Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Mar 12.
Published in final edited form as: Curr Neurol Neurosci Rep. 2019 Feb 2;19(1):5. doi: 10.1007/s11910-019-0920-4

Promoting Brain Repair and Regeneration After Stroke: A Plea for Cell-based Therapies

Ania Dabrowski 1, Thomas J Robinson 1, Ryan J Felling 1,*
PMCID: PMC7067448  NIHMSID: NIHMS1067354  PMID: 30712068

Abstract

Purpose of Review:

After decades of hype, cell-based therapies are emerging into the clinical arena for the purposes of promoting recovery after stroke. In this review we discuss the most recent science behind the role of cell-based therapies in ischemic stroke and the efforts to translate these therapies into human clinical trials.

Recent Findings:

Preclinical data support numerous beneficial effects of cell-based therapies in both small and large animal models of ischemic stroke. These benefits are driven by multifaceted mechanisms promoting brain repair through immunomodulation, trophic support, circuit reorganization, and cell replacement.

Summary:

Cell-based therapies offer tremendous potential for improving outcomes after stroke through multimodal support of brain repair. Based on recent clinical trials, cell-based therapies appear both feasible and safe in all phases of stroke. Ongoing translational research and clinical trials will further refine these therapies and have the potential to transform the approach to stroke recovery and rehabilitation.

Keywords: neurogenesis, stem cells, transplantation, stroke recovery, neuroplasticity, brain regeneration

Introduction

Stem cell therapies are emerging in the clinical arena, and bringing with them renewed hope for novel therapeutic approaches to promoting brain repair after stroke. The concept of regenerative medicine in central nervous system injury dates back more than a century, when Santiago Ramon y Cajal observed, “In adult centers the nerve paths are something fixed, ended, immutable. Everything may die, nothing may be regenerated. It is key for the science of the future to change, if possible, this decree.”[1] Over several decades we have learned much about the potential for regeneration in the CNS, with the recognition of neural stem and progenitor cells (NSPs) persisting in the brain throughout life. Reynolds and Weiss first demonstrated the ability to isolate multipotential progenitors from the brains of adult rodents.[2] Animal models then demonstrated increased neurogenesis from these progenitors after stroke in both the immature and aged brain. Attention has more recently turned toward transplantation of exogenous cells to support and augment endogenous repair mechanisms. Originally stymied by ethical considerations surrounding the use of embryonic stem cells (ESCs), the brakes have been released by a plethora of mechanisms for generating neural progenitors from adult tissues. These include most notably induced pluripotent stem cells (iPSCs) which can be generated from an individual’s own somatic cells. Today we have tremendous capabilities to generate many different specific cell types. In many ways this has outpaced our ability to study the effects of different cell types as means of therapy. In this review article we will discuss the variety of cell-based therapies under investigation, possible mechanisms of action, and the current evidence available from human clinical trials. Finally we propose a roadmap for future research to accelerate the development and optimization of cell-based therapies as critical treatments for stroke recovery.

Pathways for Cell-Based Therapies

The term “stem cell” has existed in the literature for more than a century, and by strict definition necessitates the characteristic capacities for self-renewal and differentiation into other cell types.[3] Stem cells range from pluripotent ESCs from which entire organisms arise, to more restricted organ-specific stem cells. Experimental observations also suggest that stem cells and their progeny exist on a continuum, with at least some potential of bidirectional phenotypic lability.[4] As applied to regenerative medicine, the key characteristics of stem cells present a double-edged sword. The expansion and multipotential differentiation capacities are therapeutically promising, but also present the feared possibility of tumorigenicity.[5-7] Many of the cell types that have been investigated in stroke have been are either more restricted progenitors or stem cells that have been modified to limit this risk, but nonetheless are commonly referred to collectively as stem cell therapy.

Exogenous Cell Administration

ESCs are derived from blastocysts and represent the most pluripotent cell state available for potential therapeutic purposes.[8-10] This pluripotency also raises concerns regarding tumorigenicity following transplantation.[5, 11] These cells can be directed in vitro toward neural lineages, as reviewed elsewhere.[12-14] Most experimental approaches have used such directed differentiation prior to transplantation to reduce the risk of uncontrolled expansion. After transplantation in preclinical stroke models, ESCs have can engraft and survive for up to 12 weeks.[15-18] Some studies have demonstrated migration of transplanted cells whether transplanted ipsilesionally or contralesionally,[16] but others have not observed significant migration.[15] These cells can differentiate into multiple neuronal subtypes as well as glia,[16, 17] develop electrophysiological properties of mature neurons,[16] and form structural connections within the host brain.[18]

NSPs are more restricted stem cells. They are able self-replicate, but differentiation is restricted neuronal and glial subtypes.[19, 20] In addition to ESCs and iPSCs, NSPs can be derived from fetal and adult tissue.[21, 22] Adult NSPs reside in the subventricular zone (SVZ) in the wall of the lateral ventrical and the subgranular zone (SGZ) of the dentate gyrus in the hippocampus.[23] While in general considered multipotential, NSPs may actually have region-specific lineage restrction.[24] NSPs have been administered directly into the brain either through stereotactic neurosurgery or intra-arterially in preclinical animal models of stroke, and a recent meta-analysis found many pleiotropic benefits on behavioral and structural outcomes.[25, 26]

Mesenchymal stem cells (MSCs) reside in tissue of mesodermal lineage such as adipose tissue, bone marrow, umbilical cord blood, and others.[27] The first identified and most commonly used MSCs are bone-marrow derived MSCs, a subset of bone marrow mononuclear cells (BMMNCs).[28] Along with the ability to differentiate into a range of mesenchymal tissue, MSCs can also differentiate into ectodermal and endodermal lineages, including neural cells.[29, 30] This possibly due to an even more specific subset of MSCs, the recently described multilineage-differentiating stress enduring (Muse) cells that comprise a small portion of bone marrow-derived MSCs.[31] These cells may also play a role in the unique ability of MSCs to migrate towards areas of injury and spontaneously differentiate and integrate with damaged tissue. [32, 31, 33] MSCs can be isolated and expanded from patients as an autologous source of cells, thus reducing the risk of immune system activation.[34-36] MSCs have both anti-inflammatory and neurotrophic effect with the ability to secrete multiple factors including BDNF, NGF, FGF, and VEFG.[37]

Induced pluripotent stem cells (iPSCs) are dedifferentiated somatic cells, most commonly fibroblasts, transformed via induction of defined transcription factors.[38-40] Similar to ESCs, iPSCs are returned to their pluripotent state and have the ability to differentiate into different neuronal cell types, including NSCs.[41, 42] Unlike ESCs, however, autologous iPSCs have less immunogenicity due to their derivation from the patient’s own tissue, avoid the moral and legal issues surrounding the cultivation and use of ESCs, and afford nearly limitless customization.[43-45]. Transplantation of iPSC-derived NSPs leads to regeneration of mature and functional neurons and axonal projections through trophic support and enhances neurogenesis and angiogenesis following ischemic stroke, promoting improved neurologic outcomes.[46-49]

Endogenous Neurogenesis

Once considered to be a static organ, we now know that the brain has the capacity to generate new cells during postnatal neurodevelopment and long after. Joseph Altman first demonstrated new cells being born in the adult rodent brain using 3H-thymidine incorporation assays.[50] Kaplan and Hinds later confirmed similar results demonstrating newly born neurons in the rat dentate gyrus and olfactory bulb using electron microscopy.[51] Adult neurogenesis is now a well-established feature of the rodent brain, occurring in discrete neurogenic niches: the subventricular zone of the lateral ventricles and the subgranular zone of the dentate gyrus.[52, 53] Despite these early findings, the issue of adult neurogenesis remained contentious due to the unknown source of these cells and primate research that suggested adult neurogenesis may be limited to rodents.[54] Postnatal neurogenesis has since been confirmed in the human hippocampus, taking advantage of patients who had received the thymidine analog bromodeoxyuridine (BrdU) as chemotherapy and thus labeled newborn cells at the time of treatment.[55] More recent studies suggest that basal levels of endogenous neurogenesis in humans are very low, with the possible exception of the perinatal period.[56, 57] Animal models have repeatedly demonstrated increased neurogenesis after stroke, both in immature and adult rodents.[58-63] Key questions remain as to the functional importance of this apparent regenerative response, but numerous studies have demonstrated correlations between behavioral recovery after stroke.[64] In humans, evidence is much more sparse given technical limitations, but some studies hint that a similar phenomenon may occur.[65, 66]

Other avenues for promoting cell-based therapy

In addition to stem cells themselves, a number of adjunctive technologies are emerging with the potential to further advance these therapies. The use of bioscaffolds such as biologically derived and synthetic hydrogels greatly aid in the transplantation and subsequent survivability of exogenous stem cells in the stroke cavity.[67, 68] These substances allow for in situ tissue regeneration and provide a non-reactive matrix that can act both as structural support system for stem cells as well as a vehicle for drug delivery.[69, 70] Imaging techniques including optical imaging, magnetic resonance imaging (MRI), and positron emission tomography (PET) offer the ability to track and monitor cells from the point of administration[71-73] Cells to be transplanted are labelled with magnetic markers, typically superparamagnetic iron oxide (SPIO) nanoparticles. In addition to the primary function as MRI markers, SPIO-labeled cells can be physically manipulated via an external magnet through fluid compartments, potentially indicating a method of manual direction through the ventricular system.[74]. These technologies may open even further avenues for the application of cell-based therapies in stroke.

Mechanisms of Action

The holy grail of stem cell therapy is to replace cells that are lost or damaged as a consequence of disease or injury. In the context of stroke, this is an enormous ask given that a stroke indiscriminately destroys all brain tissue, often leaving behind a region devoid of the infrastructure that was laid down during development. In order to achieve cell replacement, therapies will have to accomplish (i) delivery of cells to the infarct territory; (ii) allowing or promoting the differentiation of those cells into a diverse population including various types of neurons, glia, and blood vessels; and (iii) re-establishment of complex connections and networks both locally and remotely. Fortunately cell-based therapies provide numerous mechanisms for enhancing repair of the brain following injury, independently of actual cell replacement.[75]

Modulation of neuroinflammation

Stroke represents an evolution of injury over time, from acute necrosis due during ischemia to secondary cell death due to inflammation.[76] An overly simplistic view of inflammation would suggest that proinflammatory cytokines and the cellular immune response aggravate injury, impair neurogenesis, and impede neural repair after stroke.[77, 78] The true interaction between inflammation and the regenerative response to the brain is likely much more complex, and some inflammatory mediators may actually help to promote repair.[79, 80] Microglia play a biphasic role in ischemic stroke with shifting polarization between pro-inflammatory and anti-inflammatory phenotypes, a phenomenon that can be targeted therapeutically with cell-based therapy in preclinical models.[81-83] Accumulating evidence in both humans and animals support a significant role for immunomodulation as one pillar of stem cell therapies in enhancing recovery after stroke.[84-88] This mechanism of action is particularly applicable to peripherally administered stem cells because they can exert their effects through the systemic immune system rather than requiring direct localization near the stroke.

Remodeling of Neural Networks and Cell Replacement

Data from animal models human patients suggests that after ischemic stroke neural circuitry in areas surrounding damaged tissue reorganizes to regain previously lost function.[89] These changes include axon sprouting, dendritic remodeling, and new synapse formation, and can be facilitated by functionally-directed rehabilitation.[90-94] Expanding evidence suggests that stem cells promote neural circuit regeneration through multiple intertwined mechanisms, promoting repair reorganization of existing cells as well as limited incorporation of new cells into the regenerating circuit.

One important mechanism through which stem cells promote neural circuit remodeling is secretion of neurotrophic factors. Infusion of mesenchymal stem cells engineered to express brain-derived neurotrophic factor (BDNF), placental growth factor (PGF), glial cell-line derived neurotrophic factor (GDNF), or vascular endothelial growth factor (VEGF) and angiopoietin into rodent models of ischemic stroke improved functional outcomes.[95-98] The functional improvement in these experiments correlated with decreased infarct volume and improved vascular regrowth into the injured parenchyma. Although cells can be engineered to overexpress neurotrophic factors, MSCs exposed to the ischemic post-stroke environment also appear to inherently upregulate production of neurotrophic factors.[99] Neurotrophic factors are known to be crucial to neural circuit development at sequential stages of development, from promoting neurogenesis, through dendrite and axon growth, to synaptogenesis and synaptic refinement.[100] Cell-based therapies may act in part by re-inducing developmental programs of neural circuit formation.[101] Emerging evidence also suggests that exosomes may provide a critical mechanism by which stem cells exert their effects in promoting remodeling after injury.[102]

Indeed, all of the anticipated effects of neurotrophic signaling in the stroke-damaged brain have been observed after stem cell transplant. When transplanted into the ischemic brain, exogenous NSPs can augment neurogenesis and angiogenesis from resident precursors thus increasing the population of cells that may potentially be integrated into the recovering circuit.[103-105] Transplanting human NSPs into stroke-injured brain also promotes remodeling of both neuronal axons and dendrites, with increased connectivity within damaged circuits and improved axon function as evidenced by increased cargo transport along the length of axons.[106] Accompanying in vitro studies suggest that these effects were at least in part mediated by VEGF and thrombospondin.

Bystander or paracrine effects are clearly important factors underlying the efficacy of stem cells in promoting repair and regeneration, but cell replacement likely has a role as well. Arvidsson and colleagues observed that less than 20% of newly generated cells survived and matured into NeuN-expressing neurons.[59] Despite this sobering fact, a minority of cells do survive, migrate into sites of injury, and even functionally integrates into local circuitry, developing similar electrophyiological signatures compared to pre-existing neighbors.[107] There is evidence that stem cells can generate mature neurons that form functional afferent and efferent connections. Neural precursor cells derived from explants of immature medial ganglionic eminence (which developmentally is the source of inhibitory interneurons) were directly implanted into stroke-damaged brain, and found to differentiate into neurons.[108, 109] These explant-derived neurons received functional synaptic connections, as measured electrophysiologically by postsynaptic potentials, and were able to generate action potentials, although the target of their connectivity was not defined. Following transplantation, iPSCs that had been primed toward cortical neuronal phenotypes also functionally integrate into damaged circuitry following transplantation.[49] These cells differentiated into both excitatory and inhibitory mature neurons (as assessed both immunohistochemically and electrophysiologically) and received functional synaptic inputs from native cortex. While most effort has emphasized neuronal production, some investigators have also observed oligodendrogenesis.[110] Understanding the role of glia in both promoting and limiting regeneration in the brain will be critical for further promotion of cell-based therapy.[111, 112]

Clinical Trials of Cell-based Therapy in Stroke

Based upon encouraging results from preclinical studies of cell-based therapies in animal models of stroke, investigators have embarked on pioneering human studies over the past two decades. Most of these have been small, open-label, single arm studies. Table 1 summarizes many of the published trials to date. The majority of these clinical trials have been initial phase I/II trials of feasibility and safety, with small numbers of patients and often not randomized or controlled.

Table 1.

Published clinical trials of cell-based therapies in ischemic stroke. Quality of evidence provided is based on GRADE assessment of the trial design.[142] Phases of stroke: Acute = within the first week; Subacute = 1 week to 3 months; Chronic = more than 3 months. Abbreviations: MSC – mesenchymal stem cells; BMMNC – bone marrow mononuclear cells; hNSP – human neural stem/progenitor cells; SAE – significant adverse event; mRS – modified Rankin Scale; BI – Barthel Index; FM – Fugl Meyer scale; ESS – European Stroke Scale; NIHSS – NIH Stroke Scale.

    Stroke phase Cell type Mode of delivery Treatment Control Safety concerns Outcomes Quality of evidence
Kondziolka[114] 2000 Chronic
6m-5y
hNSP Surgical 12 N/A  No SAE -Improved ESS mean 2.9 pts at 6 months
-No change in NIHSS or BI at 6 months
-No change in infarct volume
-Increased [F18]FDG uptake on PET in 6/11
Low
-open label
-single arm
Kondziolka[113] 2005 Chronic
1-5y
hNSP Surgical 18 4
No sham
 No SAE -Improvement in ESS mean 2.7 pts at 6 months
compared to 0.75pts in control group (p=0.148)
Moderate
-randomized
-observer blinded
Rabinovich[129] 2005 Chronic
4m-2y
Fetal tissue Intrathecal 10 11
No sham
Fever, meningismus
within 48h
transplant
 -Improved Karnovskii functional activity scale, quality of life Very low
-nonrandomized
-unblinded
-nonstandard outcome measure
Savitz[115] 2005 Chronic
1.5-10y
porcine lateral ganglionic eminence cells Surgical 5 N/A Enhancing MRI lesions in 2/5 patients -Improvement of NIHSS by 1 point over 4 years in 3/5 patients and by 4 points in 1 patient
-Subjective functional improvement in 3/5 patients
Very low
-open label
-single arm
-subjective outcome
Bang[130] 2005 Subacute
4-5w
autologous MSC Intravenous 5 25 No SAE -Improved BI at 3 and 6m (not significant at 12m) Moderate
-randomized
-controlled
-observer blinded
Suarez-Monteguido[131] 2009 Chronic
3y-8y
BMMNC Surgical 5 N/A No SAE -Imporved motor function (NIHSS and SSS)
-Improved spasticity 1.Ashworth)
-Improved BI (mean 4 pts at 6m, 10 pts at 12m)
 Very low
-small number
-single arm
-multiple outcomes
Lee[132] 2010 Subacute
4-5w
autologous
MSC
Intravenous 16 36
No sham
No difference v. ctl -Improved mRS shift
-Improved survival (HR 0.344)
Moderate
-randomized
-controlled
-observer blinded
Savitz[133] 2011 Acute
24-72h
autologous
BMMNC
Intravenous 10 historical No SAE -710 patients with better 90d mRS than expected based on historical controls
−5/9 mRS 0-2 at 6m
−7/9 BI ≥90 at 6m
Very Low
-single arm
-unblinded
Honmou[120] 2011 subacute to chronic
43-133d
autologous
MSC
Intravenous 12 N/A No SAE -Increased daily rate of change in NIHSS during first week after infusion
−20% reduction in lesion size 1 week after infusion
Very Low
-single arm
-unblinded
Friedrich[134] 2012  Acute
3-7d
autologous
BMMNC
Intravenous 20 N/A  No SAE -7/20 patients with “satisfactory” improvement in NIHSS at 24 hours or mRS at 90 days. Very Low
-single arm
-unblinded
Moniche[135] 2012 Acute
5-9d
Autologous
BMMNC
Intraarterial 10 10
No sham
Seizure in 2 treated patients at 3m
-no significant difference in mRS or BI at 90d
-increased BNGF at 8 days compared to controls
Low
-Nonrandomized
-Controlled
-Observer blinded
Bhasin[136] 2011 Chronic
3m-1y
Autologous MSC Intravenous 6 6
Matched for age, chronicity, lesion size, severity
No SAE -no significant difference in FM, BI, Ashworth at 6m Low
-Nonrandomized
-Controlled
-Unblinded
Bhasin[137] 2012 Chronic
3m-2y
autologous BMMNC Intravenous 12 12 No SAE  -improved BI at 24w Low
-Nonrandomized
-Controlled
-Unblinded
Bhasin[138] 2013  Chronic
3m-2y
 Autologous MSC v BM-
MNC
 Intravenous  20 20  No SAE -Improvement in 24w BI compared to controls  Low
-Nonrandomized
-Controlled
-Unblinded
-multiple comparisons
Bhasin[139]* 2017 Chronic
3m-1y
Autologous
MSC
Intravenous 6 6 No SAE -No significant difference in FM or BI at 4y Low
-Nonrandomized
-Controlled
-Unblinded
Wang[140] 2013 Chronic
1-7y
autologous
BMMNC
Intravenous 8 N/A No SAE -Mean decrease NIHSS 3.1pts at 12m
-Mean increase BI 20.6pts at 12m
Very Low
-Single arm
-Unblinded
Prasad[141] 2014 Subacute
1-4w
autologous
MSC
Intravenous 58 60
No sham
 No SAE  -No significant difference in mRS shift or BI at 6m  Moderate
-Randomized
-Observer blinded
Kalladka[118] 2016 Chronic
6-60m
hNSC Surgical 11
Dose-escalation
N/A 2 SAE related to procedure (not to the cells) -improved mRS by 1 grade in 4/11 patients Low
-Single arm
-Unblinded
Steinberg[119] 2016 Chronic
6-60m
Allogeneic modified MSC Surgical 18
Dose-escalation
N/A 2 AEs definitely or probably related to surgery, none related to cells -Increase mean ESS by 6.88 points at 12m
-Decrease mean NIHSS by 2 points at 12m
-Increase mean total FM by 19.2 points at 12m
-Increase mean motor FM 11.4 points at 12m
-no change in mRS at 12m
-no clear dose response
Low
-Single arm
-Unblinded
Hess[85] 2017 Acute
24-48h
MAPC
(MultiStem)
Intravenous 65 61
Placebo
23% treatment-related adverse event (halitosis, fever/chills, nausea -Primary outcome (mRS≤2, NIHSS improvement ≥ 75%, BI≥95) not significantly different at 90d
-BI≥95 favored MAPC at 1y
-mRS≤1 favored MAPC at 1y
-Excellent outcome (mRS≤1, NIHSS≤1, & BI≥95) favored MAPC at 1y
-Secondary outcomes more strongly significant for patients treated within 36h

 High
-Randomized
-double-blinded
-placebo-controlled

In one of the earliest efforts, Kondziolka and colleagues investigated the effects of stereotactic transplantation of human embryonic carcinoma-derived precursor cells (termed LBS-neurons) in chronic basal ganglia stroke. In their first study they found slight improvements in the European Stroke Scale at 6 months compared to the patients’ baseline, but in their follow up phase II study there were no significant differences between transplanted patients and control patients.[113, 114] In both studies there were no adverse cell-related events, although procedure-related complications did occur. One of the major criticisms of these studies was the use of a cancer-derived cell line and the risk for tumorigenicity given limited follow up of only one year. This led to a pilot study of porcine embryonic precursor cells derived from the lateral ganglionic eminence, but this study was terminated by the FDA after 2/5 patients developed adverse events.[115]

An alternative approach has utilized an immortalized human neural stem cell line derived from fetal cortical brain tissue (CTX0E03 cell line). These cells have been engineered with a retrovirally-delivered c-mycERTAM transgene to allow large scale expansion and banking.[116] In vivo models have demonstrated rapid epigenetic silencing of this transgene within the first week after transplantation, supporting a low risk of uncontrolled expansion and tumorigenicity.[117] In a phase I safety trial (PISCES), Kalladka and colleagues transplanted increasing doses of these cells into the ipsilesional putamen of 11 men with ischemic stroke 14-51 months prior to enrollment.[118] Importantly their trial did not include immunosuppression as preclinical models suggested that low immunogenic responses to the CTX0E03 cells. The primary outcome of this phase I trial was safety, and they saw no significant adverse events that they attributed to the cell therapy, but several related to the neurosurgical procedure. While not powered or designed for efficacy, several patients did experience improvements in multiple measurement scales including the modified Rankin scale. While typically patients are not expected to make significant improvements at the timepoints in this study, it is not possible to attribute causality to the cell therapy in the absence of a control group. An important caveat to this study is that only men were included to reduce the risk of incidental exposure to tamoxifen, a commonly used treatment for breast cancer, because the transgene is under control of a tamoxifen-inducible promoter. Whether the safety of this treatment will be generalizable to women remains to be seen. A phase II has recently been completed, but not yet published. The company’s website indicates that although the primary endpoint was not met, but enough benefits were observed in some subjects to prompt planning of a definitive trial.

The previously described studies used neuronal precursors, others MSC transplantation has also been explored in clinical trials. Steinberg and colleagues used bone marrow-derived MSCs that had been transiently transfected with Notch1 to promote differentiation to a neuronal lineage.[119] These cells were stereotactically implanted to multiple locations in the peri-infarct tissue under MRI guidance, with a goal of bracketing the stroke with stem cells. Transplantations were performed in the chronic phase at a mean of 22 months after stroke (range 7-36). Similar to prior studies, adverse events were rare and largely attributable to the neurosurgical procedure rather than the cells. In 18 patients, there were only 4 treatment-emergent adverse events that were possibly related to cell therapy and none that were probably or definitely deemed attributable to cell therapy, but there were 22 adverse events with a possible/probable/definite relationship to the neurosurgical procedure (most commonly headache). Similar to the PISCES trial, it is difficult to draw strong conclusions on efficacy in the absence of a control group, but the investigators observed statistically significant improvements in the European Stroke Scale, the NIH Stroke Scale, and the Fugl-Meyer at timepoints when substantial improvement would typically be unexpected.

Honmou and colleagues investigated IV infusion of autologous MSCs in the subacute to chronic phase of ischemic stroke, and they observed no significant adverse effects.[120] Interestingly they did see an increased rate of improvement in NIHSS in the first 1-2 weeks post-infusion, but there was no control group and evaluators were not blinded. Additionally, many of these patients received infusion within 3 months after stroke, a time window in which some spontaneous recovery of impairment is expected. They also saw progressive reduction of lesion volumes, reaching a mean of 20% reduction at 1 week post-infusion compared to 1 day after infusion, at a time when such lesion evolutions may not be expected.[121]

The application of cell-based therapies during the acute phase of stroke has mostly been limited to systemic administration of bone marrow derived precursors (MSCs, MAPCs, BMMCs). The MASTERS trial is one of the largest studies to date and was performed in a multicenter, placebo-controlled, double-blinded fashion.[85] Bone marrow derived stem/progenitor cells were administered intravenously between 24 and 48 hours after stroke onset. There was no difference in the primary or secondary safety endpoints of dose-limiting toxicity, neurological worsening due to the investigational product, secondary infections, or laboratory/cardiac abnormalities. While overall the frequency of treatment emergent adverse events were more common in the treatment group, these were mostly deemed mild to moderate. The primary efficacy endpoint was the multivariate global stroke recovery at 90 days (mRS ≤ 2, 75% improvement in NIHSS, and Barthel Index ≥ 95). Exploratory analyses suggested benefit in terms of excellent outcome (defined as mRS ≤ 1, BI ≥ 95, and NIHSS ≤ 1) at one year. Additionally when considering only those patients treated within 24-36 hours, mRS shift analysis and excellent outcome at 90 days both favored MAPC treatment, and the one year outcomes were even more strongly in favor of MAPC treatment. The authors interpretation of these results posited that MAPC treatment may ameliorate secondary inflammation after stroke, and that these benefits may take even more time to become evident than our typical 90 day outcomes. They also note the suggestion that time window of treatment may be important.

Conclusions

The momentum behind cell-based therapies for stroke recovery remains substantial, but while early studies have shown hints of promise true efficacy has not yet been achieved. In 2007, investigators from academia, government, and industry convened a consortium to lay a path forward, and from this emerged the Stem Cells as an Emerging Paradigm in Stroke (STEPS) series of guidelines.[122-124] Preclinical studies have shown that stem cells through both immunomodulatory mechanisms and through post-stroke neural circuit remodeling in a twofold manner: by enhancing mechanisms of intrinsic circuit remodeling (secreting neurotrophic factors, increasing neurogenesis, and promoting plasticity), and by maturating into neurons that directly incorporate into the neural circuit. Whether these mechanisms are independent or synergistically bound requires further exploration. While the quality of clinical evidence remains limited, safety and feasibility have been demonstrated for multiple cell types, routes of administration, and times of administration. Future studies should establish biomarkers so that as clinical trials progress we will be able to re-evaluate biological targets to optimize efficacy. An iterative process between the clinic and the laboratory is essential to refine the approach for cell-based therapy and ultimately reach the desired endpoints. Bioengineering advances promise to allow customization of both cells and scaffolds to enhance therapeutic benefits.[125-127] No therapies in current standard clinical practice improve outcomes beyond the proportional recovery expected from spontaneous biological repair mechanisms.[128] Cell-based therapies offer the potential to dramatically shift the paradigm of stroke rehabilitation and recovery. It is imperative that we continue to refine and drive these therapies toward the goal of improving functional restoration in our patients.

Acknowledgments

Disclosures

The authors have received grant support from NIH/NINDS for stem cell research (K08NS097704 awarded to RJF). The authors have no conflicts of interest. All reported studies/experiments with human or animal subjects performed by the authors have been previously published and complied with all applicable ethical standards(including the Helsinki declaration and its amendments, institutional/national research committee standards, and international/national/institutional guidelines).

References

  • 1.Ramon y Cajal S. Degeneration and regeneration of the nervous system. London: Oxford University Press; 1928. [Google Scholar]
  • 2.Reynolds BA, Weiss S. Generation of neurons and astrocytes from isolated cells of the adult mammalian central nervous system. Science. 1992;255(5052):1707–10. [DOI] [PubMed] [Google Scholar]
  • 3.Ramalho-Santos M, Willenbring H. On the origin of the term “stem cell”. Cell stem cell. 2007;1(1):35–8. doi: 10.1016/j.stem.2007.05.013. [DOI] [PubMed] [Google Scholar]
  • 4.Quesenberry PJ, Colvin G, Dooner G, Dooner M, Aliotta JM, Johnson K. The stem cell continuum: cell cycle, injury, and phenotype lability. Annals of the New York Academy of Sciences. 2007;1106:20–9. doi: 10.1196/annals.1392.016. [DOI] [PubMed] [Google Scholar]
  • 5.Blum B, Benvenisty N. The tumorigenicity of human embryonic stem cells. Adv Cancer Res. 2008;100:133–58. doi: 10.1016/S0065-230X(08)00005-5. [DOI] [PubMed] [Google Scholar]
  • 6.Kawai H, Yamashita T, Ohta Y, Deguchi K, Nagotani S, Zhang X et al. Tridermal tumorigenesis of induced pluripotent stem cells transplanted in ischemic brain. Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism. 2010;30(8):1487–93. doi: 10.1038/jcbfm.2010.32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Chen SJ, Chang CM, Tsai SK, Chang YL, Chou SJ, Huang SS et al. Functional improvement of focal cerebral ischemia injury by subdural transplantation of induced pluripotent stem cells with fibrin glue. Stem Cells Dev. 2010;19(11):1757–67. doi: 10.1089/scd.2009.0452. [DOI] [PubMed] [Google Scholar]
  • 8.Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS et al. Embryonic stem cell lines derived from human blastocysts. Science. 1998;282(5391):1145–7. [DOI] [PubMed] [Google Scholar]
  • 9.Ilic D, Ogilvie C. Concise Review: Human Embryonic Stem Cells-What Have We Done? What Are We Doing? Where Are We Going? Stem cells. 2017;35(1):17–25. doi: 10.1002/stem.2450. [DOI] [PubMed] [Google Scholar]
  • 10.Ware CB. Concise Review: Lessons from Naive Human Pluripotent Cells. Stem cells. 2017;35(1):35–41. doi: 10.1002/stem.2507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Erdo F, Buhrle C, Blunk J, Hoehn M, Xia Y, Fleischmann B et al. Host-dependent tumorigenesis of embryonic stem cell transplantation in experimental stroke. Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism. 2003;23(7):780–5. doi: 10.1097/01.WCB.0000071886.63724.FB. [DOI] [PubMed] [Google Scholar]
  • 12.Peljto M, Wichterle H. Programming embryonic stem cells to neuronal subtypes. Current opinion in neurobiology. 2011;21(1):43–51. doi: 10.1016/j.conb.2010.09.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Gaspard N, Vanderhaeghen P. Mechanisms of neural specification from embryonic stem cells. Current opinion in neurobiology. 2010;20(1):37–43. doi: 10.1016/j.conb.2009.12.001. [DOI] [PubMed] [Google Scholar]
  • 14.Elkabetz Y, Studer L. Human ESC-derived neural rosettes and neural stem cell progression. Cold Spring Harb Symp Quant Biol. 2008;73:377–87. doi: 10.1101/sqb.2008.73.052. [DOI] [PubMed] [Google Scholar]
  • 15.Hicks AU, Lappalainen RS, Narkilahti S, Suuronen R, Corbett D, Sivenius J et al. Transplantation of human embryonic stem cell-derived neural precursor cells and enriched environment after cortical stroke in rats: cell survival and functional recovery. The European journal of neuroscience. 2009;29(3):562–74. doi: 10.1111/j.1460-9568.2008.06599.x. [DOI] [PubMed] [Google Scholar]
  • 16.Buhnemann C, Scholz A, Bernreuther C, Malik CY, Braun H, Schachner M et al. Neuronal differentiation of transplanted embryonic stem cell-derived precursors in stroke lesions of adult rats. Brain : a journal of neurology. 2006;129(Pt 12):3238–48. doi: 10.1093/brain/awl261. [DOI] [PubMed] [Google Scholar]
  • 17.Daadi MM, Maag AL, Steinberg GK. Adherent self-renewable human embryonic stem cell-derived neural stem cell line: functional engraftment in experimental stroke model. PloS one. 2008;3(2):e1644. doi: 10.1371/journal.pone.0001644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Hayashi J, Takagi Y, Fukuda H, Imazato T, Nishimura M, Fujimoto M et al. Primate embryonic stem cell-derived neuronal progenitors transplanted into ischemic brain. Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism. 2006;26(7):906–14. doi: 10.1038/sj.jcbfm.9600247. [DOI] [PubMed] [Google Scholar]
  • 19.Gage FH. Neurogenesis in the adult brain. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2002;22(3):612–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Doetsch F, Caille I, Lim DA, Garcia-Verdugo JM, Alvarez-Buylla A. Subventricular zone astrocytes are neural stem cells in the adult mammalian brain. Cell. 1999;97(6):703–16. [DOI] [PubMed] [Google Scholar]
  • 21.Liu YP, Lang BT, Baskaya MK, Dempsey RJ, Vemuganti R. The potential of neural stem cells to repair stroke-induced brain damage. Acta neuropathologica. 2009;117(5):469–80. doi: 10.1007/s00401-009-0516-1. [DOI] [PubMed] [Google Scholar]
  • 22.Noisa P, Urrutikoetxea-Uriguen A, Li M, Cui W. Generation of human embryonic stem cell reporter lines expressing GFP specifically in neural progenitors. Stem Cell Rev. 2010;6(3):438–49. doi: 10.1007/s12015-010-9159-9. [DOI] [PubMed] [Google Scholar]
  • 23.Bond AM, Ming GL, Song H. Adult Mammalian Neural Stem Cells and Neurogenesis: Five Decades Later. Cell stem cell. 2015;17(4):385–95. doi: 10.1016/j.stem.2015.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Larimer P, Spatazza J, Espinosa JS, Tang Y, Kaneko M, Hasenstaub AR et al. Caudal Ganglionic Eminence Precursor Transplants Disperse and Integrate as Lineage-Specific Interneurons but Do Not Induce Cortical Plasticity. Cell reports. 2016;16(5):1391–404. doi: 10.1016/j.celrep.2016.06.071.●●The authors demonstrate that not all neural precursor cells have equal capacity to induce neuroplasticity. Following transplantation, precursor cells from both the medial and caudal ganglionic emincences disperse throughout the brain and laminate appropriately as interneurons. The ability to induce ocular dominance plasticity is restricted to genetically specified “medial” precurors even if isolated anatomically from the caudal ganglionic eminence.
  • 25.Chen L, Zhang G, Gu Y, Guo X. Meta-Analysis and Systematic Review of Neural Stem Cells therapy for experimental ischemia stroke in preclinical studies. Sci Rep. 2016;6:32291. doi: 10.1038/srep32291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Huang H, Qian K, Han X, Li X, Zheng Y, Chen Z et al. Intraparenchymal Neural Stem/Progenitor Cell Transplantation for Ischemic Stroke Animals: A Meta-Analysis and Systematic Review. Stem Cells Int. 2018;2018:4826407. doi: 10.1155/2018/4826407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Kern S, Eichler H, Stoeve J, Kluter H, Bieback K. Comparative analysis of mesenchymal stem cells from bone marrow, umbilical cord blood, or adipose tissue. Stem cells. 2006;24(5):1294–301. doi: 10.1634/stemcells.2005-0342. [DOI] [PubMed] [Google Scholar]
  • 28.Banerjee S, Williamson DA, Habib N, Chataway J. The potential benefit of stem cell therapy after stroke: an update. Vasc Health Risk Manag. 2012;8:569–80. doi: 10.2147/VHRM.S25745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Greco SJ, Zhou C, Ye JH, Rameshwar P. An interdisciplinary approach and characterization of neuronal cells transdifferentiated from human mesenchymal stem cells. Stem Cells Dev. 2007;16(5):811–26. doi: 10.1089/scd.2007.0011. [DOI] [PubMed] [Google Scholar]
  • 30.Sanchez-Ramos J, Song S, Cardozo-Pelaez F, Hazzi C, Stedeford T, Willing A et al. Adult bone marrow stromal cells differentiate into neural cells in vitro. Experimental neurology. 2000;164(2):247–56. doi: 10.1006/exnr.2000.7389. [DOI] [PubMed] [Google Scholar]
  • 31.Dezawa M. Muse Cells Provide the Pluripotency of Mesenchymal Stem Cells: Direct Contribution of Muse Cells to Tissue Regeneration. Cell Transplant. 2016;25(5):849–61. doi: 10.3727/096368916X690881. [DOI] [PubMed] [Google Scholar]
  • 32.Kuroda Y, Kitada M, Wakao S, Nishikawa K, Tanimura Y, Makinoshima H et al. Unique multipotent cells in adult human mesenchymal cell populations. Proceedings of the National Academy of Sciences of the United States of America. 2010;107(19):8639–43. doi: 10.1073/pnas.0911647107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Yamauchi T, Kuroda Y, Morita T, Shichinohe H, Houkin K, Dezawa M et al. Therapeutic effects of human multilineage-differentiating stress enduring (MUSE) cell transplantation into infarct brain of mice. PloS one. 2015;10(3):e0116009. doi: 10.1371/journal.pone.0116009.●This study demonstrates possible independent effects and mechanisms for different subpopulations of bone marrow derived progenitors in stroke recovery.
  • 34.Aggarwal S, Pittenger MF. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood. 2005;105(4):1815–22. doi: 10.1182/blood-2004-04-1559. [DOI] [PubMed] [Google Scholar]
  • 35.Hess DC, Hill WD. Cell therapy for ischaemic stroke. Cell Prolif. 2011;44 Suppl 1:1–8. doi: 10.1111/j.1365-2184.2010.00718.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Rosado-de-Castro PH, Schmidt Fda R, Battistella V, Lopes de Souza SA, Gutfilen B, Goldenberg RC et al. Biodistribution of bone marrow mononuclear cells after intra-arterial or intravenous transplantation in subacute stroke patients. Regen Med. 2013;8(2):145–55. doi: 10.2217/rme.13.2. [DOI] [PubMed] [Google Scholar]
  • 37.Jiang J, Wang Y, Liu B, Chen X, Zhang S. Challenges and research progress of the use of mesenchymal stem cells in the treatment of ischemic stroke. Brain & development. 2018. doi: 10.1016/j.braindev.2018.03.015. [DOI] [PubMed] [Google Scholar]
  • 38.Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J, Frane JL, Tian S et al. Induced pluripotent stem cell lines derived from human somatic cells. Science. 2007;318(5858):1917–20. doi: 10.1126/science.1151526. [DOI] [PubMed] [Google Scholar]
  • 39.Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131(5):861–72. doi: 10.1016/j.cell.2007.11.019. [DOI] [PubMed] [Google Scholar]
  • 40.Polo JM, Liu S, Figueroa ME, Kulalert W, Eminli S, Tan KY et al. Cell type of origin influences the molecular and functional properties of mouse induced pluripotent stem cells. Nature biotechnology. 2010;28(8):848–55. doi: 10.1038/nbt.1667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Han X, Yu L, Ren J, Wang M, Liu Z, Hu X et al. Efficient and Fast Differentiation of Human Neural Stem Cells from Human Embryonic Stem Cells for Cell Therapy. Stem Cells Int. 2017;2017:9405204. doi: 10.1155/2017/9405204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Denham M, Dottori M. Neural differentiation of induced pluripotent stem cells. Methods in molecular biology. 2011;793:99–110. doi: 10.1007/978-1-61779-328-8_7. [DOI] [PubMed] [Google Scholar]
  • 43.Marei HE, Hasan A, Rizzi R, Althani A, Afifi N, Cenciarelli C et al. Potential of Stem Cell-Based Therapy for Ischemic Stroke. Front Neurol. 2018;9:34. doi: 10.3389/fneur.2018.00034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Kokaia Z, Llorente IL, Carmichael ST. Customized Brain Cells for Stroke Patients Using Pluripotent Stem Cells. Stroke; a journal of cerebral circulation. 2018;49(5):1091–8. doi: 10.1161/STROKEAHA.117.018291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Kokaia Z, Tornero D, Lindvall O. Transplantation of reprogrammed neurons for improved recovery after stroke. Prog Brain Res. 2017;231:245–63. doi: 10.1016/bs.pbr.2016.11.013.●The authors provide a comprehensive review of iPSC-derived precursors for stroke recovery,
  • 46.Chau MJ, Deveau TC, Song M, Gu X, Chen D, Wei L. iPSC Transplantation increases regeneration and functional recovery after ischemic stroke in neonatal rats. Stem cells. 2014;32(12):3075–87. doi: 10.1002/stem.1802. [DOI] [PubMed] [Google Scholar]
  • 47.Oki K, Tatarishvili J, Wood J, Koch P, Wattananit S, Mine Y et al. Human-induced pluripotent stem cells form functional neurons and improve recovery after grafting in stroke-damaged brain. Stem cells. 2012;30(6):1120–33. doi: 10.1002/stem.1104. [DOI] [PubMed] [Google Scholar]
  • 48.Tornero D, Wattananit S, Gronning Madsen M, Koch P, Wood J, Tatarishvili J et al. Human induced pluripotent stem cell-derived cortical neurons integrate in stroke-injured cortex and improve functional recovery. Brain : a journal of neurology. 2013;136(Pt 12):3561–77. doi: 10.1093/brain/awt278. [DOI] [PubMed] [Google Scholar]
  • 49.Tornero D, Tsupykov O, Granmo M, Rodriguez C, Gronning-Hansen M, Thelin J et al. Synaptic inputs from stroke-injured brain to grafted human stem cell-derived neurons activated by sensory stimuli. Brain : a journal of neurology. 2017;140(3):692–706. doi: 10.1093/brain/aww347.●●The authors demonstrate that iPSC-derived cortically primed neurons engraft and receive direct synaptic inputs from host thalamic neurons. This occurs in a somatotopic fashion, and transplanted neurons can exhibit activation from physiological stimuli.
  • 50.Altman J. Are new neurons formed in the brains of adult mammals? Science. 1962;135(3509):1127–8. [DOI] [PubMed] [Google Scholar]
  • 51.Kaplan MS, Hinds JW. Neurogenesis in the adult rat: electron microscopic analysis of light radioautographs. Science. 1977;197(4308):1092–4. [DOI] [PubMed] [Google Scholar]
  • 52.Ihrie RA, Alvarez-Buylla A. Lake-front property: a unique germinal niche by the lateral ventricles of the adult brain. Neuron. 2011;70(4):674–86. doi: 10.1016/j.neuron.2011.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Bonaguidi MA, Song J, Ming GL, Song H. A unifying hypothesis on mammalian neural stem cell properties in the adult hippocampus. Current opinion in neurobiology. 2012;22(5):754–61. doi: 10.1016/j.conb.2012.03.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Rakic P. Limits of neurogenesis in primates. Science. 1985;227(4690):1054–6. [DOI] [PubMed] [Google Scholar]
  • 55.Eriksson PS, Perfilieva E, Bjork-Eriksson T, Alborn AM, Nordborg C, Peterson DA et al. Neurogenesis in the adult human hippocampus. Nature medicine. 1998;4(11):1313–7. doi: 10.1038/3305. [DOI] [PubMed] [Google Scholar]
  • 56.Bhardwaj RD, Curtis MA, Spalding KL, Buchholz BA, Fink D, Bjork-Eriksson T et al. Neocortical neurogenesis in humans is restricted to development. Proceedings of the National Academy of Sciences of the United States of America. 2006;103(33):12564–8. doi: 10.1073/pnas.0605177103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Paredes MF, James D, Gil-Perotin S, Kim H, Cotter JA, Ng C et al. Extensive migration of young neurons into the infant human frontal lobe. Science. 2016;354(6308). doi: 10.1126/science.aaf7073.●This study demonstrates a previously unappreciated persistence of neurogenesis and migration in the postnatal human brain, albeit largely limited to infants.
  • 58.Felling RJ, Snyder MJ, Romanko MJ, Rothstein RP, Ziegler AN, Yang Z et al. Neural stem/progenitor cells participate in the regenerative response to perinatal hypoxia/ischemia. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2006;26(16):4359–69. doi: 10.1523/JNEUROSCI.1898-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Arvidsson A, Collin T, Kirik D, Kokaia Z, Lindvall O. Neuronal replacement from endogenous precursors in the adult brain after stroke. Nature medicine. 2002;8(9):963–70. doi: 10.1038/nm747. [DOI] [PubMed] [Google Scholar]
  • 60.Plane JM, Liu R, Wang TW, Silverstein FS, Parent JM. Neonatal hypoxic-ischemic injury increases forebrain subventricular zone neurogenesis in the mouse. Neurobiology of disease. 2004;16(3):585–95. doi: 10.1016/j.nbd.2004.04.003. [DOI] [PubMed] [Google Scholar]
  • 61.Parent JM. The role of seizure-induced neurogenesis in epileptogenesis and brain repair. Epilepsy Res. 2002;50(1-2):179–89. [DOI] [PubMed] [Google Scholar]
  • 62.Lindvall O, Kokaia Z. Neurogenesis following Stroke Affecting the Adult Brain. Cold Spring Harb Perspect Biol. 2015;7(11). doi: 10.1101/cshperspect.a019034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Felling RJ, Levison SW. Enhanced neurogenesis following stroke. Journal of neuroscience research. 2003;73(3):277–83. doi: 10.1002/jnr.10670. [DOI] [PubMed] [Google Scholar]
  • 64.Lagace DC. Does the endogenous neurogenic response alter behavioral recovery following stroke? Behav Brain Res. 2012;227(2):426–32. doi: 10.1016/j.bbr.2011.08.045. [DOI] [PubMed] [Google Scholar]
  • 65.Jin K, Wang X, Xie L, Mao XO, Zhu W, Wang Y et al. Evidence for stroke-induced neurogenesis in the human brain. Proceedings of the National Academy of Sciences of the United States of America. 2006;103(35):13198–202. doi: 10.1073/pnas.0603512103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Marti-Fabregas J, Romaguera-Ros M, Gomez-Pinedo U, Martinez-Ramirez S, Jimenez-Xarrie E, Marin R et al. Proliferation in the human ipsilateral subventricular zone after ischemic stroke. Neurology. 2010;74(5):357–65. doi: 10.1212/WNL.0b013e3181cbccec. [DOI] [PubMed] [Google Scholar]
  • 67.Lam J, Lowry WE, Carmichael ST, Segura T. Delivery of iPS-NPCs to the Stroke Cavity within a Hyaluronic Acid Matrix Promotes the Differentiation of Transplanted Cells. Adv Funct Mater. 2014;24(44):7053–62. doi: 10.1002/adfm.201401483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.George PM, Oh B, Dewi R, Hua T, Cai L, Levinson A et al. Engineered stem cell mimics to enhance stroke recovery. Biomaterials. 2018;178:63–72. doi: 10.1016/j.biomaterials.2018.06.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Cook DJ, Nguyen C, Chun HN, I LL, Chiu AS, Machnicki M et al. Hydrogel-delivered brain-derived neurotrophic factor promotes tissue repair and recovery after stroke. Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism. 2017;37(3):1030–45. doi: 10.1177/0271678X16649964.●●This study demonstrates the potential for hydrogel scaffolds to promote endogenous repair mechanisms including neurogenesis.
  • 70.Moshayedi P, Nih LR, Llorente IL, Berg AR, Cinkornpumin J, Lowry WE et al. Systematic optimization of an engineered hydrogel allows for selective control of human neural stem cell survival and differentiation after transplantation in the stroke brain. Biomaterials. 2016;105:145–55. doi: 10.1016/j.biomaterials.2016.07.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Manley NC, Steinberg GK. Tracking stem cells for cellular therapy in stroke. Curr Pharm Des. 2012;18(25):3685–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Walczak P, Wojtkiewicz J, Nowakowski A, Habich A, Holak P, Xu J et al. Real-time MRI for precise and predictable intra-arterial stem cell delivery to the central nervous system. Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism. 2017;37(7):2346–58. doi: 10.1177/0271678X16665853. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Walczak P, Zhang J, Gilad AA, Kedziorek DA, Ruiz-Cabello J, Young RG et al. Dual-modality monitoring of targeted intraarterial delivery of mesenchymal stem cells after transient ischemia. Stroke; a journal of cerebral circulation. 2008;39(5):1569–74. doi: 10.1161/STROKEAHA.107.502047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Janowski M, Walczak P, Kropiwnicki T, Jurkiewicz E, Domanska-Janik K, Bulte JW et al. Long-term MRI cell tracking after intraventricular delivery in a patient with global cerebral ischemia and prospects for magnetic navigation of stem cells within the CSF. PloS one. 2014;9(2):e97631. doi: 10.1371/journal.pone.0097631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Janowski M, Wagner DC, Boltze J. Stem Cell-Based Tissue Replacement After Stroke: Factual Necessity or Notorious Fiction? Stroke; a journal of cerebral circulation. 2015;46(8):2354–63. doi: 10.1161/STROKEAHA.114.007803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Anrather J, Iadecola C. Inflammation and Stroke: An Overview. Neurotherapeutics : the journal of the American Society for Experimental NeuroTherapeutics. 2016;13(4):661–70. doi: 10.1007/s13311-016-0483-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Ben-Hur T, Ben-Menachem O, Furer V, Einstein O, Mizrachi-Kol R, Grigoriadis N. Effects of proinflammatory cytokines on the growth, fate, and motility of multipotential neural precursor cells. Molecular and cellular neurosciences. 2003;24(3):623–31. [DOI] [PubMed] [Google Scholar]
  • 78.Erlandsson A, Lin CH, Yu F, Morshead CM. Immunosuppression promotes endogenous neural stem and progenitor cell migration and tissue regeneration after ischemic injury. Experimental neurology. 2011;230(1):48–57. doi: 10.1016/j.expneurol.2010.05.018. [DOI] [PubMed] [Google Scholar]
  • 79.Ekdahl CT, Kokaia Z, Lindvall O. Brain inflammation and adult neurogenesis: the dual role of microglia. Neuroscience. 2009;158(3):1021–9. doi: 10.1016/j.neuroscience.2008.06.052. [DOI] [PubMed] [Google Scholar]
  • 80.Thored P, Heldmann U, Gomes-Leal W, Gisler R, Darsalia V, Taneera J et al. Long-term accumulation of microglia with proneurogenic phenotype concomitant with persistent neurogenesis in adult subventricular zone after stroke. Glia. 2009;57(8):835–49. doi: 10.1002/glia.20810. [DOI] [PubMed] [Google Scholar]
  • 81.Hu X, Leak RK, Shi Y, Suenaga J, Gao Y, Zheng P et al. Microglial and macrophage polarization-new prospects for brain repair. Nature reviews Neurology. 2015;11(1):56–64. doi: 10.1038/nrneurol.2014.207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Ma Y, Wang J, Wang Y, Yang GY. The biphasic function of microglia in ischemic stroke. Progress in neurobiology. 2017;157:247–72. doi: 10.1016/j.pneurobio.2016.01.005.●The authors nicely review the dual functions of inflammation, in particularly microglia, in mediating both stroke-related injury and repair.
  • 83.Ohtaki H, Ylostalo JH, Foraker JE, Robinson AP, Reger RL, Shioda S et al. Stem/progenitor cells from bone marrow decrease neuronal death in global ischemia by modulation of inflammatory/immune responses. Proceedings of the National Academy of Sciences of the United States of America. 2008;105(38):14638–43. doi: 10.1073/pnas.0803670105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Boshuizen MCS, Steinberg GK. Stem Cell-Based Immunomodulation After Stroke: Effects on Brain Repair Processes. Stroke; a journal of cerebral circulation. 2018;49(6):1563–70. doi: 10.1161/STROKEAHA.117.020465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Hess DC, Wechsler LR, Clark WM, Savitz SI, Ford GA, Chiu D et al. Safety and efficacy of multipotent adult progenitor cells in acute ischaemic stroke (MASTERS): a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Neurol. 2017;16(5):360–8. doi: 10.1016/S1474-4422(17)30046-7.●●This is one of the more extensive and rigorously designed trials of cell-based therapy. While efficacy outcomes were not significantly positive, the study demonstrates the feasibility of cell-based therapy in a very acute window utilizing an “off the shelf” cell product.
  • 86.Yang B, Hamilton JA, Valenzuela KS, Bogaerts A, Xi X, Aronowski J et al. Multipotent Adult Progenitor Cells Enhance Recovery After Stroke by Modulating the Immune Response from the Spleen. Stem cells. 2017;35(5):1290–302. doi: 10.1002/stem.2600. [DOI] [PubMed] [Google Scholar]
  • 87.Yang B, Li W, Satani N, Nghiem DM, Xi X, Aronowski J et al. Protective Effects of Autologous Bone Marrow Mononuclear Cells After Administering t-PA in an Embolic Stroke Model. Translational stroke research. 2018;9(2):135–45. doi: 10.1007/s12975-017-0563-1. [DOI] [PubMed] [Google Scholar]
  • 88.Stonesifer C, Corey S, Ghanekar S, Diamandis Z, Acosta SA, Borlongan CV. Stem cell therapy for abrogating stroke-induced neuroinflammation and relevant secondary cell death mechanisms. Progress in neurobiology. 2017;158:94–131. doi: 10.1016/j.pneurobio.2017.07.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Murphy TH, Corbett D. Plasticity during stroke recovery: from synapse to behaviour. Nature reviews Neuroscience. 2009;10(12):861–72. doi: 10.1038/nrn2735. [DOI] [PubMed] [Google Scholar]
  • 90.Carmichael ST, Chesselet MF. Synchronous neuronal activity is a signal for axonal sprouting after cortical lesions in the adult. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2002;22(14):6062–70. doi:20026605. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Dancause N, Barbay S, Frost SB, Plautz EJ, Chen D, Zoubina EV et al. Extensive cortical rewiring after brain injury. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2005;25(44):10167–79. doi: 10.1523/JNEUROSCI.3256-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Brown CE, Aminoltejari K, Erb H, Winship IR, Murphy TH. In vivo voltage-sensitive dye imaging in adult mice reveals that somatosensory maps lost to stroke are replaced over weeks by new structural and functional circuits with prolonged modes of activation within both the peri-infarct zone and distant sites. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2009;29(6):1719–34. doi: 10.1523/JNEUROSCI.4249-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Jones TA, Schallert T. Use-dependent growth of pyramidal neurons after neocortical damage. The Journal of neuroscience : the official journal of the Society for Neuroscience. 1994;14(4):2140–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Biernaskie J, Corbett D. Enriched rehabilitative training promotes improved forelimb motor function and enhanced dendritic growth after focal ischemic injury. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2001;21(14):5272–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Horita Y, Honmou O, Harada K, Houkin K, Hamada H, Kocsis JD. Intravenous 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 rat. Journal of neuroscience research. 2006;84(7):1495–504. doi: 10.1002/jnr.21056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Toyama K, Honmou O, Harada K, Suzuki J, Houkin K, Hamada H et al. Therapeutic benefits of angiogenetic gene-modified human mesenchymal stem cells after cerebral ischemia. Experimental neurology. 2009;216(1):47–55. doi: 10.1016/j.expneurol.2008.11.010. [DOI] [PubMed] [Google Scholar]
  • 97.Liu H, Honmou O, Harada K, Nakamura K, Houkin K, Hamada H et al. Neuroprotection by PlGF gene-modified human mesenchymal stem cells after cerebral ischaemia. Brain : a journal of neurology. 2006;129(Pt 10):2734–45. doi: 10.1093/brain/awl207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Nomura T, Honmou O, Harada K, Houkin K, Hamada H, Kocsis JD. I.V. infusion of brain-derived neurotrophic factor gene-modified human mesenchymal stem cells protects against injury in a cerebral ischemia model in adult rat. Neuroscience. 2005;136(1):161–9. doi: 10.1016/j.neuroscience.2005.06.062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Teixeira FG, Carvalho MM, Sousa N, Salgado AJ. Mesenchymal stem cells secretome: a new paradigm for central nervous system regeneration? Cellular and molecular life sciences : CMLS. 2013;70(20):3871–82. doi: 10.1007/s00018-013-1290-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Park H, Poo MM. Neurotrophin regulation of neural circuit development and function. Nature reviews Neuroscience. 2013;14(1):7–23. doi: 10.1038/nrn3379. [DOI] [PubMed] [Google Scholar]
  • 101.Cramer SC, Chopp M. Recovery recapitulates ontogeny. Trends in neurosciences. 2000;23(6):265–71. [DOI] [PubMed] [Google Scholar]
  • 102.Zhang ZG, Chopp M. Exosomes in stroke pathogenesis and therapy. The Journal of clinical investigation. 2016;126(4):1190–7. doi: 10.1172/JCI81133.●Excellent review about a exosomes, and emerging field related to stem cells but beyond the scope of this current review.
  • 103.Hsieh JY, Wang HW, Chang SJ, Liao KH, Lee IH, Lin WS et al. Mesenchymal stem cells from human umbilical cord express preferentially secreted factors related to neuroprotection, neurogenesis, and angiogenesis. PloS one. 2013;8(8):e72604. doi: 10.1371/journal.pone.0072604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Sheikh AM, Yano S, Mitaki S, Haque MA, Yamaguchi S, Nagai A. A Mesenchymal stem cell line (B10) increases angiogenesis in a rat MCAO model. Experimental neurology. 2018;311:182–93. doi: 10.1016/j.expneurol.2018.10.001. [DOI] [PubMed] [Google Scholar]
  • 105.Jin K, Xie L, Mao X, Greenberg MB, Moore A, Peng B et al. Effect of human neural precursor cell transplantation on endogenous neurogenesis after focal cerebral ischemia in the rat. Brain research. 2011;1374:56–62. doi: 10.1016/j.brainres.2010.12.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Andres RH, Horie N, Slikker W, Keren-Gill H, Zhan K, Sun G et al. Human neural stem cells enhance structural plasticity and axonal transport in the ischaemic brain. Brain : a journal of neurology. 2011;134(Pt 6):1777–89. doi: 10.1093/brain/awr094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Hou SW, Wang YQ, Xu M, Shen DH, Wang JJ, Huang F et al. Functional integration of newly generated neurons into striatum after cerebral ischemia in the adult rat brain. Stroke; a journal of cerebral circulation. 2008;39(10):2837–44. doi: 10.1161/STROKEAHA.107.510982. [DOI] [PubMed] [Google Scholar]
  • 108.Daadi MM, Lee SH, Arac A, Grueter BA, Bhatnagar R, Maag AL et al. Functional engraftment of the medial ganglionic eminence cells in experimental stroke model. Cell Transplant. 2009;18(7):815–26. doi: 10.3727/096368909X470829. [DOI] [PubMed] [Google Scholar]
  • 109.Martinez-Cerdeno V, Noctor SC, Espinosa A, Ariza J, Parker P, Orasji S et al. Embryonic MGE precursor cells grafted into adult rat striatum integrate and ameliorate motor symptoms in 6-OHDA-lesioned rats. Cell stem cell. 2010;6(3):238–50. doi: 10.1016/j.stem.2010.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Zhang R, Chopp M, Zhang ZG. Oligodendrogenesis after cerebral ischemia. Frontiers in cellular neuroscience. 2013;7:201. doi: 10.3389/fncel.2013.00201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Adams KL, Gallo V. The diversity and disparity of the glial scar. Nature neuroscience. 2018;21(1):9–15. doi: 10.1038/s41593-017-0033-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Li Y, Liu Z, Xin H, Chopp M. The role of astrocytes in mediating exogenous cell-based restorative therapy for stroke. Glia. 2014;62(1):1–16. doi: 10.1002/glia.22585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Kondziolka D, Steinberg GK, Wechsler L, Meltzer CC, Elder E, Gebel J et al. Neurotransplantation for patients with subcortical motor stroke: a phase 2 randomized trial. Journal of neurosurgery. 2005;103(1):38–45. doi: 10.3171/jns.2005.103.1.0038. [DOI] [PubMed] [Google Scholar]
  • 114.Kondziolka D, Wechsler L, Goldstein S, Meltzer C, Thulborn KR, Gebel J et al. Transplantation of cultured human neuronal cells for patients with stroke. Neurology. 2000;55(4):565–9. [DOI] [PubMed] [Google Scholar]
  • 115.Savitz SI, Dinsmore J, Wu J, Henderson GV, Stieg P, Caplan LR. Neurotransplantation of fetal porcine cells in patients with basal ganglia infarcts: a preliminary safety and feasibility study. Cerebrovasc Dis. 2005;20(2):101–7. doi: 10.1159/000086518. [DOI] [PubMed] [Google Scholar]
  • 116.Thomas RJ, Hope AD, Hourd P, Baradez M, Miljan EA, Sinden JD et al. Automated, serum-free production of CTX0E03: a therapeutic clinical grade human neural stem cell line. Biotechnol Lett. 2009;31(8):1167–72. doi: 10.1007/s10529-009-9989-1. [DOI] [PubMed] [Google Scholar]
  • 117.Stevanato L, Corteling RL, Stroemer P, Hope A, Heward J, Miljan EA et al. c-MycERTAM transgene silencing in a genetically modified human neural stem cell line implanted into MCAo rodent brain. BMC neuroscience. 2009;10:86. doi: 10.1186/1471-2202-10-86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Kalladka D, Sinden J, Pollock K, Haig C, McLean J, Smith W et al. Human neural stem cells in patients with chronic ischaemic stroke (PISCES): a phase 1, first-in-man study. Lancet. 2016;388(10046):787–96. doi: 10.1016/S0140-6736(16)30513-X. [DOI] [PubMed] [Google Scholar]
  • 119.Steinberg GK, Kondziolka D, Wechsler LR, Lunsford LD, Coburn ML, Billigen JB et al. Clinical Outcomes of Transplanted Modified Bone Marrow-Derived Mesenchymal Stem Cells in Stroke: A Phase 1/2a Study. Stroke; a journal of cerebral circulation. 2016;47(7):1817–24. doi: 10.1161/STROKEAHA.116.012995.●●While interpretation is limited by the design as an uncontrolled study, this trial demonstrated clinically significant improvements in chronic stroke patients following stem cell transplantation.
  • 120.Honmou O, Houkin K, Matsunaga T, Niitsu Y, Ishiai S, Onodera R et al. Intravenous administration of auto serum-expanded autologous mesenchymal stem cells in stroke. Brain : a journal of neurology. 2011;134(Pt 6):1790–807. doi: 10.1093/brain/awr063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Gaudinski MR, Henning EC, Miracle A, Luby M, Warach S, Latour LL. Establishing final infarct volume: stroke lesion evolution past 30 days is insignificant. Stroke; a journal of cerebral circulation. 2008;39(10):2765–8. doi: 10.1161/STROKEAHA.107.512269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Savitz SI, Chopp M, Deans R, Carmichael T, Phinney D, Wechsler L et al. Stem Cell Therapy as an Emerging Paradigm for Stroke (STEPS) II. Stroke; a journal of cerebral circulation. 2011;42(3):825–9. doi: 10.1161/STROKEAHA.110.601914. [DOI] [PubMed] [Google Scholar]
  • 123.Savitz SI, Cramer SC, Wechsler L, Consortium S. Stem cells as an emerging paradigm in stroke 3: enhancing the development of clinical trials. Stroke; a journal of cerebral circulation. 2014;45(2):634–9. doi: 10.1161/STROKEAHA.113.003379.●●Consensus guidelines from the STEPS consortiums outlining best practices moving forward in the design of clinical trials for cell-based therapies. These guidelines are essential to ensure the quality of trials and the continued generation of mechanistic knowledge in the trial setting so that therapies can be refined and optimized based on clinical results.
  • 124.Stem Cell Therapies as an Emerging Paradigm in Stroke P. Stem Cell Therapies as an Emerging Paradigm in Stroke (STEPS): bridging basic and clinical science for cellular and neurogenic factor therapy in treating stroke. Stroke; a journal of cerebral circulation. 2009;40(2):510–5. doi: 10.1161/STROKEAHA.108.526863. [DOI] [PubMed] [Google Scholar]
  • 125.Bernstock JD, Peruzzotti-Jametti L, Ye D, Gessler FA, Maric D, Vicario N et al. Neural stem cell transplantation in ischemic stroke: A role for preconditioning and cellular engineering. Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism. 2017;37(7):2314–9. doi: 10.1177/0271678X17700432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Chan SJ, Love C, Spector M, Cool SM, Nurcombe V, Lo EH. Endogenous regeneration: Engineering growth factors for stroke. Neurochemistry international. 2017. doi: 10.1016/j.neuint.2017.03.024. [DOI] [PubMed] [Google Scholar]
  • 127.Madl CM, Heilshorn SC, Blau HM. Bioengineering strategies to accelerate stem cell therapeutics. Nature. 2018;557(7705):335–42. doi: 10.1038/s41586-018-0089-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Krakauer JW, Marshall RS. The proportional recovery rule for stroke revisited. Annals of neurology. 2015;78(6):845–7. doi: 10.1002/ana.24537.●●Summarizes the concept of proportional recovery and discusses two studies which predict who may or may not follow this rule, providing a benchmark for expected spontaneous recovery against which stem cell therapies may be compared.
  • 129.Rabinovich SS, Seledtsov VI, Banul NV, Poveshchenko OV, Senyukov VV, Astrakov SV et al. Cell therapy of brain stroke. Bull Exp Biol Med. 2005;139(1):126–8. [DOI] [PubMed] [Google Scholar]
  • 130.Bang OY, Lee JS, Lee PH, Lee G. Autologous mesenchymal stem cell transplantation in stroke patients. Annals of neurology. 2005;57(6):874–82. doi: 10.1002/ana.20501. [DOI] [PubMed] [Google Scholar]
  • 131.Suarez-Monteagudo C, Hernandez-Ramirez P, Alvarez-Gonzalez L, Garcia-Maeso I, de la Cuetara-Bernal K, Castillo-Diaz L et al. Autologous bone marrow stem cell neurotransplantation in stroke patients. An open study. Restorative neurology and neuroscience. 2009;27(3):151–61. doi: 10.3233/RNN-2009-0483. [DOI] [PubMed] [Google Scholar]
  • 132.Lee JS, Hong JM, Moon GJ, Lee PH, Ahn YH, Bang OY et al. A long-term follow-up study of intravenous autologous mesenchymal stem cell transplantation in patients with ischemic stroke. Stem cells. 2010;28(6):1099–106. doi: 10.1002/stem.430. [DOI] [PubMed] [Google Scholar]
  • 133.Savitz SI, Misra V, Kasam M, Juneja H, Cox CS Jr., Alderman S et al. Intravenous autologous bone marrow mononuclear cells for ischemic stroke. Annals of neurology. 2011;70(1):59–69. doi: 10.1002/ana.22458. [DOI] [PubMed] [Google Scholar]
  • 134.Friedrich MA, Martins MP, Araujo MD, Klamt C, Vedolin L, Garicochea B et al. Intra-arterial infusion of autologous bone marrow mononuclear cells in patients with moderate to severe middle cerebral artery acute ischemic stroke. Cell Transplant. 2012;21 Suppl 1:S13–21. doi: 10.3727/096368912X612512 . [DOI] [PubMed] [Google Scholar]
  • 135.Moniche F, Gonzalez A, Gonzalez-Marcos JR, Carmona M, Pinero P, Espigado I et al. Intra-arterial bone marrow mononuclear cells in ischemic stroke: a pilot clinical trial. Stroke; a journal of cerebral circulation. 2012;43(8):2242–4. doi: 10.1161/STROKEAHA.112.659409. [DOI] [PubMed] [Google Scholar]
  • 136.Bhasin A, Srivastava MV, Kumaran SS, Mohanty S, Bhatia R, Bose S et al. Autologous mesenchymal stem cells in chronic stroke. Cerebrovasc Dis Extra. 2011;1(1):93–104. doi: 10.1159/000333381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Bhasin A, Srivastava M, Bhatia R, Mohanty S, Kumaran S, Bose S. Autologous intravenous mononuclear stem cell therapy in chronic ischemic stroke. J Stem Cells Regen Med. 2012;8(3):181–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Bhasin A, Srivastava MV, Mohanty S, Bhatia R, Kumaran SS, Bose S. Stem cell therapy: a clinical trial of stroke. Clin Neurol Neurosurg. 2013;115(7):1003–8. doi: 10.1016/j.clineuro.2012.10.015. [DOI] [PubMed] [Google Scholar]
  • 139.Bhasin A, Kumaran SS, Bhatia R, Mohanty S, Srivastava MVP. Safety and Feasibility of Autologous Mesenchymal Stem Cell Transplantation in Chronic Stroke in Indian patients. A four-year follow up. J Stem Cells Regen Med. 2017;13(1):14–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Wang L, Ji H, Li M, Zhou J, Bai W, Zhong Z et al. Intrathecal Administration of Autologous CD34 Positive Cells in Patients with Past Cerebral Infarction: A Safety Study. ISRN neurology. 2013;2013:128591. doi: 10.1155/2013/128591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Prasad K, Sharma A, Garg A, Mohanty S, Bhatnagar S, Johri S et al. Intravenous autologous bone marrow mononuclear stem cell therapy for ischemic stroke: a multicentric, randomized trial. Stroke; a journal of cerebral circulation. 2014;45(12):3618–24. doi: 10.1161/STROKEAHA.114.007028. [DOI] [PubMed] [Google Scholar]
  • 142.Atkins D, Best D, Briss PA, Eccles M, Falck-Ytter Y, Flottorp S et al. Grading quality of evidence and strength of recommendations. Bmj. 2004;328(7454):1490. doi: 10.1136/bmj.328.7454.1490. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES