279
Views
25
CrossRef citations to date
0
Altmetric
Review

Brain repair: cell therapy in stroke

&
Pages 31-44 | Published online: 21 Feb 2014

Abstract

Stroke affects one in every six people worldwide, and is the leading cause of adult disability. Some spontaneous recovery is usual but of limited extent, and the mechanisms of late recovery are not completely understood. Endogenous neurogenesis in humans is thought to contribute to repair, but its extent is unknown. Exogenous cell therapy is promising as a means of augmenting brain repair, with evidence in animal stroke models of cell migration, survival, and differentiation, enhanced endogenous angiogenesis and neurogenesis, immunomodulation, and the secretion of trophic factors by stem cells from a variety of sources, but the potential mechanisms of action are incompletely understood. In the animal models of stroke, both mesenchymal stem cells (MSCs) and neural stem cells (NSCs) improve functional recovery, and MSCs reduce the infarct volume when administered acutely, but the heterogeneity in the choice of assessment scales, publication bias, and the possible confounding effects of immunosuppressants make the comparison of effects across cell types difficult. The use of adult-derived cells avoids the ethical issues around embryonic cells but may have more restricted differentiation potential. The use of autologous cells avoids rejection risk, but the sources are restricted, and culture expansion may be necessary, delaying treatment. Allogeneic cells offer controlled cell numbers and immediate availability, which may have advantages for acute treatment. Early clinical trials of both NSCs and MSCs are ongoing, and clinical safety data are emerging from limited numbers of selected patients. Ongoing research to identify prognostic imaging markers may help to improve patient selection, and the novel imaging techniques may identify biomarkers of recovery and the mechanism of action for cell therapies.

Introduction

Stroke is the most common cause of adult-acquired disability in the developedCitation1 and developing world.Citation2 With an aging population, the incidence and prevalence of stroke are predicted to rise.Citation3 Stroke is an acute-onset clinical syndrome that develops following a vascular insult to the brain. Brain ischemia resulting from thromboembolism or less frequently, in situ thrombosis, constitutes 80%–85%, and hemorrhage resulting from hypertension or vessel wall pathology constitutes 15%–20% of all strokes. Following vascular occlusion, a complex chain of events occurs at a molecular level, leading to irreversible tissue injury, including failure of energy synthesis, loss of transmembrane ionic gradients dependent on active transport, cell depolarization, and excitotoxicity due to the excess release of excitatory neurotransmitters. In the region with severely reduced blood flow (the ischemic core), these processes result in rapid cell necrosis affecting all the cellular elements (neurons, glia, and blood vessels). A region around the core (the ischemic penumbra) transiently maintains a collateral blood supply sufficient for cell viability. Restoring perfusion can salvage penumbral tissue, and timely recanalization is the most robust predictor of good clinical prognosis following ischemic stroke.Citation4 Early thrombolysis with intravenous recombinant tissue plasminogen activator increases the likelihood of recanalization and a recovery to independence defined on scales of disability and handicap.Citation5 Alternative reperfusion strategies have not yet shown benefit. Secondary processes following ischemic injury and cell necrosis include an inflammatory response, with the activation of microglia, infiltration of tissue by neutrophils and macrophages from the blood, and blood–brain barrier breakdown. Inflammatory mediators can act as chemoattractants for both the endogenous and exogenous cells involved in tissue repair. At the network level, regions of the brain that were previously connected to the infarcted area reorganize, at least in terms of the brain activation patterns seen on functional magnetic resonance imaging (fMRI). Rehabilitation exploits the combination of functional reorganization and adaptation after stroke.Citation6

Immediately after stroke, several events, including edema, deafferentation, and inflammation, occur around the infarct, and some early functional recovery can be attributed to the resolution of edema and inflammation. However, this is usually limited, and other processes, including immunomodulation, angiogenesis, endogenous neurogenesis, and altered gene expression, may be involved in the longer-term recovery of function. The apparent translational failure of neuroprotective strategiesCitation7 that aim to interrupt or slow the injurious postischemic biochemical/molecular events may be attributed to various factors, including the heterogeneity of clinical stroke populations,Citation8 inadequate sample sizes, and dose-limiting drug toxicities. However, recent critiques of the preclinical literature have suggested that the discrepancies between the preclinical and clinical studies are likely also to have arisen from publication bias,Citation9 the limited replication of results, and experimental methodological flaws that inflated the estimates of effect size and led to the potential selection of inappropriate therapeutic candidates.Citation10 A series of Stroke Therapy Academic Industry Roundtable (STAIR)Citation11 meetings produced recommendations on the minimum standards for preclinical evidence that should, ideally, underpin the selection of drug candidates for clinical testing, as well as the clinical trial methods. An equivalent process, entitled Stem Cells as an Emerging Paradigm in Stroke (STEPS), has provided a forum for methodological discussions in the cell therapy field.Citation12,Citation13

Overview of stem cell therapy in stroke

Stem cells are undifferentiated cells that have the capacity to self-renew and differentiate into a range of tissues. Stroke therapy has distinct requirements compared with other neurological diseases, like Parkinson’s disease or multiple sclerosis, since stroke is nonprogressive, involves a focal loss of tissue of all cell types, and is typically associated with a degree of endogenous recovery. Stem cell therapy is not, therefore, restricted to a paradigm of the replacement of a tissue, or a specific neuronal cell type (the focus in Parkinson’s disease, for example), but potentially extends to effects on inflammation, immunomodulation, and the stimulation of endogenous recovery. Cell therapies probably act on multiple mechanisms in ischemic stroke, depending upon the timing and mode of administration; however, unlike neuroprotectant drugs, cell therapies have the advantage that they may be able to respond dynamically to an environment that varies both temporally and spatially after ischemia, rather than targeting a single pathway or mechanism of action. Interaction with the host environment appears to dictate the phenotypic properties of stem cell grafts. Stem cells come from various sources, and although they share some common properties, they also differ in many respects and behave differently in terms of their rate of differentiation, trophic factor secretion, and in their stimulation of endogenous processes when in a pathologic environment. No studies have compared the different cell types in the same experiment.

Endogenous stem cells

Until the middle of the 20th century, it was generally believed that neurogenesis in the mammalian nervous system was restricted to fetal development and that regeneration did not occur in the adult brain. In 1965, Altman and DasCitation14 first reported postnatal neurogenesis in the rat brain, and by the late 20th century, there was evidence of similar endogenous neurogenesis in humans.Citation15 In animals and humans, neuroblasts are known to be produced in the subventricular zone,Citation16 subgranular zone of the hippocampal dentate gyrus,Citation17 and, albeit controversially, in the newly discovered subcallosal zone that lies between the hippocampus and corpus callosum in rats.Citation18 Increased neuroblast production following ischemic stroke has been observed in the rat subventricular zone, and cortical neuroblasts have been reported in both a rat stroke modelCitation19 and in human brain biopsy specimens of penumbral tissue that were acquired for diagnostic purposes after stroke.Citation20,Citation21 Neuroblast production has also been stimulated experimentally by extrinsic growth factors, like hepatocyte growth factor,Citation22 and specific molecules, such as statinsCitation23 and fluoxetine,Citation24 but few of these neuroblasts appear able to migrate to the boundary of ischemic damage,Citation25 calling into question their functional relevance – amplifying and sustaining this endogenous poststroke neurogenesis response and overcoming the low rate of cell survival may be relevant for functional gains. An improved understanding of the role of changes in the expression of the developmental genes and associated proteins that are observed along the ischemic border after strokeCitation26 may also be important in developing cell or pharmacologic augmentation therapies that will capitalize on endogenous neuroregenerative capacity.

Olfactory ensheathing cells are a self-renewing population of cells that display the properties of both glia and Schwann cells and are found at the junction between the central and peripheral nervous systems. Their main properties have led them to be studied more in the context of spinal cord and nerve root injuries, but their neuroplastic effects have been tested in murine models of stroke and they have been found to promote neurite outgrowth.Citation27 Few preclinical studies exist, and their clinical application remains unclear in stroke.Citation28

Exogenous stem cells

The application of exogenous cell therapy in neurology began with neurodegenerative diseases, for which fetal ventral mesencephalic tissue was transplanted with the intention of replacement of a specific cell type, such as the dopaminergic neurons of the basal ganglia.Citation29 Cell replacement for stroke requires the regeneration of multiple functionally specialized cell types, with differing ratios in different brain regions, but extends also to glial cells and blood vessels since the injury involves the entire neurovascular unit.

Neural stem cells

Whether neural stem cells (NSCs) should be defined by their tissue of origin or their capacity to generate neural tissue is not universally agreed.Citation15 The following discussion considers the tissue of origin to define NSCs. Cells sourced from ectodermal tissue, such as the central nervous system (CNS), have restricted differentiation potential and can further be categorized into embryonic,Citation30 fetal,Citation31 or adult,Citation30 by origin. The use of adult-derived cells does not share the ethical and practical concerns of the use of embryonic or fetal cells. Cells from adult murine brain have been harvested, expanded in culture, and reimplanted as an allogeneic source. Isolated cells can be induced to form neurospheres, which are then expanded in vitro before delivery via various routes, including stereotactic (ST) injection to the brain, and intravenous (IV), intra-arterial (IA) and intracerebroventricular (ICV) administration. The differentiation spectrum of NSCs is restricted to neurons, astrocytes, or oligodendrocytes and can be influenced by intrinsic factors,Citation32 such as neuron-restrictive silencing factor, and extrinsic factors, such as experimental hypoxiaCitation33 and epidermal growth factors. The transmission of infectious agents by culture media is a concern that can be addressed only incompletely by applying strict Good Manufacturing Practice standards. Human fetal brain cortex cells have been immortalized by the insertion of c-MycCitation31 and v-MycCitation34 transcription factor genes, in order to enhance cell survival or allow the regulation of cell replication (for example, where c-Myc expression is under the regulatory control of a modified estrogen receptor).Citation35 The majority of NSC experimental stroke studies have used STCitation31,Citation36Citation38 intracerebral delivery, with implantation ranging from hoursCitation39 to 6 weeksCitation38 after stroke. Cell migration to ischemic regions has been reported following implantation by ST,Citation38 IV,Citation40 or IACitation41 routes. ST-implanted human NSCs have migrated up to 1.2 mm in the lesioned hemispheres compared with 0.2 mm in naïve rat brain.Citation42 Whether more distant migration occurs is unclear. Cell survival varies and depends on the timing and mode of delivery. Following ST implantation, proximity to the lesion influences survival,Citation38 while very few cells reach the brain following IV administration as they are filtered by the pulmonary vascular bed and sequestered in the spleen.Citation43 Slightly greater cell survival in the CNS is seen after IA delivery.Citation44 Although many cells die early after administration, bioluminescent human NSCs ST-implanted 7 days after middle cerebral arterial occlusion (MCAo) have been observed to survive beyond 2 months, with over 50% cell survival confirmed on histology.Citation45 Surviving cells exhibit a wide spectrum of fates, ranging from 78% remaining in an immature stateCitation36 at week 5, to unquantified numbers of differentiated neurons forming synapses with host cells.Citation45 The expression of neuronal cell surface markers does not necessarily indicate functioning neuronal tissue, still less, useful integration, and the contribution of the surviving cells to an observed functional improvement is still unclear. A change in neurological or behavioral function has been the preferred outcome, rather than infarct volume, as NSC studies have mostly chosen to implant at subacute time points, when infarcts are well-established. A modified neurological severity score (NSS), which provides a composite score based on motor, sensory, reflex, and balance responses, has been used commonly in preclinical rodent studies to assess change, reporting significant improvements compared with sham controls, following NSC therapy.Citation46,Citation47 However, a wide range of behavioral tests has been employed. The reporting of results differs across laboratories,Citation48 and the reproducibility of tests across observers and also across time has seldom been reported. Despite the lack of clarity regarding the mechanisms of action, NSCs are believed to alter white matter tissue structure, and a noninvasive method to measure this would be valuable. The effects of NSC treatment on white matter reorganization can be monitored by measuring water diffusionCitation49 using magnetic resonance imaging (MRI) with diffusion tensor imaging (DTI) sequences. Although DTI in small animals is compromised by the relatively lesser volume of white matter present in rodents compared with humans, there are also some advantages to use of DTI, including the ability to apply longer scan acquisition times and higher magnetic field strength. The white matter reorganization observed on histology was coincident with improved fractional anisotropy, and fiber tracking maps revealed similar orientation patterns to that seen on immunohistology.Citation41

Mesenchymal stem cells

Since the first bone marrow-derived mesenchymal stem cells (MSCs),Citation50 many other cell types with similar properties from various tissues, including bone marrow mononuclear cells, adipose-derived stem cells, umbilical cord blood cells (UCBCs), endothelial progenitor cells (EPCs), peripheral blood progenitor cells, cluster of differentiation (CD)34+ cells from placenta, periosteal stem cells, and amniotic fluid cells have all been proposed as potential alternatives. The relative ease of cell acquisition without ethical difficulties has fuelled interest in MSCs, but the specific characterization of MSCs has not been consistent over time,Citation51,Citation52 making study comparability difficult. In vitro cultures contain a mix of committed and noncommitted progenitors that can form, not only mesodermal, but under certain circumstances, also ectodermal cell types, like neurons, but it is unclear whether the MSCs differentiated along neuronal lines in culture will have the same properties as do NSCs. Human neuronal MSCs, which have the ability to differentiate into neuronal cells following transfection of the Notch intracellular domain, were ST-implanted 4 days after MCAo in gerbils and compared with human MSC. In the human neuronal MSC group, better cell survival and functional recovery were observed despite the absence of synaptic connection between the transplanted and recipient cerebral cells on fluorescence-in-situ-hybridization (FISH), suggesting that the neuronal differentiation did not contribute to the MSC beneficial effects.Citation53 In experiments with MSCs derived from donor rats,Citation54 mice,Citation55 rabbit,Citation56 (autologous or allogeneic), or humansCitation57 (xenogeneic), cells have been transplanted by IV,Citation57 IA,Citation58 ST,Citation59 or intracisternalCitation58 routes into animals, from hoursCitation57 to 1 monthCitation60 after induction of stroke with either temporary or permanent MCAo. Homing of the transplanted MSCs appears to occur via a complex multistep process that includes interactions with the stromal cell-derived factor 1 (SDF-1) (also called C-X-C motif chemokine 12 [CXCL12]) chemokine receptor.Citation61 Homing signals originate from within the active inflammatory zone in the injured tissue. MSC migration to specific sites has been observed in stroke studies, where they have been found to travel preferentially to the ischemic boundary, following IVCitation57 and ST delivery.Citation62 Few cells have been shown to survive in the studies of xenogeneic cell implantation. With no immunosuppression, cell survival of up to 2 weeks has been reported on ST implantation, but the proportion of surviving cells has not been quantifiedCitation63 and has qualitatively been described as being a small proportion only. Long-term cell engraftment has not been detected with IV administration on histology.Citation64 In another study, out of 3×106 MSCs delivered IV, only 3% of administered cells expressed neuronal markers in vivo,Citation65 further supporting the concept that tissue replacement is not likely to be a functionally relevant mechanism of action for this cell type. Trophic factors, such as brain-derived neurotrophic factor (BDNF), glial-derived neurotrophic factor (GDNF), vascular endothelial growth factor (VEGF), neurotrophin-3 (NT3), fibroblast growth factor (FGF), and thrombospondins, secreted by the MSCsCitation66,Citation67 in response to the local microenvironment may, along with their stimulation of neurogenesis,Citation66 angiogenesis,Citation68 and immunomodulation,Citation69 underlie functional recovery. Astrocytes are known to maintain normal neuronal function,Citation70 forming an important pathway for endogenous repair.Citation71 Exogenous MSCs have been observed to influence astrocyte survival and astrocyte trophic factor gene expression after anaerobic insult, by upregulating several kinase pathways and protein functions.Citation72 After ischemia, astrocytes form gliotic scar tissue, which may be helpful in limiting tissue inflammation but can impede axonal regeneration. IA-implanted MSCs have shown histological evidence of improved axon-myelin remodeling after stroke,Citation73 but it is unknown whether this mechanism is relevant in other routes of MSC administration. MSCs naturally adopt different trophic factor expression dependent on the injured host neural tissue.Citation74 Higher levels of BDNF, NT3, and VEGF have been detected at the ischemic boundary 14 days after ST human MSC transplantation in rat brains compared with controls that received saline.Citation75 The expression of VEGF and FGF has been consistently high at the ischemic boundary, potentially driving endothelial cell proliferation and angiogenesis, and facilitating regional blood flow.Citation76 In contrast with the NSC studies, infarct volume has been the preferred outcome measure for experimental MSC therapy, which has been predominantly administered in an acute or early subacute IV delivery paradigm, with significant reduction in infarct volumes and good correlation noted between histology and imaging measures.Citation77 Significant improvements have also been reported in behavioral measures, which have included assessments of sensorimotor function, motor coordination, and placing deficits during locomotion (treadmill test),Citation77 forelimb function and placing deficits (limb placement test),Citation78 motor coordination and balance (rotarod test),Citation75 and a composite of motor, sensory, reflex, and balance responses (NSS).Citation79 Other cell types including UCBCs, EPCs, adipose-derived stem cells, and hematopoietic progenitor cells (CD34+ cells), share some of the properties of bone marrow-derived MSCs and have been found to have similar effects in animal models. In animal experiments, UCBCs respond to ischemic region homing signals, migrate to the lesioned hemisphere following IV administration, and differentiate, as evidenced by immunohistochemical neuronal and astrocytic markers.Citation80 Some MCAo rat studies have failed to detect IV-administered UCBC in the lesions despite improvement in spontaneous activity and behavioral motor tests, suggesting a trophic factor-mediated response.Citation81,Citation82 CD34+ cells form a significant component of UCBCs that have been enriched either from the umbilical cord, peripheral blood, or bone marrow, and administered separately. IV administration of CD34+ in MCAo models has shown ischemic border zone neovascularization that has in turn, stimulated endogenous neurogenesis.Citation83 EPCs represent cells with varying cell expression markers,Citation84 typically CD34+, CD133+, and kinase insert domain receptor (KDR+) (also known as vascular endothelial growth factor receptor-2), with an angiogenic mechanism of action and found to reduce infarct volumes in rat stroke models when administered IV a day after MCAo.Citation85 Several Phase I and II MSC clinical trials are ongoing ().

Table 1 Clinical trials listed on http://www.clinicaltrials.gov (as of August 5, 2013)

Embryonic stem cells

Embryonic stem cells (ESCs) are derived from the first stages of embryonic development: the first human ESC lines were established in 1998 from the inner cell wall of the blastocyst stage.Citation86 Religious and moral objections have been raised to the medical use of embryonic material; however, it is not widely recognized that the ESCs used for medical research are generally obtained from in vitro fertilization programs. Media reports often fail to distinguish ESCs from other stem cell types, leading to public confusion. ESCs are pluripotent and able to differentiate into tissues of all three germ layers. Although at first glance this might appear advantageous, regulatory control over ESC differentiation may be necessary before therapeutic use, since ESCs tend to form teratomas when grafted,Citation87 with the postischemic environment possibly promoting teratoma formation.Citation88 ESC studies in animal stroke models have been concerned with mechanistic aspects rather than functional efficacy, and report only isolation, neutralization,Citation89 and the electrophysiological activity of differentiated neuronal cells.Citation90 Undifferentiated ESCs grafted into rat brains have differentiated and integrated with host tissues in stroke models,Citation91 showing improved functional outcomes on the cylinder test, which measures the spontaneous use of forelimbs.Citation92

ESCs remain widely researched as a source for in vitro generation of neuronal cell lines for drug screening, mechanistic investigation, or therapeutic use. ESCs can be stimulated to differentiate into specific neuronal populations or glia, with appropriately timed use of growth and inhibitory factors in relevant media and culture conditions. ESCs have been preferentially differentiated to a glutamatergic neuronal phenotype of the auditory nerves, with a view to specific tissue regeneration of the auditory nerve.Citation93 Similar preferentially differentiated cell cultures can be used for in vitro studies, to investigate several critical stroke-related molecular processes. Such studies provide tight experimental control despite limitations of their ability to investigate the role of cell interactions. The cellular effects of oxygen-glucose deprivation, hypothermia, oxidative stress, and excitotoxicity have been modeled with chosen degrees of injury, helping to improve our understanding of certain key pathological processes.Citation94

Induced pluripotent stem cells

Nobel Laureate Shinya Yamanaka and his colleague Kazutoshi Takahashi first demonstrated that differentiated murine cells could be reprogrammed to an embryonic-like state, with cells having the morphology, growth properties, and cell surface markers of ESCs, calling them induced pluripotent stem cells (iPSC). Similar iPSCs were later derived from adult human somatic cells.Citation95 The Yamanaka method involved the transfection of cells with four key nuclear transcription factors, under ESC culture conditions;Citation96 subsequent studies have identified alternative methods.Citation97 While superficially appealing as a means of obtaining ESC-like cells from adult tissue, the limited yield of these methods, the potential risks of clinical use of material obtained from viral transfection, and the multiple potentially oncogenic transcription factor genes, as well as (for stroke) the time required for culture expansion, all present significant clinical hurdles that are currently being investigated.Citation98 iPSCs can potentially generate autologous patient-specific cells, avoiding the ethical, moral, and legal issues of ESCs but may share the tumorigenicity issues of ESCs.Citation87 The intracerebral implantation of undifferentiated iPSCs in a rat MCAo model showed cell expansion to form large tridermal teratomas, with little behavioral improvement compared with controls, despite differentiated neuroblasts and mature neurons being seen in the ischemic lesion.Citation99 As is the case for ESCs, partial in vitro differentiation may be necessary before therapeutic uses can be contemplated. A recent study that used human iPSC-derived long-term expandable neuroepithelial-like stem cells in a T cell deficient rat MCAo model with a 4-month observation period found no new tumors or transplant overgrowth, suggesting that predifferentiation of iPSCs and the generation of long-term self-renewing neural cell lines may offer an effective strategy for minimizing the risk for tumor formation.Citation100 The reports of improvement in function, reduced infarct volume, and differentiated neuronal cells with electrophysiological properties and host synaptic connections following the intracerebral implantation of iPSCs derived from human fibroblastsCitation100,Citation101 are promising, but other studies using ST delivery of iPSCs have reported no functional improvement.Citation102

Stem cells and the immune system

Transplanted stem cell survival may be influenced by host immune responses, but the transplanted cells may themselves modulate the host inflammatory microenvironment after stroke. The immunogenicity of allogeneic stem cells varies according to the expression of their major histocompatibility complex (MHC) I and II and other molecules that stimulate host CD8+ or CD4+ T cells.Citation103 MSCs express very few MHC antigens, but cell surface marker expression may be modified by the host environment, and the lack of in vitro immunogenicity may not therefore be informative about the potential for problems in clinical use. However, to date, there have been no reports of cell-related adverse events or tumorigenesis following autologous MSC administration in the small number of early Phase I clinical trials in strokeCitation104 and multiple sclerosis.Citation105 Two clinical trials, of allogeneic NSCCitation106 (NCT01151124) and MSCCitation107 (NCT01297413) lines for the treatment of stroke with no coadministered immunosuppression, are currently investigating safety outcomes, including clinical, laboratory, and imaging markers. Although there is evidence that adult stem cells have an inherent immunologically privileged status and are capable of escaping rejection,Citation108 it is unclear whether their MHC expression is altered by exposure to proinflammatory cytokines, such as occurs in ischemic tissue injury. ST-implanted neural progenitor cells have been observed to have low immunogenicity as they are not exposed to systemic immune surveillance, but the blood–brain barrier is damaged after stroke and the CNS probably does not retain this status. There are suggestions that low immunogenicity could be a unique property of NSCs, based on a lack in upregulation of the immunological response to transplantation of murine NSCs, and the lack of difference observed in animals, whether or not immunosuppressed, 2 weeks postimplantation and 4 weeks post-MCAo.Citation109 Some xenogeneic animal stroke studies have coadministered immunosuppressant drugs on the assumption that the recipient species would reject donor cells of human origin. Whether or not xenogeneic studies necessitate immunosuppression is still unclear. Many studies have not reported the use of immunosuppression or have not considered studying its effects in detail.Citation36,Citation40,Citation45 Immunosuppressant drugs have independent neuroprotective effects in animal models of stroke, and their use was identified as a significant factor in modifying effect size estimates in a meta-analysis of animal studies.Citation110

Transplanted stem cells initiate a dynamic sequence of host immunomodulatory actions on exposure to the host inflammatory microenvironment. They not only integrate and differentiate but also home in, extravasate into the CNS, and modulate immune responses in situ.Citation111 NSCs are reported to show more tropism towards inflammatory sites than do MSCs.Citation112 Both NSCs and MSCs exhibit host immune modulation in vivo. MSCs release neurotrophic factors, such as BDNF, provide trophic support for vulnerable neurons in the ischemic penumbra, support endogenous oligodendrogenesis, and regulate anti-inflammatory responses, leading to enhanced tissue sparing.Citation65 NSCs attenuate brain inflammation, modulate microglia activation, limit demyelination, and promote host-driven repair.Citation113

Clinical trial design

Ideally, preclinical evidence of efficacy, information on the optimal timing and mode of delivery, and toxicity (including tumorigenesis and possibly gene silencing studies) should be considered in clinical trial planning. The STEPSCitation12,Citation13 meetings have suggested essential minimum criteria for the design of cell therapy stroke trials, by incorporating general principles from the earlier STAIR proposals that primarily concerned pharmaceutical development.Citation11 Although these recommend that preclinical studies include more than one strain of rodent, animals of varying ages, and that there be independent confirmation from one or more laboratories, in reality these recommendations are rarely followed due to high costs and potential commercial restrictions, and preclinical information may thus be limited.

The selection of an appropriate target stroke population will be influenced by the phase of study, expected mode of action of the cell therapy under study, and preclinical data. For studies primarily collecting safety data, chronic stroke patients with a broad range of severity who are not within the natural recovery period are likely to be candidates. The dose of stem cells for humans would usually be estimated based on animal studies and will need further human testing to define the maximum tolerated dose, minimum effective dose, and ideally, a dose-response curve. For a safety trial, an ascending dose design could be incorporated, especially for ST-delivered cells. For studies gathering efficacy data, subjects are likely to be in the acute or subacute stage after stroke, having deficits that are measurable by well-validated clinical scales, and whose natural evolution and variability over time after stroke are understood. Biomarkers, such as imaging, may offer greater biological confidence in the effects of treatment, with sample sizes that are smaller than are necessary to distinguish differences in the clinical disability scales; imaging markers should correlate with clinically relevant measures. For long-term safety follow up, the prevalence of significant comorbidities in stroke populations and the intensity of observation in a typically disabled and elderly population need to be considered in order to minimize trial subject attrition.

Stroke lesion sizes and locations are heterogeneous, and there is considerable interindividual variation in the neuroanatomical systems involved. Experimental stroke induction is a more controlled event, intended to produce a consistent lesion size and distribution. Anatomical characterization will thus play a significant role in patient selection in trials, not only from the perspective of surgical planning and feasibility for studies using delivery by ST implantation, but also, more generally as a prognostic marker. For example, corticospinal tract integrity predicts motor impairmentCitation114 and the probability of motor recovery.Citation115 Likewise, the timing of the ST intervention can be challenging in the acute stage, when lesion size varies considerably with improving edema and anatomical remodeling. While IV delivery is more straightforward from this aspect, a persistent occlusion of the target artery compromises IA cell delivery and may significantly reduce cell penetration or compromise survival at the target site. Timing will also be influenced by knowledge of the natural course of recovery and how this aligns with the chosen cell’s mechanism of action to maximize effect.

The feasibility of blinding patients and trialists to treatment allocation varies by the treatment delivery route and the requirement for placebo controls. Both placebo and blinding are relatively easier for IV therapy than for more invasive delivery routes since there are procedural complications from either IA or direct intracerebral delivery. Whether the scientifically rigorous inclusion of placebo controls to permit double blinding is sufficiently justified instead of potentially less hazardous sham alternatives that could yield a single-blind study (for example, a small incision in the groin rather than an IA placebo injection, or a scalp incision or burr hole rather than an injection of placebo fluid into the brain) may depend on the stage of research and the procedural risks. Functional change, for example, serial clinical scores or functional brain imaging, including both pre- and post-treatment periods, may reduce variance. Domain-specific endpoints, as suggested by Cramer et al,Citation116 may be more relevant than broad global outcome scales that traverse multiple neural systems; at the same time, they may restrict the trial entry criteria to those patients with very specific deficits, and thus reduce the generalizability of trial results (for example, motor outcome endpoints necessitate motor deficits at entry but a positive effect may arguably not be applicable to speech deficits).Citation117 Imaging-based outcomes may help to compare metrics, as they could be applied to both humans and animals. The use of imaging biomarkers for recovery prediction is promising, and these are currently being tested. The major confound of providing routine physical rehabilitation treatment in stroke recovery trials is an unresolved issue. While some studies of therapy inputs for specific clinical problems have identified dose-response relationships,Citation118,Citation119 for many routinely applied interventions, the efficacy or dose relationship is unknown. Even where evidence supports the therapy interventions, few clinical services deliver the optimal dose routinely, and there is enormous variability across sites and healthcare systems. Since animal studies have reported that concomitant specific physical rehabilitation may be a prerequisite of stem cell efficacy, this represents a major challenge in clinical study design.

Advances and future prospects in stem cell therapy for stroke

Many clinical investigations are documented on trials databases as planned or underway (), but the great majority of these studies are safety and tolerability studies, with small sample sizes and unspecified control groups. A wide range of cell types is being investigated, but most studies plan autologous bone marrow-derived cell administration by intravascular routes at subacute time points. These studies, if completed, will contribute valuable safety data that is a necessary prelude to large-scale efficacy trials, but ultimately, large randomized controlled trials with broad clinical endpoints will be required to judge the balance of risks and benefits.

The genetic modification of stem cells (for example to enhance the delivery of trophic factors, like BDNFCitation120 or VEGF,Citation121 or to address large scale manufacturing through conditional cell immortalizationCitation30) may offer advantages for allogeneic cell therapies. The allogeneic approaches offer the hypothetical advantage of immediate “off the shelf” availability, which is not possible with autologous cells, even if cells are not culture-expanded prior to administration. Laboratory research into the use of nonviral vectors for stable modification of cells, in vivo cell tracking, and the modification of stem cell gene expression profiles, is ongoing and will improve our understanding of cell function.Citation122 Tissue replacement as a therapeutic goal is almost certainly beyond the scope of the current therapeutic approaches in stroke, but the development of extracellular matrix bioscaffolds, to provide structural support for human NSCs, is a promising and potentially relevant approach for chronic stroke and other forms of brain injury.Citation123 The concept of stem cell–secreted extracellular membrane vesicles, providing extracellular waves of information capable of inducing multiple functional responses in adjacent and distant target cells, has emerged recently; the relevance of the bidirectional genetic information exchange between stem and target cells via MSC-secreted extracellular membrane vesiclesCitation124 is under investigation as a possible means of modifying graft–host interactions.

Parallel advances in biomaterial engineering and nanotechnology could provide an inert scaffold for ex-vivo stem cell expansion and intracranial delivery,Citation125 and may in future address the limitation, for current cell therapy paradigms, of the major loss of brain tissue after stroke that leaves only a cystic cavity.

Applying novel imaging techniques to monitor stem cell effects and identify biomarkers is likely to be the key to the neurological application of cell therapies. Conventional structural imaging is unlikely to be helpful, but modalities, such as motor task fMRI can predict treatment responseCitation126 and provide a measure of the balance of interhemispheric control,Citation127 and DTI can provide information on axonal integrity, which correlates with functional recovery.Citation128 Approaches such as resting state fMRI may allow the assessment of the effect of stem cells at a network level on either hemisphere.Citation129 Multimodal approachesCitation130 combining fMRI and DTI are advancing, and more work with stem cell–treated subjects will improve the use of imaging-based biomarkers for patient selection, baseline stratification, and outcome assessment.

Conclusion

Contrary to long-held beliefs, we now know that the brain is highly malleable after an ischemic insult. Endogenous neurogenesis, angiogenesis, and synaptogenesis occurs in humans, albeit at a rate that is able to provide only partial functional recovery in the majority of cases. Cell therapy offers a potential for multimodal action that is promising within the domain of brain repair therapies. Despite the almost certain publication bias in animal stroke studies, stem cell experiments have shown evidence of cell migration to the lesion, survival, and varying degrees of differentiation. Both tissue-specific NSCs and non-tissue-specific MSCs have been associated with significantly improved behavioral outcomes. A comprehensive understanding of their mechanism of action is lacking, but tissue replacement is now believed likely to constitute only a minor contribution (if any) to the therapeutic effect. Accordingly, a cell type’s capacity to differentiate along specific pathways is likely to be a less relevant consideration. The multiple mechanisms of action of stem cells include the secretion of trophic factors, immunomodulation, and anti-inflammatory effects. The great majority of the early cell therapy clinical studies have involved adult-derived cells of either autologous or allogeneic origin, and no major safety issues have been identified to date, although the numbers of subjects have been extremely small and follow-up periods limited. Several clinical trials are ongoing or planned, mostly using MSC cells delivered by IV infusion.

Disclosure

KWM is the chief investigator of the PISCES trial of human neural stem cells in stroke and a related Phase II trial. Both trials are funded by ReNeuron Ltd. DK is a subinvestigator for the PISCES trial. The authors report no other conflicts of interest.

References

  • NorrvingBKisselaBThe global burden of stroke and need for a continuum of careNeurology2013803 Suppl 2S5S1223319486
  • TruelsenTHeuschmannPUBonitaRStandard method for developing stroke registers in low-income and middle-income countries: experiences from a feasibility study of a stepwise approach to stroke surveillance (STEPS Stroke)Lancet Neurol20076213413917239800
  • DonnanGAFisherMMacleodMDavisSMStrokeLancet200837196241612162318468545
  • RhaJHSaverJLThe impact of recanalization on ischemic stroke outcome: a meta-analysisStroke200738396797317272772
  • HackeWDonnanGFieschiCATLANTIS Trials InvestigatorsECASS Trials InvestigatorsNINDS rt-PA Study Group InvestigatorsAssociation of outcome with early stroke treatment: pooled analysis of ATLANTIS, ECASS, and NINDS rt-PA stroke trialsLancet2004363941176877415016487
  • LanghornePBernhardtJKwakkelGStroke rehabilitationLancet201137797781693170221571152
  • GinsbergMDNeuroprotection for ischemic stroke: past, present and futureNeuropharmacology200855336338918308347
  • MuirKWHeterogeneity of stroke pathophysiology and neuroprotective clinical trial designStroke20023361545155012052989
  • MacleodMRO’CollinsTHowellsDWDonnanGAPooling of animal experimental data reveals influence of study design and publication biasStroke20043551203120815060322
  • DirnaglUIadecolaCMoskowitzMAPathobiology of ischaemic stroke: an integrated viewTrends Neurosci199922939139710441299
  • Stroke Therapy Academic Industry Roundtable (STAIR)Recommendations for standards regarding preclinical neuroprotective and restorative drug developmentStroke199930122752275810583007
  • Stem Cell Therapies as an Emerging Paradigm in Stroke ParticipantsStem Cell Therapies as an Emerging Paradigm in Stroke (STEPS): bridging basic and clinical science for cellular and neurogenic factor therapy in treating strokeStroke200940251051519095993
  • SavitzSIChoppMDeansRCarmichaelSTPhinneyDWechslerLStem Cell Therapy as an Emerging Paradigm for Stroke (STEPS) IIStroke201142382582921273569
  • AltmanJDasGDPost-natal origin of microneurones in the rat brainNature196520750009539565886931
  • GageFHMammalian neural stem cellsScience200028754571433143810688783
  • WangCLiuFLiuYYIdentification and characterization of neuroblasts in the subventricular zone and rostral migratory stream of the adult human brainCell Res201121111534155021577236
  • ErikssonPSPerfilievaEBjörk-ErikssonTNeurogenesis in the adult human hippocampusNat Med1998411131313179809557
  • SeriBHerreraDGGrittiAComposition and organization of the SCZ: a large germinal layer containing neural stem cells in the adult mammalian brainCereb Cortex200616Suppl 1i103i11116766696
  • ShimadaISPetersonBMSpeesJLIsolation of locally derived stem/progenitor cells from the peri-infarct area that do not migrate from the lateral ventricle after cortical strokeStroke2010419e552e56020671247
  • LindvallOKokaiaZMartinez-SerranoAStem cell therapy for human neurodegenerative disorders-how to make it workNat Med200410SupplS42S5015272269
  • JinKWangXXieLEvidence for stroke-induced neurogenesis in the human brainProc Natl Acad Sci U S A200610335131981320216924107
  • ShangJDeguchiKOhtaYStrong neurogenesis, angiogenesis, synaptogenesis, and antifibrosis of hepatocyte growth factor in rats brain after transient middle cerebral artery occlusionJ Neurosci Res2011891869520963849
  • ChenJZhangZGLiYStatins induce angiogenesis, neurogenesis, and synaptogenesis after strokeAnn Neurol200353674375112783420
  • ZhaoCDengWGageFHMechanisms and functional implications of adult neurogenesisCell2008132464566018295581
  • ArvidssonACollinTKirikDKokaiaZLindvallONeuronal replacement from endogenous precursors in the adult brain after strokeNat Med20028996397012161747
  • CramerSCChoppMRecovery recapitulates ontogenyTrends Neurosci200023626527110838596
  • ShyuWCLiuDDLinSZImplantation of olfactory ensheathing cells promotes neuroplasticity in murine models of strokeJ Clin Invest200811872482249518596986
  • ChoiDGladwinKOlfactory ensheathing cells, part II: source of cells and application to patientsWorld Neurosurg EpubJuly242013
  • PeschanskiMDeferGN’GuyenJPBilateral motor improvement and alteration of L-dopa effect in two patients with Parkinson’s disease following intrastriatal transplantation of foetal ventral mesencephalonBrain199411734874998032859
  • TakahashiKYasuharaTShingoTEmbryonic neural stem cells transplanted in middle cerebral artery occlusion model of rats demonstrated potent therapeutic effects, compared to adult neural stem cellsBrain Res2008123417218218703033
  • PollockKStroemerPPatelSA conditionally immortal clonal stem cell line from human cortical neuroepithelium for the treatment of ischemic strokeExp Neurol2006199114315516464451
  • MassirerKBCarromeuCGriesi-OliveiraKMuotriARMaintenance and differentiation of neural stem cellsWiley Interdiscip Rev Syst Biol Med20113110711421061307
  • De FilippisLDeliaDHypoxia in the regulation of neural stem cellsCell Mol Life Sci201168172831284421584807
  • CacciEVillaAParmarMGeneration of human cortical neurons from a new immortal fetal neural stem cell lineExp Cell Res2007313358860117156776
  • StevanatoLCortelingRLStroemerPc-MycERTAM transgene silencing in a genetically modified human neural stem cell line implanted into MCAo rodent brainBMC Neurosci2009108619622162
  • AndresRHHorieNSlikkerWHuman neural stem cells enhance structural plasticity and axonal transport in the ischaemic brainBrain2011134Pt 61777178921616972
  • IshibashiSSakaguchiMKuroiwaTHuman neural stem/progenitor cells, expanded in long-term neurosphere culture, promote functional recovery after focal ischemia in Mongolian gerbilsJ Neurosci Res200478221522315378509
  • DarsaliaVAllisonSJCusulinCCell number and timing of transplantation determine survival of human neural stem cell grafts in stroke-damaged rat brainJ Cereb Blood Flow Metab201131123524220531461
  • RosenblumSWangNSmithTNTiming of intra-arterial neural stem cell transplantation after hypoxia-ischemia influences cell engraftment, survival, and differentiationStroke20124361624163122535265
  • ChuKParkKILeeSTCombined treatment of vascular endothelial growth factor and human neural stem cells in experimental focal cerebral ischemiaNeurosci Res200553438439016198014
  • JiangQZhangZGDingGLMRI detects white matter reorganization after neural progenitor cell treatment of strokeNeuroimage20063231080108916860575
  • KellySBlissTMShahAKTransplanted human fetal neural stem cells survive, migrate, and differentiate in ischemic rat cerebral cortexProc Natl Acad Sci U S A200410132118391184415280535
  • PendharkarAVChuaJYAndresRHBiodistribution of neural stem cells after intravascular therapy for hypoxic-ischemiaStroke20104192064207020616329
  • LiLJiangQDingGEffects of administration route on migration and distribution of neural progenitor cells transplanted into rats with focal cerebral ischemia, an MRI studyJ Cereb Blood Flow Metab201030365366219888287
  • DaadiMMLiZAracAMolecular and magnetic resonance imaging of human embryonic stem cell-derived neural stem cell grafts in ischemic rat brainMol Ther20091771282129119436269
  • ZhangPLiJLiuYHuman neural stem cell transplantation attenuates apoptosis and improves neurological functions after cerebral ischemia in ratsActa Anaesthesiol Scand20095391184119119650809
  • SongMKimYJKimYHRohJKimSUYoonBWEffects of duplicate administration of human neural stem cell after focal cerebral ischemia in the ratInt J Neurosci2011121845746121574891
  • HicksASchallertTJolkkonenJCell-based therapies and functional outcome in experimental strokeCell Stem Cell20095213914019664986
  • BeaulieuCThe basis of anisotropic water diffusion in the nervous system – a technical reviewNMR Biomed2002157–843545512489094
  • FriedensteinAJPetrakovaKVKurolesovaAIFrolovaGPHeterotopic of bone marrow. Analysis of precursor cells for osteogenic and hematopoietic tissuesTransplantation1968622302475654088
  • HorwitzEMLe BlancKDominiciMInternational Society for Cellular TherapyClarification of the nomenclature for MSC: The International Society for Cellular Therapy position statementCytotherapy20057539339516236628
  • DominiciMLe BlancKMuellerIMinimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statementCytotherapy20068431531716923606
  • XuHMikiKIshibashiSTransplantation of neuronal cells induced from human mesenchymal stem cells improves neurological functions after stroke without cell fusionJ Neurosci Res201088163598360920936694
  • Gutiérrez-FernándezMRodríguez-FrutosBAlvarez-GrechJFunctional recovery after hematic administration of allogenic mesenchymal stem cells in acute ischemic stroke in ratsNeuroscience201117539440521144885
  • CuiXChenJZacharekANitric oxide donor upregulation of stromal cell-derived factor-1/chemokine (CXC motif) receptor 4 enhances bone marrow stromal cell migration into ischemic brain after strokeStem Cells200725112777278517641243
  • ChenZZJiangXDZhangLLBeneficial effect of autologous transplantation of bone marrow stromal cells and endothelial progenitor cells on cerebral ischemia in rabbitsNeurosci Lett20084451364118755241
  • HoritaYHonmouOHaradaKHoukinKHamadaHKocsisJDIntravenous administration of glial cell line-derived neurotrophic factor gene-modified human mesenchymal stem cells protects against injury in a cerebral ischemia model in the adult ratJ Neurosci Res20068471495150416998918
  • RuanGPHanYBWangTHComparative study among three different methods of bone marrow mesenchymal stem cell transplantation following cerebral infarction in ratsNeurol Res201335221222023452580
  • BraunRGAndrewsEMKartjeGLKinematic analysis of motor recovery with human adult bone marrow-derived somatic cell therapy in a rat model of strokeNeurorehabil Neural Repair201226789890622619255
  • ShenLHLiYChenJTherapeutic benefit of bone marrow stromal cells administered 1 month after strokeJ Cereb Blood Flow Metab200727161316596121
  • RosenkranzKKumbruchSLebermannKThe chemokine SDF-1/CXCL12 contributes to the ‘homing’ of umbilical cord blood cells to a hypoxic-ischemic lesion in the rat brainJ Neurosci Res20108861223123319937807
  • KangSKLeeDHBaeYCKimHKBaikSYJungJSImprovement of neurological deficits by intracerebral transplantation of human adipose tissue-derived stromal cells after cerebral ischemia in ratsExp Neurol2003183235536614552877
  • KimSSYooSWParkTSNeural induction with neurogenin1 increases the therapeutic effects of mesenchymal stem cells in the ischemic brainStem Cells20082692217222818617687
  • Mora-LeeSSirerol-PiquerMSGutiérrez-PérezMTherapeutic effects of hMAPC and hMSC transplantation after stroke in micePLoS One201278e4368322952736
  • ChenJLiYWangLTherapeutic benefit of intravenous administration of bone marrow stromal cells after cerebral ischemia in ratsStroke20013241005101111283404
  • ChoppMLiYTreatment of neural injury with marrow stromal cellsLancet Neurol2002129210012849513
  • WakabayashiKNagaiASheikhAMTransplantation of human mesenchymal stem cells promotes functional improvement and increased expression of neurotrophic factors in a rat focal cerebral ischemia modelJ Neurosci Res20108851017102519885863
  • WuYChenLScottPGTredgetEEMesenchymal stem cells enhance wound healing through differentiation and angiogenesisStem Cells200725102648265917615264
  • LiJZhuHLiuYHuman mesenchymal stem cell transplantation protects against cerebral ischemic injury and upregulates interleukin-10 expression in MacacafascicularisBrain Res20101334657220353760
  • Sidoryk-WegrzynowiczMWegrzynowiczMLeeEBowmanABAschnerMRole of astrocytes in brain function and diseaseToxicol Pathol201139111512321075920
  • SofroniewMVReactive astrocytes in neural repair and protectionNeuroscientist200511540040716151042
  • GaoQLiYChoppMBone marrow stromal cells increase astrocyte survival via upregulation of phosphoinositide 3-kinase/threonine protein kinase and mitogen-activated protein kinase kinase/extracellular signal-regulated kinase pathways and stimulate astrocyte trophic factor gene expression after anaerobic insultNeuroscience2005136112313416198497
  • ShenLHLiYChenJIntracarotid transplantation of bone marrow stromal cells increases axon-myelin remodeling after strokeNeuroscience2006137239339916298076
  • AndrewsEMTsaiSYJohnsonSCHuman adult bone marrow-derived somatic cell therapy results in functional recovery and axonal plasticity following stroke in the ratExp Neurol2008211258859218440506
  • BaoXWeiJFengMTransplantation of human bone marrow-derived mesenchymal stem cells promotes behavioral recovery and endogenous neurogenesis after cerebral ischemia in ratsBrain Res2011136710311320977892
  • BaoXFengMWeiJTransplantation of Flk-1+ human bone marrow-derived mesenchymal stem cells promotes angiogenesis and neurogenesis after cerebral ischemia in ratsEur J Neurosci2011341879821692879
  • HonmaTHonmouOIihoshiSIntravenous infusion of immortalized human mesenchymal stem cells protects against injury in a cerebral ischemia model in adult ratExp Neurol20061991566615967439
  • KurozumiKNakamuraKTamiyaTMesenchymal stem cells that produce neurotrophic factors reduce ischemic damage in the rat middle cerebral artery occlusion modelMol Ther20051119610415585410
  • KohSHKimKSChoiMRImplantation of human umbilical cord-derived mesenchymal stem cells as a neuroprotective therapy for ischemic stroke in ratsBrain Res2008122923324818634757
  • ChenJSanbergPRLiYIntravenous administration of human umbilical cord blood reduces behavioral deficits after stroke in ratsStroke200132112682268811692034
  • BorlonganCVHadmanMSanbergCDSanbergPRCentral nervous system entry of peripherally injected umbilical cord blood cells is not required for neuroprotection in strokeStroke200435102385238915345799
  • WillingAELixianJMillikenMIntravenous versus intrastriatal cord blood administration in a rodent model of strokeJ Neurosci Res200373329630712868063
  • TaguchiASomaTTanakaHAdministration of CD34+ cells after stroke enhances neurogenesis via angiogenesis in a mouse modelJ Clin Invest2004114333033815286799
  • YoderMCHuman endothelial progenitor cellsCold Spring Harb Perspect Med201227114
  • IskanderAKnightRAZhangZGIntravenous administration of human umbilical cord blood-derived AC133+ endothelial progenitor cells in rat stroke model reduces infarct volume: magnetic resonance imaging and histological findingsStem Cells Transl Med20132970371423934909
  • ThomsonJAItskovitz-EldorJShapiroSSEmbryonic stem cell lines derived from human blastocystsScience19982825391114511479804556
  • Ben-DavidUBenvenistyNThe tumorigenicity of human embryonic and induced pluripotent stem cellsNat Rev Cancer20111126827721390058
  • SeminatoreCPolentesJEllmanDThe postischemic environment differentially impacts teratoma or tumor formation after transplantation of human embryonic stem cell-derived neural progenitorsStroke201041115315919940279
  • Azevedo-PereiraRLDaadiMMIsolation and purification of self-renewable human neural stem cells for cell therapy in experimental model of ischemic strokeMethods Mol Biol2013105915716723934842
  • Drury-StewartDSongMMohamadOHighly efficient differentiation of neural precursors from human embryonic stem cells and benefits of transplantation after ischemic stroke in miceStem Cell Res Ther2013449323928330
  • BühnemannCScholzABernreutherCNeuronal differentiation of transplanted embryonic stem cell-derived precursors in stroke lesions of adult ratsBrain2006129Pt 123238324817018551
  • DaadiMMMaagALSteinbergGKAdherent self-renewable human embryonic stem cell-derived neural stem cell line: functional engraftment in experimental stroke modelPLoS One200832e164418286199
  • ReyesJHO’SheaKSWysNLGlutamatergic neuronal differentiation of mouse embryonic stem cells after transient expression of neurogenin 1 and treatment with BDNF and GDNF: in vitro and in vivo studiesJ Neurosci20082848126221263119036956
  • AntonicASenaESDonnanGAHowellsDWHuman in vitro models of ischemic stroke: a test bed for translationTransl Stroke Res2012330630924323805
  • YuJVodyanikMASmuga-OttoKInduced pluripotent stem cell lines derived from human somatic cellsScience200731858581917192018029452
  • TakahashiKYamanakaSInduction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factorsCell2006126466367616904174
  • YamanakaSBlauHMNuclear reprogramming to a pluripotent state by three approachesNature2010465729970471220535199
  • MalikNRaoMSA review of the methods for human iPSC derivationMethods Mol Biol2013997233323546745
  • KawaiHYamashitaTOhtaYTridermal tumorigenesis of induced pluripotent stem cells transplanted in ischemic brainJ Cereb Blood Flow Metab20103081487149320216552
  • OkiKTatarishviliJWoodJHuman-induced pluripotent stem cells form functional neurons and improve recovery after grafting in stroke-damaged brainStem Cells20123061120113322495829
  • JiangMLvLJiHInduction of pluripotent stem cells transplantation therapy for ischemic strokeMol Cell Biochem20113541–2677521465238
  • JensenMBYanHKrishnaney-DavisonRAl SawafAZhangSCSurvival and differentiation of transplanted neural stem cells derived from human induced pluripotent stem cells in a rat stroke modelJ Stroke Cerebrovasc Dis201322430430822078778
  • PluchinoSCossettiCHow stem cells speak with host immune cells in inflammatory brain diseasesGlia20136191379140123633288
  • BhasinASrivastavaMVKumaranSSAutologous mesenchymal stem cells in chronic strokeCerebrovasc Dis Extra2011119310422566987
  • ConnickPKolappanMCrawleyCAutologous mesenchymal stem cells for the treatment of secondary progressive multiple sclerosis: an open-label phase 2a proof-of-concept studyLancet Neurol201211215015622236384
  • ReNeuron LimitedPilot Investigation of Stem Cells in Stroke (PISCES)ClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2010 [updated March 27, 2013]. Available from: http://clinicaltrials.gov/ct2/show/NCT01151124. NLM identifier: NCT01151124Accessed November 20, 2013
  • Stemedica Cell Technologies, IncA Study of Allogeneic Mesenchymal Bone Marrow Cells in Subjects With Ischemic StrokeClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2011 [updated December 13, 2012]. Available from: http://clinicaltrials.gov/ct2/show/NCT01297413?term=NCT01297413&rank=1. NLM identifier: NCT01297413Accessed November 20, 2013
  • BifariFPacelliLKramperaMImmunological properties of embryonic and adult stem cellsWorld J Stem Cells201023506021607122
  • ModoMRezaiePHeuschlingPPatelSMaleDKHodgesHTransplantation of neural stem cells in a rat model of stroke: assessment of short-term graft survival and acute host immunological responseBrain Res20029581708212468031
  • LeesJSSenaESEganKJStem cell-based therapy for experimental stroke: a systematic review and meta-analysisInt J Stroke20127758258822687044
  • KokaiaZMartinoGSchwartzMLindvallOCross-talk between neural stem cells and immune cells: the key to better brain repair?Nat Neurosci20121581078108722837038
  • MartinoGPluchinoSThe therapeutic potential of neural stem cellsNat Rev Neurosci20067539540616760919
  • MartinoGPluchinoSBonfantiLSchwartzMBrain regeneration in physiology and pathology: the immune signature driving therapeutic plasticity of neural stem cellsPhysiol Rev20119141281130422013212
  • LindenbergRRengaVZhuLLBetzlerFAlsopDSchlaugGStructural integrity of corticospinal motor fibers predicts motor impairment in chronic strokeNeurology201074428028720101033
  • SterrAShenSSzameitatAJHerronKAThe role of corticospinal tract damage in chronic motor recovery and neurorehabilitation: a pilot studyNeurorehabil Neural Repair201024541341920516488
  • CramerSCKoroshetzWJFinklesteinSPThe case for modality-specific outcome measures in clinical trials of stroke recovery-promoting agentsStroke20073841393139517332455
  • QuinnTJDawsonJWaltersMRLeesKRFunctional outcome measures in contemporary stroke trialsInt J Stroke20094320020519659822
  • CookeEVMaresKClarkATallisRCPomeroyVMThe effects of increased dose of exercise-based therapies to enhance motor recovery after stroke: a systematic review and meta-analysisBMC Med201086020942915
  • FrenchBThomasLLeathleyMDoes repetitive task training improve functional activity after stroke? A Cochrane systematic review and meta-analysisJ Rehabil Med201042191420111838
  • KurozumiKNakamuraKTamiyaTBDNF gene-modified mesenchymal stem cells promote functional recovery and reduce infarct size in the rat middle cerebral artery occlusion modelMol Ther20049218919714759803
  • ToyamaKHonmouOHaradaKTherapeutic benefits of angiogenetic gene-modified human mesenchymal stem cells after cerebral ischemiaExp Neurol20092161475519094989
  • FontesALakshmipathyUAdvances in genetic modification of pluripotent stem cellsBiotechnol Adv2013317994100123856320
  • BibleEDell’AcquaFSolankyBNon-invasive imaging of transplanted human neural stem cells and ECM scaffold remodeling in the stroke-damaged rat brain by (19)F- and diffusion-MRIBiomaterials201233102858287122244696
  • HuangYCParoliniODengLThe potential role of microvesicles in mesenchymal stem cell-based therapyStem Cells Dev201322684184423216256
  • ChaiCLeongKWBiomaterials approach to expand and direct differentiation of stem cellsMol Ther200715346748017264853
  • CramerSCParrishTBLevyRMPredicting functional gains in a stroke trialStroke20073872108211417540966
  • CramerSCNellesGBensonRRA functional MRI study of subjects recovered from hemiparetic strokeStroke19972812251825279412643
  • StinearCMBarberPASmalePRCoxonJPFlemingMKByblowWDFunctional potential in chronic stroke patients depends on corticospinal tract integrityBrain2007130Pt 117018017148468
  • CarterARShulmanGLCorbettaMWhy use a connectivity-based approach to study stroke and recovery of function?Neuroimage20126242271228022414990
  • WangLETittgemeyerMImperatiDDegeneration of corpus callosum and recovery of motor function after stroke: a multimodal magnetic resonance imaging studyHum Brain Mapp201233122941295622020952
  • Federal University of Rio de JaneiroStudy of Autologous Stem Cell Transplantation for Patients With Ischemic StrokeClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2007 [updated May 18, 2011]. Available from: http://clinicaltrials.gov/ct2/show/NCT00473057?term=NCT00473057&rank=1. NLM identifier: NCT00473057Accessed November 20, 2013
  • Manipal Acunova LtdIntravenous Autologous Bone Marrow-derived Stem Cells Therapy for Patients With Acute Ischemic StrokeClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2011 Available from: http://clinicaltrials.gov/ct2/show/NCT01501773?term=NCT01501773&rank=1. NLM identifier: NCT01501773Accessed November 20, 2013
  • Chaitanya Hospital, PuneA Clinical Trial to Study the Safety and Efficacy of Bone Marrow Derived Autologous Cell for the Treatment of Stroke (BMACS)ClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2013 [updated April 12, 2013]. Available from: http://clinicaltrials.gov/ct2/show/NCT01832428?term=NCT01832428&rank=1. NLM identifier: NCT01832428Accessed November 20, 2013
  • National University of MalaysiaIntravenous Autologous Mesenchymal Stem Cells Transplantation to Treat Middle Cerebral Artery InfarctClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2011 [updated October 26, 2011]. Available from: http://clinicaltrials.gov/ct2/show/NCT01461720?term=NCT01461720&rank=1. NLM identifier: NCT01461720Accessed November 20, 2013
  • University of California, IrvineSafety of Escalating Doses of Intravenous Bone Marrow-Derived Mesenchymal Stem Cells in Patients With a New Ischemic StrokeClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2013 [updated May 8, 2013]. Available from: http://clinicaltrials.gov/show/NCT01849887. NLM identifier: NCT01849887Accessed November 20, 2013
  • Wenzhou Medical UniversityAutologous Bone Marrow Mesenchymal Stem Cell Transplantation for Chronic StrokeClinicalTrials gov [website on the Internet]Bethesda, MDUS National Library of Medicine2012 [updated October 22, 2012]. Available from: http://clinicaltrials.gov/show/NCT01714167. NLM identifier: NCT01714167Accessed November 20, 2013
  • Samsung Medical CenterThe STem Cell Application Researches and Trials In NeuroloGy-2 (STARTING-2) StudyClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2012 [updated August 11, 2013]. Available from: http://clinicaltrials.gov/ct2/show/NCT01716481?term=NCT01716481&rank=1. NLM identifier: NCT01716481Accessed November 20, 2013
  • University Hospital, GrenobleIntravenous Stem Cells After Ischemic Stroke (ISIS)ClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2009 [updated September 4, 2013]. Available from: http://clinicaltrials.gov/ct2/show/NCT00875654?term=NCT00875654&rank=1. NLM identifier: NCT00875654Accessed November 20, 2013
  • Hospital Universitario Central de AsturiasAutologous Bone Marrow Stem Cells in Middle Cerebral Artery Acute Stroke TreatmentClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2008 [updated November 27, 2011]. Available from: http://clinicaltrials.gov/ct2/show/NCT00761982?term=NCT00761982&rank=1. NLM identifier: NCT00761982Accessed November 20, 2013
  • Imperial College LondonAutologous Bone Marrow Stem Cells in Ischemic StrokeClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2007 [updated July 2, 2012]. Available from: http://clinicaltrials.gov/ct2/show/NCT00535197?term=NCT00535197&rank=1. NLM identifier: NCT00535197Accessed November 20, 2013
  • Celgene CorporationStudy of Human Placenta-derived Cells (PDA001) to Evaluate the Safety and Effectiveness for Patients With Ischemic StrokeClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2011 [updated August 12, 2013]. Available from: http://clinicaltrials.gov/ct2/show/NCT01310114?term=NCT01310114&rank=1. NLM identifier: NCT01310114Accessed November 20, 2013
  • Ageless Regenerative InstituteStudy to Assess the Safety and Effects of Autologous Adipose-Derived Stromal Cells in Patients After StrokeClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2011 [updated October 28, 2013]. Available from: http://clinicaltrials.gov/ct2/show/NCT01453829?term=NCT01453829&rank=1. NLM identifier: NCT01453829Accessed November 20, 2013
  • Instituto de Investigación Hospital Universitario La PazReparative Therapy in Acute Ischemic Stroke With Allogenic Mesenchymal Stem Cells From Adipose Tissue, Safety Assessment, a Randomised, Double Blind Placebo Controlled Single Center Pilot Clinical Trial (AMASCIS-01)ClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2012 [updated September 6, 2013]. Available from: http://clinicaltrials.gov/ct2/show/NCT01678534?term=NCT01678534&rank=1. NLM identifier: NCT01678534Accessed November 20, 2013
  • The University of Texas Health Science Center, HoustonSafety/Feasibility of Autologous Mononuclear Bone Marrow Cells in Stroke PatientsClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2009 [updated May 4, 2012]. Available from: http://clinicaltrials.gov/ct2/show/NCT00859014?term=NCT00859014&rank=1. NLM identifier: NCT00859014Accessed November 20, 2013
  • Zhejiang HospitalAutologous Hematopoietic Stem Cell Transplantation in Ischemic Stroke (AHSCTIS)ClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2012 [updated January 24, 2012]. Available from: http://clinicaltrials.gov/ct2/show/NCT01518231?term=NCT01518231&rank=1. NLM identifier: NCT01518231Accessed November 20, 2013
  • China Medical University HospitalStudy of Purified Umbilical Cord Blood CD34+ Stem Cell on Chronic Ischemic StrokeClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2011 [updated July 31, 2012]. Available from: http://clinicaltrials.gov/ct2/show/NCT01438593?term=NCT01438593&rank=1. NLM identifier: NCT01438593Accessed November 20, 2013
  • Athersys, IncStudy to Examine the Effects of MultiStem in Ischemic StrokeClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2011 [updated August 22, 2013]. Available from: http://clinicaltrials.gov/ct2/show/NCT01436487?term=NCT01436487&rank=1. NLM identifier: NCT01436487Accessed November 20, 2013
  • AldagenStudy of ALD-401 Via Intracarotid Infusion in Ischemic Stroke SubjectsClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2011 [updated July 29, 2013]. Available from: http://clinicaltrials.gov/ct2/show/NCT01273337?term=NCT01273337&rank=1. NLM identifier: NCT01273337Accessed November 20, 2013
  • SanBio, IncA Study of Modified Stem Cells in Stable Ischemic StrokeClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2011 [updated August 30, 2013]. Available from: http://clinicaltrials.gov/ct2/show/NCT01287936?term=NCT01287936&rank=1. NLM identifier: NCT01287936Accessed November 20, 2013
  • Southern Medical University, ChinaAutologous Bone Marrow Stromal Cell and Endothelial Progenitor Cell Transplantation in Ischemic Stroke (AMETIS)ClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2011 [updated November 8, 2011]. Available from: http://clinicaltrials.gov/ct2/show/NCT01468064?term=NCT01468064&rank=1. NLM identifier: NCT01468064Accessed November 20, 2013