the heart, or stem cells can be injected directly into the ventricular wall via a percutaneous transendocardial or a surgical transepicardial approach ( Fig. 101-1 ).

TABLE 101-1
Administration Models
Routes Advantages Disadvantages
Intravenous injection Simplest Few migrate into the target
Intracoronary injection Can be administered into patent artery, higher cell homing versus intravenous infusion, low risk of arrhythmia Unsuitable for larger stem cells, hinder cellular transmigration, exacerbation of atherosclerosis
Intramyocardial injection Most reliable under direct visualization; highest cell dose Invasive, potential for cell “clumping”

FIGURE 101-1, Delivery routes for transplanted cells for the treatment of ischemic heart disease.

Intravenous Infusion

Intravenous infusion is the simplest method of cell administration. Given this delivery strategy is nonspecific, it relies heavily on proper homing of the infused cells to the ischemic myocardium. Thus, the disadvantage of this method is that a low percentage of the infused cells home it to the intended target. Cells can also become trapped in other organs, particularly in lymphoid tissues, so that an additional small proportion of cells enters the coronary circulation and migrates into ischemic myocardium. This is particularly disadvantageous with pluripotent cells increasing the opportunity for off-site proliferation. In addition, with larger cell types, such as mesenchymal stem cells (MSCs) or skeletal myoblasts, microembolization can occur because these cells may be incapable of penetrating beyond a given capillary bed, making them unlikely to reach their target.

Intracoronary Infusion

Infusing stem cells via a patent coronary artery can be performed at the time of selective coronary angiography, and it is well-suited for the specific delivery of cells to the ischemic territory of the myocardium. The obvious advantage of intracoronary infusion is that cells can travel directly into myocardial regions in which nutrient-rich blood flow and oxygen supply are preserved, ensuring a favorable environment for the survival of cells, a prerequisite for stable engraftment. Indeed, in a study performed by Hofmann and colleagues, homing of unselected bone marrow cells to the infarct region was apparent only after intracoronary delivery but not after intravenous infusion. One disadvantage; however, is that high coronary flow after stem cell injection can prevent low-affinity adhesion to the myocardial capillaries, thus hindering cellular transmigration into the infarcted zone. Another disadvantage to delivering stem cells in this fashion to a potentially diseased segment of the coronary artery is that the combination of stem cells and the proangiogenic milieu of the plaque may serve to exacerbate macrovascular and microvascular disease. Similar to intravenous delivery, intracoronary infusion of cells might not be suitable for certain types of larger stem cells, such as skeletal myoblasts and MSCs, which can be prone to embolization. Despite some of these limitations, intracoronary injection of stem cells has been the preferred method of delivery in most clinical studies thus far.

Intramyocardial Injection

Intramyocardial injections constitute an invasive approach to delivering therapeutic cell products compared with the other methods mentioned. This approach is performed by delivering cell suspensions into target myocardial areas via direct injection during minimally invasive thoracoscopic or open procedures. It can be the preferred method of delivery particularly when there is a lack of patent coronary arteries supplying the ischemic region or when the procedure is performed in tandem with mechanical revascularization. The surgical injection process is simple and can be performed under direct visualization, allowing evaluation by inspection of the potential target zones. As an alternative to user-based visualization of cell injection, electromechanical mapping (e.g., using the NOGA system) can serve to improve the reliability of surgical transepicardial injections, or assist transendocardial injections as a percutaneous procedure. Multiple injections of concentrated cell suspensions at minimal volumes appear preferable to less frequent, larger volume injection. This method is feasible and potentially the most reliable in ensuring stem cells reach the intended or injured areas of the myocardium. Regardless, it is still difficult to predict the survival and function of progenitor cells injected into uniformly necrotic tissue, and the ischemic or necrotic area may prove too hostile to ensure proper cell engraftment and survival.

There are disadvantages to intramyocardial injections. Some nonspecific delivery can occur often because of cell leakage during injection. Moreover, cell clumping can prevent uniform cell distribution in the target area. This method of delivering stem cells may also be inappropriate in situations of diffuse disease such as nonischemic dilated cardiomyopathy where focal deposits of directly injected cells might be poorly matched to the underlying pathophysiology.

Summary

The optimal route for administering stem cells has yet to be determined, although the method of choice will likely be specific depending on a number of factors such as the cell product, patient clinical characteristics, and disease setting. In more acutely ischemic scenarios such as acute myocardial infarction (AMI), the release of chemotactic stimuli into the peripheral circulation, in response to the ischemic injury, may favor homing of intravenous or intracoronary infused cells. However, in situations in which these signals have been abated, as may be the case for chronic ischemia or old scar, injection of the cells directly into the cardiac muscle may produce a more favorable outcome. To date, most trials report a low frequency of complications regardless of route of administration associated with cell base therapies supporting the safety of any of these approaches.

Adult STEM or Progenitor Cells for Cardiac Cell Therapy

A number of adult stem or progenitor cell types have been investigated for the treatment of heart disease ( Fig. 101-2 ). They have been procured from the bone marrow (bone marrow–derived mononuclear cells [BMCs]), the peripheral circulation, adipose tissue, skeletal muscle, or cardiac muscle biopsy specimens. Pluripotent cells such as the embryonic stem cell (ESC) or the inducible pluripotent stem cell (IPSC) have also received much attention. Most have been investigated extensively in preclinical models, and some in the clinical arena. Some emerging cell types, such as the IPSC, have not yet reached clinical trial. The most pertinent cell types, either for emerging relevance or historical importance, are discussed in the following sections, and a summary is provided in Table 101-2 . No consensus has been reached on the ideal cell type; however, some candidates appear more promising than others. If the ideal cell type does exist, it should fulfill most if not all the following criteria :

  • 1

    Stem cells should be safe. In other words, they should not form tumors, predispose patients to fatal arrhythmias, or elicit immune reactions.

  • 2

    Stem cells should improve heart function, quality of life, and mortality.

  • 3

    Stem cell harvest and culture methods should be clinically feasible and not cost-prohibitive.

  • 4

    Stem cells should be available as a standard, “off-the-shelf” product.

  • 5

    Stem cell use should not be controversial or ethically ambiguous.

FIGURE 101-2, Cell types currently undergoing clinical trial and emerging cell types. EPC, Endothelial progenitor cell; HSC, hematopoietic stem cell.

TABLE 101-2
Cell Types
Cell Type Phenotypic Markers Advantages Disadvantages
Skeletal myoblasts CD56, Desmin Easily harvested and expanded; good survival rates; differentiate into myotubes Poor differentiation into cardiomyocytes; high risk for arrhythmias
Embryonic stem cells CD133, Oct 4, SSEA-3/4, Sox Differentiate into cardiomyocytes, endothelial cells, and smooth muscle cells Ethical challenges, tumorigenicity, ectopic differentiation, immunogenicity
Bone marrow–derived stem cells
Hematopoietic stem cells CD34, CD133, CD45, Sca-1, c-kit Improve cardiac function, promote angiogenesis Mechanistically, may only reconstitute peripheral blood leukocytes
Endothelial stem cells CD34, CD133, VEGFR2, CD144, Sca-1 Increase blood vessel formation, prevent cardiomyocyte apoptosis Variable efficacy
Mesenchymal stem cells CD106, c-kit Have reduced immunogenicity (at least initially), easy to expand and maintain ex vivo; robust secretome Large cell type
Adipose tissue–derived cells Less expensive, less invasive to harvest, and potentially available in great quantities Isolation needs to be optimized for clinical use
Side population cells CD45, CD59, CD43, CD49d, CD31, and integrin markers Differentiate into cardiomyocytes and endothelial cells Little if any preclinical data
Inducible pluripotent cells Oct4, Sox2, Klf4, c-Myc, Lin28, Nanog (introduced) Yet to be tested
Cardiac stem cells c-kit, Sca-1, isl 1 May generate cardiomyocytes; strong paracrine repertoire Very low numbers, isolation difficult; limited clinical experience

The following sections will cover the predominant cell types emphasizing clinical data. For emerging cell products that have not yet reached clinical trial, animal studies instead will be the focus. A summary of select clinical trials is provided in Table 101-3 (note that this table is not exhaustive, but is provided as an example).

TABLE 101-3
Select Clinical Trial Results
Study Cell Therapy Patient Cell Type and Cell Number Route Follow-up Results
TOPCARE-AMI AMI (n = 29)
AMI (n = 30)
2.1 × 10 8 BMCs
1.6 × 10 7 CPCs
IC 1 yr LVEF↑; perfusion↑; remodeling↓
BOOST AMI (n = 30) 2.4 × 10 9 BMCs IC 5 yr LVEF↑ (6 months only), no change vs. control at 5 years
Fernandez-Aviles et al AMI (n = 20) 7.8 × 10 7 BMCs IC 11 mo LVEF↑; ESV↓;
MAGIC AMI (n = 10) 1.5 × 10 9 G-CSF-mobilized cells IC 6 mo LVEF↑; ESV↓; regional perfusion↑
Choi et al AMI (n = 10) 2 × 10 9 G-CSF-mobilized cells N/A 6 mo LVEF↑
Ge et al AMI (n = 10) 4 × 10 7 BMC IC 6 mo LVEF↑; perfusion↑
Strauer et al AMI (n = 10) 2.8 × 10 7 BMCs IC 3 mo Infarct size↓; LV perfusion↑
Chen et al AMI (n = 34) 4.8-6 × 10 10 BMCs IC 6 mo LVEF↑; perfusion↑; wall motion↑
REPAIR-AMI AMI (n = 102) BMCs (50 mL bone marrow) IC 1 yr LVEF↑; infarct size↓; ↓remodeling
Janssens et al AMI (n = 33) 3 × 10 8 BMSCs IC 4 mo Infarct size↓; regional systolic function↑
Li et al AMI (n = 35) 7.3 × 10 7 PBSCs IC 6 mo LVEF↑; wall motion↑
ASTAMI AMI (n = 50) BMCs (50 mL bone marrow) IC 6 mo No benefit
TOPCARE-CHD CMI (n = 28)
CMI (n = 24)
2.1 × 10 8 BMCs
2.2 × 10 7 CPCs
IC 3 mo Wall motion↑
Katritsis et al CMI (n = 11) 2-4 × 10 6 BMCs and EPCs IC 4 mo Perfusion defects↓; infarct size↓
Perin et al HF (n = 14) 3 × 10 7 BMCs IM 4 mo LVEF↑; regional perfusion↑
Stamm et al CMI (n = 20) 5 × 10 6 AC133 + BMCs IM 6 mo LVEF↑; perfusion↑
Ahmadi et al ICM (n = 19) N/A IM 6 mo WMSI↓
Hendrikx et al CMI (n = 10) 6 × 10 7 BMCs IM 4 mo LVEF↑; systolic thickening↑; Defect score↓
Losordo et al RA (n = 18) 5-50 × 10 4 CD34 + cells/kg IM 6 mo Exercise tolerance↑; perfusion↑
Mocini et al ICM (n = 18) 2.9 × 10 8 BMCs IM 3 mo LVEF↑; wall motion↑
Tse et al ICM (n = 8) BMCs (from 40 mL bone marrow) IM 3 mo LVEF↑; wall motion↑
G-CSF-STEMI AMI (n = 44) G-CSF mobilized cells N/A 1 yr ↑Perfusion 1 week and 1 month post treatment
STEMMI AMI (n = 78) G-CSF mobilized cells N/A 6 mo No improvement, some inverse association between circulating mesenchymal cells and systolic recovery
REVIVAL-2 AMI (n = 56) G-CSF mobilized cells N/A 4-6 mo No improvement
FINCELL AMI (n = 40) BMCs (360 × 10 6 ) IC 6 mo ↑LVEF
Cao et al AMI (n = 41) BMCs (5 × 10 7 ) IC 4 yr ↑LVEF
Hare et al AMI (n = 53) Allogeneic MSCs (0.5-5 × 10 6 cell/kg) IC 6 mo ↑LVEF and global symptom score in patients with anterior MI
REGENT AMI (n = 80) BMCs or CD34 + CXCR4 + selected cells (1.78 × 10 8 cells; 1.90 × 10 6 cells) IC 6 mo ↑LVEF in patients with baseline EF < 37%
BALANCE AMI (n = 62) BMCs (6.1 × 10 7 cells) IC 5 yr ↑contractility; ↑hemodynamic status; ↑exercise capacity; ↓mortality
SCIPIO HF (n = 16) c-kit+ CSCs (1 × 10 6 cells) IC 1 yr ↑LVEF; ↓infarct size
HEBE AMI (n = 200) BMCs or PBMCs (≈290 × 10 6 ) IC 4 mo No improvements
Losordo et al RA (n = 167) CD34+ selected BMCs (1-5 × 10 5 cells/kg) NOGA IM 1 yr ↓weekly angina frequency; ↑exercise tolerance
Quyyunmi et al AMI (n = 16) CD34+ selected BMCs (10 × 10 6 cells) IC 6 mo ↑perfusion; ↑LVEF; ↓infarct size
BONAMI et al AMI (n = 49) BMCs (98 × 10 6 cells) IC 3 mo ↑myocardial viability; ↓non-viable segments
Williams et al ICM (n = 8) BMCs or MSCs (100-200 × 10 6 cells) IM 1 yr ↑regional contractility; ↓end-diastolic volumes; ↓infarct sizes
Chih et al RA (n = 18) G-CSF mobilized cells N/A 42 wk No improvements
POSEIDON ICM (n = 5) Allogeneic and autologous MSCs (20-, 100-, 200- × 10 6 cells) IC 1 yr ↑functional capacity; ↑quality of life; ↓ventricular remodeling
TAC-HFT ICM (n = 65) MSCs and BMCs IC 1 yr ↓MLHFQ both cell types
↑6MWT; ↑regional function; ↓infarct sizes MSCs only
CADUCEUS AMI (n = 31) CDCs (25 × 10 6 cells) IC 6 mo ↓scar mass; ↑viable heart mass; ↑regional contractility; ↑regional systolic wall thickening
Perin et al HF (n = 10) ALDH+ BMCs (2.27 × 10 6 ) NOGA IM 6 mo ↓LV end-systolic volume
SWISS-AMI AMI (n = 200) BMCs IC 4 mo No improvements
TIME AMI (n = 65) BMCs (150 × 10 6 cells) IC 1 yr No improvements
AMI, Acute myocardial infarction; BMC, bone marrow--derived mononuclear cell; CDC, cardiosphere derived cells; CMI, chronic myocardial infarction; CPC, circulating progenitor cell; CTO, chronic total occlusion; EF, ejection fraction; EPC, endothelial progenitor cell; ESV, end-systolic volume; G-CSF, granulocyte colony-stimulating factor; HF, heart failure; IC, intracoronary; IM, intramyocardial; LV, left ventricular; MSC, mesenchymal stem cell; RA, refractory angina pectoris.

Bone Marrow–Derived Cells

The bone marrow hosts a myriad of cell populations, including hematopoietic and nonhematopoietic cells and progenitors. Interest in autologous BMCs for the treatment of ischemic cardiac syndromes was first stimulated by the discovery of the putative endothelial progenitor cell (EPC) by Asahara and colleagues in 1997. These EPCs, originally from the bone marrow, were found to circulate in the peripheral blood homing to sites of tissue ischemia to participate in postnatal vasculogenesis. Since then, cells derived from the bone marrow have been the preferred cell type in both preclinical and clinical cardiac cell therapy studies. BMCs can be harvested via either bone marrow aspiration or peripheral blood apheresis followed by isolation of the mononuclear cell fraction by density-gradient ultracentrifugation. Upon isolation of the mononuclear fraction, BMCs can either be kept unfractionated or be further purified to isolate subpopulations identified by specific surface antigenic expression. In some cases, depending on desired cell type, fractionated BMCs may require further ex vivo expansion in culture. The subpopulations of BMCs that have been studied in the context as cardiac regenerative therapies includes hematopoietic stem cells (HSCs), EPCs, MSCs, and purified CD133 + or CD34 + cells (see Fig. 101-2 ). Bone marrow products have gained much favor from investigators mainly because of their ease of procurement and expansion in culture, and they tend to possess a relatively high concentration of adult progenitors.

Unfractionated Bone Marrow Cells

The capacity for unselected or unfractionated BMCs to promote cardiac repair has been controversial, yet they remain the most thoroughly investigated cell type in clinical trial. Their popularity is driven largely by practical reasons. For example, BMCs are (1) derived from straightforward harvest protocols, (2) do not require complex or prolonged ex vivo management, and (3) contain a small niche of stem and progenitor cells despite representing a heterogeneous population. BMCs have been investigated in AMI, chronic ICM, and HF, but for the sake of brevity the following section will focus on trials of AMI. Readers interested in the results of BMC therapy in other clinical settings can consult several excellent reviews.

The safety and feasibility of BMC therapy was established from early, small trials of AMI such as the “Transplantation of Progenitor Cells and Regeneration Enhancement in Acute Myocardial Infarction” (TOPCARE-AMI). The demonstration that cell therapy may be safe, and possibly effective, prompted further inquiry into mid-sized, double-blind, randomized trials such as the “Reinfusion of Enriched Progenitor Cells and Infarct Remodelling in Acute Myocardial Infarction” (REPAIR-AMI) and the “BOne marrow transfer to enhance ST-elevation infarct regeneration” (BOOST) trials. Initial follow-up analysis from both the REPAIR-AMI and BOOST trials reported benefits associated with BMC therapy. BMCs were infused 3-7 days after successful reperfusion and were associated with superior cardiac function (5.5%-6.7% improved left ventricular ejection fraction [LVEF]) compared with placebo at 6 months postintervention. In addition, cell therapy was not associated with an increased risk for adverse clinical events. Substudies of both trials were subsequently released by trial investigators with equally promising results. For example, patients receiving BMCs in BOOST demonstrated improvements in diastolic function, whereas there was a reduction in a prespecified cumulative end-point of death, MI, or necessity for revascularization at 1-year follow-up of the REPAIR-AMI. Several other trials also support the benefit of BMC therapy in patients with AMI. Trials by Assmus and colleagues, Cao and colleagues, Yousef and colleagues, and the “FINish stem CELL study” (FINCELL) reported positive benefits associated with BMC therapy including improved global ventricular function, a reduction in morbidity and mortality, and a neutral effect on severe adverse reactions, including arrhythmias and restenosis of the stented coronary artery.

Unfortunately, despite the positive early experience with BMCs in patients, other trials have not been so promising. Moreover, follow-up from some of the earlier, positive trials were beginning to release disappointing long-term results. For example, the 2-year results of the REPAIR-AMI could not conclusively associate BMC therapy with improved morbidity and mortality ; in BOOST, whereas the difference in LVEF improvement between cell therapy and placebo were significant at 6-month follow-up, the effect was not sustained at 18 months. A subgroup of patients with pronounced transmural infarcts may have benefited from cell therapy, although this was required to be tested prospectively in a randomized trial. Regardless, the lack of beneficial effects associated with BMCs in BOOST was sustained in the 5-year results. Examples of results from recent trials include the “BONe marrow in Acute Myocardial Infarction” (BONAMI), which have been mixed, and those from the “Autologous Stem-Cell Transplantation in Acute Myocardial Infarction” (ASTAMI) trial, which were altogether negative. Aside from a small improvement in exercise time, ASTAMI did not associate BMCs with improvements in global LV function, or ventricular remodelling.

Before reaching clinical trial, many elements such as cell dose, frequency, or time of delivery had not been systematically determined for stem cell therapies. In some trials, such as the REPAIR-AMI, only until patients were stratified based on time of delivery after reperfused MI did some suggestion arise that timing of delivery influenced the efficacy of the therapy. This prespecified subgroup analysis associated later cell delivery (about 7 days after reperfused MI) with more pronounced benefits. Thus, several recent trials including the TIME, late-TIME, and SWISS-AMI sought to test this hypothesis comparing the time of delivery between cells delivered early, mid, or late postinfarction. Unfortunately, results from all three trials did not associate beneficial effects with BMC therapy for AMI, consistent with the BOOST and ASTAMI trials, regardless of when cells were delivered after AMI. Results from the TIME trial may, however, require careful scrutiny as cells were procured from a novel, convenient extraction method that had not been previously validated in bioactivity assays or animals models as opposed to the already well-supported and published methods for isolation.

Establishing a concrete position on the efficacy of BMC therapy for AMI has so far been difficult and results from recent trials such as TIME, late-TIME, or SWISS-AMI have not been able to answer this question conclusively. The outlook for BMC therapy is less than optimistic. Given the conflicting evidence and emerging alternatives, favor may lean toward products with well-demonstrated capacity to produce improvements in global LV function, mortality, and quality of life. Moreover, although some cumulative evidence from recent meta-analyses suggests that BMCs are associated with modest improvements in LV function (2%-4% LVEF), the question remains of whether these improvements are clinically meaningful. The upcoming European large, phase 3 “Effect of Intracoronary Reinfusion of Bone Marrow–derived Mononuclear Cells on All-Cause Mortality in Acute Myocardial Infarction” (BAMI) trial ( NCT01569178 ) will probably represent the final consensus of BMCs in cardiac cell therapy in AMI.

Hematopoietic Stem Cells

Hematopoietic stem cells (HSCs) are a subset of BMCs that are multipotent and self-renewing and from which all blood cell types are derived. They are possibly the first stem cell therapeutic investigated in patients dating back from early trials of bone marrow transplantation for fatal leukemias. Their use can be potentially curative for genetic blood disorders such as thalassemia and immune deficiency, or malignancies such as leukemia. The prevailing hypothesis is that HSCs give rise to two lineage restricted progenitor cell types, including myeloid progenitors and lymphoid progenitors that, in turn, supply the number of lymphocytic and granulocytic cells, macrophages, platelets, and erythrocytes found in the blood. However, specific identification and classification of HSCs has been difficult largely because of the limitations of current imaging and lineage tracing methods. Generally, HSCs are identified by a lack of hematopoietic or myeloid lineage (lin) markers and, depending on the report, the presence of a number of antigenic surface markers such as CD34, CD38, CD90, CD133, CD105, CD45, and c-kit (CD117) or other markers such as the presence of aldehyde dehydrogenase. Their use as a cardiac regenerative cell product was stimulated by early studies indicating that lin , c-kit + HSCs could restore necrotic myocardium by transdifferentiating into all the major cardiac cell types (cardiomyocyte, endothelial cell, and smooth muscle cell) conferring salutary effects. Unfortunately, this notion was met with sharp criticism and conflicting data. Subsequent evidence instead indicates that HSCs are, in fact, not cardiomyogenic but adopt mature hematopoietic fates in ischemic myocardium. Whether the benefits of HSCs are derived from an ability to restore damaged myocardium or from a separate, possibly paracrinic, mechanism remains a topic of intense debate.

Consensus on what combination of surface, cytosolic, or functional markers define the HSC is lacking, thus making direct comparisons between trials difficult. Given this loose definition of HSCs, some of the trials discussed in the following section might apply equally to other BMC products, and vice versa. The MAGIC trial assessed the feasibility and efficacy of HSC infusion in patients with MI and who underwent coronary stenting. Granulocyte colony-stimulating factor (G-CSF), a well-established stem-cell mobilizer, was administered to mobilize bone marrow–derived HSCs into the peripheral blood 4 days before apheresis and cell procurement. This trial involved three separate arms including the HSC group, a control group (not placebo), and patients receiving G-CSF alone. At 6 months after infusion, patients treated with HSCs showed significant improvements in exercise capacity (450 vs. 578 seconds), myocardial perfusion (perfusion defect, 11.6% vs. 5.3%), and systolic function (LVEF, 48.7% vs. 55.1%). Although no adverse events occurred because of cell infusion combined with G-CSF, enrollment of the G-CSF cohort was eventually discontinued because of high rates of in-stent restenosis, highlighting a potential safety concern and cautioning future trials using G-CSF administration as a strategy for endogenous stem cell mobilization. Recent trials, though, such as Perin and colleagues have not reported any significant adverse cardiovascular or cerebrovascular events associated with HSC infusion in similarly high-risk patient cohorts with advanced ischemic heart failure and CAD ineligible for PCI or surgical revascularization. The difference, perhaps, is that Perin and colleagues elected to aspirate bone marrow from the iliac crest as opposed to administering G-CSF. In their trial, HSCs were then purified based on the presence of cytosolic ADH, constituting a population of primitive Lin CD34 + CD38 cells. When administered to patients with advanced ischemic heart failure ADH + HSCs were associated with a significant decrease in left-ventricular end-systolic volume and a trend toward maximal oxygen consumption at 6 months. It is difficult to draw meaningful conclusions given that only 20 patients were enrolled in the study. Overall, HSCs appear to be beneficial for the treatment of ischemic cardiomyopathies, but larger, randomized, controlled trials are required to confirm early results.

Endothelial Progenitor Cells

Before the characterization of a circulating EPC in 1997 by Asahara and colleagues, vasculogenesis, the process of de novo blood vessel development, was believed to be limited to the embryonic development of vasculature from endothelial cell progenitors or angioblasts. Now it is understood that a subset of bone marrow cells, possibly the putative EPC, mobilizes from the bone marrow to the peripheral circulation ultimately reaching sites of tissue injury or ischemia to participate in postnatal vasculogenesis. Circulating EPC levels have also been recognized as possibly providing clinical information on atherosclerotic burden and future cardiovascular risk. Despite the significant attention that has been devoted to studying EPC biology, consensus on an unambiguous definition of the EPC is still lacking and, likewise, the success of many studies has been hindered by the absence of such a definition. EPCs were first characterized as a population of cells that were CD34 + , VEGFR2 + , and adherent to fibronectin ( Fig. 101-3 A ). Today, they are either broadly identified as the mononuclear cells adherent to fibronectin or by cell surface expression that may or may not include CD31, FLK-1 (VEGFR2), CD34, CD133 (AC133), c-kit, and other endothelial markers including VE-cadherin. The criteria that distinguish an HSC from an EPC, and vice versa, may seem unclear given they seem to share many of the same surface antigenic markers. As a cell therapy product, however, EPCs are distinguished from HSCs based on specific ex vivo culture requirements. Separate techniques exist to produce what can be referred to as the early or late EPC . Both early and late EPCs first require isolating the mononuclear cell fraction from the peripheral circulation, and early EPCs are generated through subsequent culture on fibronectin for 3 to 7 days, whereas late EPCs are produced via culture for greater than 14 days on collagen type I. For a more detailed discussion on the characterization of EPC phenotypes or potential diagnostic use of circulating EPCs, interested readers are referred to an excellent review by Fadini and colleagues.

FIGURE 101-3, Endothelial progenitor cells (EPCs) and mesenchymal stem cells (MSCs) in culture. A, EPCs from the peripheral blood mononuclear cell layer, cultured on fibronectin for 4 days. Adherent cells (CD34 +/– , VEGFR-2 + ) typically take up the markers lectin and DiI-Oxidized Low Density Lipoprotein. B, MSCs cultured from human bone marrow yielding a population of adherent MSCs that characteristically express the antigens CD29 and CD44 but are negative for the hematopoietic cell marker CD45.

In preclinical animal models of ischemic cardiomyopathy EPC transplantation prevented cardiac myocyte apoptosis, resulted in increased blood vessel formation, decreased infarct size, and improved function. The capacity for human EPCs to transdifferentiate into cardiomyocytes has also been reported, but not all studies support this conclusion. Instead, recent evidence suggests that human early EPCs may be proangiogenic, enacting their salubrious properties through paracrine mechanisms. The proangiogenic effects of late EPCs, instead, is thought to be via direct incorporation into the vasculature.

Considering the attention EPCs have received in preclinical animal models of myocardial ischemia, there is comparatively little experience with their use in patients. This is likely owing to pragmatic considerations and investigator preference for unfractionated or sorted BMC products neither requiring ex vivo culture. EPC cell therapy has been explored in several settings, including AMI and chronic ischemic heart disease. As a therapy delivered shortly after reperfused AMI, the small TOPCARE-AMI trial reported that EPC transplant was safe, feasible, and potentially beneficial. EPCs were infused approximately 4 to 5 days after MI and were associated with improved LVEF (51.6 ± 9.6% to 60.1 ± 8.6%), improved regional wall motion in the infarct zone, and profound reduction in end-systolic left-ventricular volumes at 4-month follow-up, with benefits persisting in the final 1-year follow-up. Indeed, contrasting results were released by the same group in a similar trial assessing EPC cell therapy for the treatment of chronic ischemic heart disease. The randomized, controlled TOPCARE-CHD trial assessed the efficacy of BMC, EPC, or placebo treatment later after MI in patients who had a myocardial infarction at least 3 months before enrollment and a well-demarcated region of left ventricular dysfunction. Cells were infused into the vessel supplying the most dyskinetic area of the ventricle, and the results of this study contradict those of TOPCARE-AMI, as EPC delivery was significantly inferior to BMC therapy in terms of improving global left ventricular function. The absolute improvement witnessed with BMC therapy, similar to their experience in AMI, was modest. BMC therapy was associated with an increase in approximately 2.9 percentage points according to left ventricular angiography and 4.8 percentage points according to MRI. The precise mechanisms underlying the benefit of BMC therapy were not investigated in the trial, and it remains unclear as to why EPCs proved inferior in this setting. A higher number of progenitors isolated from the bone marrow aspiration may account for this discrepancy. Moreover, given that patients with CHD have been shown to have pronounced EPC dysfunction, limited cell recruitment and action at the site of injury may have also contributed.

Fractionated CD34 + Bone Marrow Cells

As an alternative to heterogeneous cell products, in recent years focus has shifted to BMCs isolated based on CD34 expression given their potent proangiogenic properties. CD34 is believed to be a cell-to-cell adhesion factor expressed in some cell types, including hematopoietic cells, endothelial cells, muscle satellite cells, hair follicle stem cells, and fibrocytes. The function of CD34 remains largely unknown, although in HSCs some evidence suggests that CD34 alters engraftment potential and other properties, such as cell adhesion and migration. Peripheral blood CD34 + cells have been shown to transdifferentiate into most of the major cardiac lineages, including endothelial cells, vascular smooth muscle cells, and cardiomyocytes, principally via cell fusion. CD34 + cells purified from peripheral blood BMCs have been shown to exhibit superior efficacy and safety for therapeutic neovascularization after MI compared with total mononuclear cells in rodent studies. They have also been shown to exhibit superior efficacy for preserving integrity and function following MI compared with unselected mononuclear cells with an equivalent number of CD34 + cells, suggesting that a purified CD34 + cell product might possess greater therapeutic potential than a mixed mononuclear cell population.

The REGENT trial investigated the therapeutic potential of fractionated CD34 + mononuclear cells for the treatment of significant systolic dysfunction (LVEF < 37%) secondary to reperfused anterior MI. Two hundred patients were randomized to intracoronary infusion of CD34 + CXCR4 + purified cells, total mononuclear cells, or a control group receiving no therapy at all (no placebo) and were followed for 6 months. Similar to REPAIR-MI, both the purified CD34 + CXCR4 + and BMC therapies were associated with a trend in modest improvement (≈3%) in systolic function. Unfortunately, the study was plagued by a high dropout rate in the control cohort, probably because of the open-label design, which may have potentially accounted for some results not reaching statistical significance. Nonetheless, for patients who received CD34 + cell therapy, a clear trend for better outcomes was evident in patients with a baseline ejection fraction (EF) below the median (<37%) compared to those whose baseline EF was above the median. In addition, a low baseline EF (<37%) was an independent predictor for pronounced benefits in terms of cardiac function. This pattern is consistent with the results reported in REPAIR-AMI. The authors also highlighted that the use of a relatively small number of CS34 + CXCR4 + cells was associated with a similar trend in functional improvement as the use of 100 times more total mononuclear cells, similar to the results published by Kawamoto and colleagues in rats. The mechanisms that could account for these benefits were not explored, but it is unlikely that it was a result of de novo cardiomyocytes. Instead, recent laboratory evidence indicates that CD34 + cells secrete exosomes that have independent angiogenic activity, potentially accounting for a significant component of the paracrinic effect of these cells.

Indeed, the hallmark of ischemic cardiomyopathies, such as myocardial infarction, is the loss of myocardium. In this scenario, cell therapy may be a futile endeavor if no meaningful restoration of myocardium can be accomplished. Moreover, the data supporting the cardiomyogenic potential of CD34 + purified cells is less than convincing. However, in situations in which instead neovascularization may be preferred, CD34 + cells could offer some benefit. Early evidence for the feasibility, safety, and benefit of purified CD34 + cells mobilized by G-CSF administration in 24 patients with refractory angina was provided by Losordo and colleagues in a pilot phase I/IIa randomized trial. Follow-up was performed in a larger double-blind, randomized phase II trial in 167 patients. CD34 + purified cells, or an equal volume of diluent (placebo), were injected with an electromechanical (NOGA) mapping injection catheter. Low-dose CD34 + therapy improved both weekly angina frequency and exercise tolerance measures. Mortality was not different between control and cell therapy cohorts at 12 months, although cell mobilization and collection procedures were associated with elevations in cardiac enzymes that will require follow-up in future studies.

CD34 + purified cells might represent a natural progression from unfractionated BMC therapies, and a more standard product compared with HSC- or EPC-based therapies based on their properties and preclinical results. However, cumulative evidence reviewed in a recent meta-analysis of BMC trials suggests that, overall, CD34 + purified cell therapy might not offer meaningful benefits in the settings of acute MI or chronic HF, possibly because of a lack of clinically relevant cardiomyogenic potential. Given their potent proangiogenic paracrinic properties, CD34 + cells could offer potential in settings where neovascularization in itself could offer substantial benefits, such as refractory angina or chronic ischemic cardiomyopathy.

Mesenchymal Stem Cells

Mesenchymal stem cells are stromal cells found within the bone marrow or adipose tissue and can be expanded via ex vivo culture (see Fig. 101-3 B ). Similar to other bone marrow cell types, they are loosely defined based on surface expression and culture characteristics which include adherence to plastic in cell culture and the surface antigen expression of CD105 + /CD90 + /CD73 + /CD34 /CD45 /CD11b or CD14 /CD19 or CD79 α /HLA DR1 . MSCs have been identified as promising candidates because of their relative ease of procurement, multilineage differentiation potential, robust secretome, antiapoptotic, immunomodulatory properties. They have also been shown to be capable of activating resident cardiac stem cells. Given their immune-privileged status, they may serve as ideal allograft candidates, although the long-term benefits as allogeneic products have been challenged. Some authors contend that MSCs can adopt cardiomyocyte-like phenotypes when injected into both healthy and infarcted myocardium; however, these results have been scrutinized, and others attribute these observations as either MSC fusion with resident cardiomyocytes or nonexistent altogether.

Purified human-derived MSCs have been shown to attenuate contractile dysfunction and to prevent adverse remodeling in rodent and swine models of myocardial infarction. In contrast with the classical method of collecting cells and deploying via infusion or injection, initial clinical attempts at harnessing MSCs assessed pharmacologic mobilization of the cells from the bone marrow using G-CSF. Assessing G-CSF mobilized cells soon after AMI, results from the “G-CSF ST-segment Elevation Myocardial Infarction” (G-CSF STEMI), “STEM cells in Myocardial Infarction” (STEMMI), and “Regenerate Vital Myocardium by Vigorous Activation of Bone Marrow Stem Cells” (REVIVAL-2) trials showed that G-CSF administration did not result in improved systolic function compared with placebo. A substudy of the STEMMI trial sought to describe the cells mobilized by G-CSF administration and to determine the association, if any, between plasma concentration of these cell types and changes in left ventricular systolic function. The authors associated an inverse relationship between circulating MSCs and systolic functional improvement following G-CSF administration after MI. This relationship was left relatively unexplored, but it could be a consequence of several mechanisms. It may be that reduced circulating MSCs is a reflection of increased homing to the infarcted myocardium, or given the large size of MSCs their increased concentration in the circulation may contribute to further microembolization and infarction.

MSCs are one of the few cell types to date that may offer a true “off-the-shelf” product given their potential immune-privileged status. Hare and colleagues assessed the safety of this type of allogeneic product in a double-blind, randomized, dose-ranging trial of 53 patients with reperfused MI. The primary end-point of the study was safety, whereas efficacy measured by ejection fraction and ventricular volumes constituted exploratory, secondary end-points. Safety profiles were similar between the allogeneic MSC product (Prochymal; Osiris Therapeutics Inc., Baltimore, MD) and placebo treatments. Patients receiving MSCs experienced fewer episodes of ventricular tachycardia and improved pulmonary forced expiratory volumes. In terms of efficacy, MSCs were associated with improved global symptom score and ejection fraction. These results highlight some potential advantages of MSCs versus other BMC products. For example, the product used in this trial was a readily available allogeneic product prepared from healthy donors, circumventing any potential issues with patient clinical condition on cell potency. It also obviates the need for an invasive and highly painful bone marrow aspiration. Results from the recent POSEIDON trial reinforce some of these benefits. POSEIDON was a small, randomized, pilot trial that compared the safety and efficacy of autologous or allograft MSC transendocardial delivery in patients with chronic ischemic LV dysfunction secondary to MI. The primary end-point was safety measured by incidence of severe adverse reactions. None of the cell products reached the predefined stopping point for incident severe adverse reactions, supporting the low immunogenicity of MSCs. In terms of secondary end-points, reflecting efficacy, the combined results appear encouraging. Autologous, but not allogeneic, therapy resulted in improvements in the 6-minute walk test and Minnesota Living with Heart Failure Questionnaire (MLHFQ) scores. Moreover, both cohorts demonstrated improvements in New York Heart Association (NYHA) class. Both were associated with reduced early enhancement defect, a measure of infarct size, end-systolic volumes, and sphericity. In addition, patients having received allogeneic therapy demonstrated improved end-diastolic volumes. Changes in EF were not apparent from the aggregate results, although a trend for an inverse dose-response relationship was observed. Increases in EF and pronounced changes in ventricular geometry were evident in patients receiving a lower dose of MSCs. The results are promising but will require substantiation in several follow-up trials given the small sample size, open-label design, and lack of a placebo-controlled group.

MSCs are an exciting candidate as the field of cell therapy progresses. Given their potent paracrinic properties and immune privilege, they may truly offer the potential for an off-the-shelf product. A major knowledge gap that remains before the potential for these cells can be fully realized is the mechanisms underpinning observed benefits. Nevertheless, given their purported benefits and considerable secretome, MSCs appear to be promising as cardiac cell therapy evolves.

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here