Abstract
Purpose of review
The central nervous system has limited capacity for regeneration after acute and chronic injury. An attractive approach to stimulate neural plasticity in the brain is to transplant stem cells in order to restore function. Here we discuss potential mechanisms of action, current knowledge and future perspectives of clinical stem cell research for stroke and traumatic brain injury.
Recent findings
Preclinical data using various models suggest stem cell therapy to be a promising therapeutic avenue. Progress has been made in elucidating the mechanism of action of various cell types used, shifting the hypothesis from neural replacement to enhancing endogenous repair processes. Translation of these findings in clinical trials is currently being pursued with emphasis on both safety as well as efficacy.
Summary
Clinical trials are currently recruiting patients in phase I and II trials to gain more insight in the therapeutic potential of stem cells in acute cerebral injury. A close interplay between results of these clinical trials and more extensive basic research is essential for future trial design: Choosing the optimal transplantation strategy and selecting the right patients.
Keywords: Neural repair, recovery, stem cell therapy, stroke, traumatic brain injury
Introduction
Stroke causes 1 in 18 deaths in the US and stroke incidence is expected to increase over the next decades.[1] The only effective treatment, intravenous tissue plasminogen activator, has a narrow time window, thereby limiting its application to a small subset of patients with ischemic stroke.[2,3] Unfortunately, many patients are disabled after stroke and require ongoing medical care, so that most yearly costs occur in this chronic setting.[4] These findings have led to research focused on mechanisms of neural repair as they relate to patterns of human recovery after stroke.
Traumatic brain injury (TBI) is a major cause of mortality and morbidity, especially among young adults, and lifelong disability is common in those who survive.[5] With no effective therapy available, interest in the potential of stem cell therapy for TBI patients is emerging. Stem-cell-based approaches therefore could provide new therapeutic avenues to restore neurological function in both patients with stroke[6] and TBI[7].
Preclinical studies using human cells
A large variety of human cell types have been used in experimental stroke (reviewed in Bliss et al.[8]) and can be divided in neural stem cells (NSCs) and non-NSCs approaches.
NSCs can give rise to neurons, astrocytes or oligodendrocytes. It is unclear which would be the most favorable cell type and how NSCs would be best prepared and modified in order to differentiate into a specific phenotype. NSCs can be generated from human embryonic and fetal stem cells. Using different techniques, NSCs have improved functional recovery after transplantation in experimental stroke.[9-12] The major concern of these cells is their capacity to form tumors, although tumorigenicity is lower for fetal-versus embryonic-derived NSCs.[13] Another approach might be to use induced pluripotent stem cells (iPSCs) generated from reprogrammed somatic cells, such as fibroblasts, to generate NSCs.[14]
Expandable neuroepithelial-like stem cells generated from human iPSCs have also been shown to improve functional recovery.[15] A potential advantage of this technology is that patients’ own cells can be used, thereby reducing the risk of cell rejection; the danger of tumor formation, however, remains.
With clinical translation of NSC transplantation underway, it is important to understand the mechanism of action for improved recovery. The initial hypothesis was that NSCs would replace lost neurons and circuits. Indeed successful synaptic integration has been documented, which could contribute to behavioral improvement.[12,16,17] However, evidence for widespread afferent and efferent neuronal projections is lacking. Transplantation of cortical neurons could be a future therapeutic option to improve integration and functioning of transplanted cells.[18] Introducing bioengineered platforms at the time of transplantation could also promote replacement of lost connections and circuitry.[19] Other modes of action have been explored with documented beneficial effects on immunomodulation, neovascularization, neural plasticity and endogenous neurogenesis.[9,20-24]
Various other human non-NSCs transplantations have improved functional recovery mostly with blood, umbilical cord and bone-marrow-derived (non-hematopoietic) cells, mesenchymal stem cells[25-32] and multipotent adult progenitor cells[33]; and in fewer studies with human embryonic[34], placental[35] and amniotic-derived[36] mesenchymal stem cells. These cells are of clinical interest as they—particularly bone marrow-derived cells—are easy to obtain from autologous donors without immunogenic and tumorigenic complications. There is no convincing evidence that these bone-marrow-derived cells functionally integrate into the brain, but various other mechanisms of action that improve endogenous repair mechanisms have been described, including secretion of neurotrophic factors[28] as well as promotion of neurogenesis[26,30], angiogenesis[27,31] and axonal plasticity[25]. Transplantation of human umbilical cord blood cells by systemic delivery has led to functional improvement by enhancing endogenous repair processes[29,32] and immortalized neural cell lines have been shown to improve functional recovery[37,38]; tumorigenicity, however, remains an issue.
Cell transplantation has also been examined in animal models of TBI using similar approaches to experimental stroke. The pathophysiology of TBI is still not clearly understood, however, thus complicating the search for optimal stem cell therapies. One can hypothesize that replacement strategies should be aimed at transplanting cells that can differentiate into endothelial cells, neurons, glia and oligodendrocytes—for remyelination, especially when considering diffuse axonal injury. Human fetal- and embryonic-derived NSCs have been shown to improve motor and cognitive function by producing neurotrophic factors, increasing angiogenesis and reducing astrogliosis.[39-44] Human blood, umbilical cord, placental and bone marrow progenitor, and mesenchymal stem cell transplantation in animal models of TBI has improved behavioral outcome. The production of neurotrophic factors could underlie this functional amelioration.[40,45-49] Limited evidence of a role for immortalized neural cell lines has been shown by transplanting NT2N human neuronal cells to improve cognitive outcome.[50]
From preclinical research to translation
Translation of preclinical data to human clinical trials is confronted with many difficulties. Experimental studies that have used a wide variety of cells lack direct large scale comparisons to help determine which cells are optimal to use in humans. Use of autologous bone marrow and potentially umbilical cord cells eliminates the risk for immune rejection. When autologous bone marrow cells are harvested by simple sorting on a Ficoll column they can be used in the subacute or chronic phase (≥ 24 hours) after stroke and TBI. Allogeneic bone marrow and umbilical cord stem cells can be ready for infusion or transplantation earlier after the cerebral event as well as in the subacute and chronic phase, but risk of rejection is a concern. Immunosupression regimens may therefore be necessary, at least for some period of time. Use of NSCs might be promising, considering their potential for neural replacement, but the risk of teratoma formation must be addressed and closely monitored, and rejection of embryonic- or fetal-derived NSCs even with immunosuppression remains a possibility. Future strategies using allogeneic iPSCs will face similar hurdles and the risk of rejection can only be dealt with by using autologous iPSCs. This latter technology, however, is not available in the acute setting because the time necessary for induction and differentiation of these cells exceeds the time frame of acute stroke and TBI (hours to day).
After establishing which cell type to use, the dose, timing and route of administration must be determined. Larger cell numbers are associated with smaller infarct sizes[29,51], but the association with specific processes of neural repair is less clear and there is no established optimal cell dosage. Using higher cell concentrations and numbers will not necessarily result in increased efficacy. Optimal transplant timing depends on several factors, including preclinical results, cell type, hypothesized mechanism of action and route of administration. Most experimental data have been generated from transplantation within 24 hours after stroke onset, but cell therapy initiated up to 4 weeks after stroke[32] has been shown to significantly enhance functional recovery. A strong association between functional efficacy and transplant timing after stroke could not be determined, but there was an association between structural outcome and time of intervention.[52] Interestingly, stroke and TBI might have different optimal timing patterns. Studies in experimental stroke suggest there is greater functional recovery from mesenschymal stem cell and NSC transplantation at 24 hours compared to 7 days after stroke[53,54], with opposite results in experimental TBI[55].
The proposed mechanism of action of cells used might influence the timing of transplantation. If a neuroprotective effect of the therapeutic intervention is suspected, acute delivery (within 24 hours) may be essential. In contrast, endogenous repair mechanisms exert their greatest potential in the subacute phase after stroke (within the first month in rodents and within 3 months in humans)[56], thus this timing might be most optimal to promote endogenous functional recovery. Hyperacute administration, however, might be less beneficial due to the hostile environment during this phase.
The mode of delivery is also influenced by various factors such as type of cell used, timing, as well as safety and efficacy profiles. Intracerebral (IC) and intracerebroventricular (ICV) techniques precisely administer cells to a chosen location, but may have higher risks due to their invasive procedures. Homing of cells in the brain has been shown to be highest with IC administration compared to intra-arterial (IA) and intravenous (IV) delivery,[57] but whether this correlates with functional improvement has not been established. Systemic delivery might be safer and more feasible, particularly in the acute and subacute phase after stroke onset. One study found that IA infusion compared to systemic IV delivery increased homing around the ischemic lesion, but with no difference in therapeutic benefit[58]. An analogous difference in engrafting after IA versus IV delivery has been described in TBI[40]. Invasive intraparenchymal delivery of cells may not be required for efficiency when neural replacement is not expected—although there may still be a therapeutic advantage with the local release of cell-secreted paracrine molecules and proteins in the peri-infarct region—and systemic infusion may be the preferred mode of delivery for these transplantation strategies from a safety perspective.
Current clinical experience
The first few clinical trials in stroke patients used invasive techniques followed then by studies that mainly applied intravenous infusion delivery strategies (Table 1). IC transplantation of cultured neuronal cells derived from a teratocarcinoma cell line was safe in all 26 patients studied.[59,60] The primary endpoint was the same between the active versus control group, but several secondary functional outcome measurements showed improvement compared to controls.[60,75] After IC transplantation of porcine fetal cells in 5 patients this phase I trial was stopped due to serious adverse events in 2 patients.[61] The first experience with human cells was obtained by transplanting a cell suspension from immature nervous and hemopoietic tissues into the subarachnoid space in 10 patients. During a 6-month follow-up a potential functional benefit in contrast to a clinically comparable control group was demonstrated.[62] All other recent studies have used autologous bone-marrow-derived cells. One trial showed IC administration of bone marrow mononuclear cells to be safe.[63] The other 8 trials used either IA or IV infusion for delivery of autologous bone-marrow-derived cells (bone marrow mononuclear cells in 6; and mesenchymal stem cells in 3) and demonstrated safety and feasibility.[64-73] IA infusion, used in 3 studies, did not result in functional improvement. The single trial evaluating both mononuclear and mesenchymal stem cells could only demonstrate a functional benefit with mononuclear cells [71]. This potential was underscored by the largest phase II trial in 52 patients with this cell type [66]. Interestingly, very limited preclinical data exists on the use of human bone marrow mononuclear cells[76] although this was the most commonly used approach in published trials. Forty-six percent (6/13) of all studies included patients within the 3-month period following stroke in which endogenous repair mechanisms are known to exert their greatest potential.
Table 1.
Published pilot clinical trials in stroke and TBI
| Study Phase (Author, year) |
Cell type | Route of administration |
Time after stroke |
Sample size |
Results | |
|---|---|---|---|---|---|---|
| STROKE | ||||||
| 1 | I (Kondziolka, 2000) [59] |
Cultured neuronal cells derived from a teratocarcinomacell line |
IC | 7-55 months | 12 | Neuronal transplantation is safe and feasible |
| 2 | II (Kondziolka, 2005) [60] |
Cultured neuronal cells derived from a teratocarcinomacell line |
IC | 1-6 years | 14 + 4 controls |
Functional improvement was documented, but no difference in the primary outcome measurement |
| 3 | I (Savitz 2005) [61] |
Porcine fetal cells treated with an anti-MHC class I antibody |
IC | 1.5 – 10 years |
5 | Cortical vein occlusion and seizures; study stopped after 5 patients |
| 4 | I (Rabinovich, 2005) [62] |
Cell suspension from immature nervous and hemopoietic tissues |
ICV | 4-24 months | 10 | No serious complications; potential functional benefit |
| 5 | I (Suárez- Monteagudo, 2009) [63] |
Autologousbone marrow mononuclear cells |
IC | 1-10 years | 5 | Safe |
| 6 | I (Barbosa da Fonseca, 2010) [64] |
Autologous bone marrow mononuclear cells |
IA | 59 - 82 days | 6 | Safe and feasible |
| 7 | I/II(Bang, 2005) [65] |
Autologous human mesenchymal stem cells |
IV | < 7 days | 5 + 25 controls |
Safe and feasible profile with potential functional benefit |
| I/II(Lee, 2010) [66] |
Autologous human mesenchymal stem cells |
IV | < 7 days | 16 + 36 controls |
Safe and feasible profile with potential functional benefit |
|
| 8 | I (Battistella, 2011) [67] |
Autologous bone marrow mononuclear cells |
IA | < 90 days | 6 | Safe and feasible |
| 9 | I (Savitz, 2011) [68] |
Autologous bone marrow mononuclear cells |
IV | 24-72 hours | 10 | Safe and feasible |
| 10 | I (Honmou, 2011) [69] |
Autologous bone marrow human mesenchymal stem cells |
IV | 36–133 days | 12 | Safe and feasible |
| 11 | I/II (Bhasin, 2011) [70] |
Autologous bone marrow human mesenchymal stem cells |
IV | 7-12 months | 6 + 6 controls |
Safe and feasible |
| I/II (Bhasin, 2012) [71] |
Autologous bone marrow mononuclear and mesenchymal stem cells |
IV | 3 months – 2 years |
20 + 20 controls |
Safe and improvement of modified Barthel Index |
|
| 12 | I (Prasad, 2012) [72] |
Autologous bone marrow mononuclear cells |
IV | 7-30 days | 11 | Safe and feasible |
| 13 | I/II (Moniche, 2012) [73] |
Autologous bone marrow mononuclear cell |
IA | 5-9 days | 10 + 10 controls |
Safe and feasible; no clear functional improvement |
|
| ||||||
| TBI | ||||||
| 1 | I (Tian, 2013) [74] |
Autologous bone marrow mesenchymal stem cell |
IT | > 1 month | 97 | Safe and feasible; potential benefit of motor function |
IA: Intra-arterial; IC: Intracerebral; ICV: Intracerebroventricular; IT: Intrathecally; IV: Intravenous; TBI: Traumatic brain injury
The first clinical trial of 97 TBI patients in whom mesenchymal stem cells were intrathecally injected was recently published. Results suggest that this stem cell therapy is safe and potentially efficacious, with documented improvement in motor functions of patients with motor disorders and in consciousness of persistent vegetative state patients, compared to baseline.[74]
Future trial perspectives
There is still insufficient evidence that stem cell therapy is beneficial for acute cerebral injury. Regarding current and future clinical studies, 29 trials registered for stroke patients are: completed (4); ongoing, not recruiting (3); recruiting (17); or not yet recruiting (5) (Table 2). In TBI only 5 trials have been designed (Table 2). In these current trials most of the inclusion criteria for stroke patients require shorter therapeutic time windows compared to the published trials, although exclusive chronic patients are still being recruited in 28% (8/29). IV delivery of autologous bone-marrow-derived mononuclear cells or mesenchymal stem cells seems to be the most preferred strategy, likely because these cells are easy to obtain, simple to deliver, safe, and there is no need for immunosuppressants. An ongoing trial using IC NSC delivery has promising preliminary 6 month and 1 year outcomes[77]. Primary outcome endpoints include feasibility, safety and tolerance, along with functional improvement using the National Institutes of Health Stroke Scale Score (NIHSSS), the modified Rankin Scale (mRS) and the Barthel Index. Potentially more extensive scales are needed for functional improvement to be demonstrated [78].
Table 2.
Registered clinical trials in stroke and TBI
| ID | Study Phase |
Cell Type | Route of administration |
Time after stroke |
Primary outcome measurement |
Region | Recruitment status |
|---|---|---|---|---|---|---|---|
| STROKE | |||||||
| NCT00950521 | II | Autologous peripheral blood CD34 Stem cell |
IC | 6-60 months | NIHSS | Taiwan | Completed |
| NCT01518231 | I | Autologous hematopoiesis stem cell |
IA | <1 year | NIHSS | China | Recruiting |
| NCT00473057 | I | Autologous bone marrow cells |
IV/IA | 3-90 days | New deficits | Brazil | Completed |
| NCT01468064 | I/II | Autologous bone marrow stromal cell and endothelial progenitor cell |
IV | 5 weeks | Adverse events |
China | Recruiting |
| NCT00908856 | I | Autologous bone marrow mononuclear cell |
IV | <72 hours | Death | United States |
Not yet recruiting |
| NCT01028794 | I/IIA | Autologous bone marrow mononuclear cells |
IV | 7-10 days | NIHSS | Japan | Recruiting |
| NCT01501773 | II | Autologous bone marrow mononuclear cells |
IV | 7-30 days | Barthel index | India | Completed |
| NCT00859014 | I | Autologous bone marrow mononuclear cells |
IV | 24-72 hours |
Safety and feasibility |
United States |
Ongoing, not recruiting |
| NCT01832428 | I/II | Autologous bone marrow mononuclear cells |
IT | Unclear | Improvement in power of muscles |
India | Recruiting |
| NCT01273337 | II | Autologous bone marrow ALD-401 |
IA | 13-19 days |
Safety (various parameters) |
United States |
Recruiting |
| NCT00535197 | I/II | Autologous CD34+ subset bone marrow stem cell |
IA | < 7 days | Adverse events |
United Kingdom |
Recruiting |
| NCT00761982 | I/II | Autologous CD34+ stem cells |
IA | 5-9 days | Adverse events & new deficits |
Spain | Completed |
| NCT01716481 | III | Autologous mesenchymal stem cell |
IV | < 90 days | mRS | Republic of Korea |
Recruiting |
| NCT00875654 | II | Autologous mesenchymal stem cells |
IV | <14 days | Feasibility and tolerance |
France | Recruiting |
| NCT01714167 | I | Autologous bone marrow mesenchymalstem cell |
IC | 3-60 months |
NIHSS | China | Recruiting |
| NCT01461720 | II | Autologous mesenchymal stem cells |
IV | 1 week - 2 months |
NIHSS, mRS, Barthel index and infarct size |
Malaysia | Recruiting |
| NCT01091701 | I/II | Ex vivo cultured adult allogenic mesenchymal stem cells |
IV | < 10 days | Adverse events and NIHSS |
Malaysia | Not yet recruiting |
| NCT01436487 | II | Adult stem cell: Multistem | IV | 1-2 days | Adverse events and mRS |
United States |
Recruiting |
| NCT01849887 | I/II | Allogeneic bone marrow mesenchymal stem cells |
IV | 24-72 hours |
Adverse events |
United States |
Not yet recruiting |
| NCT01297413 | I/II | Allogeneic bone marrow adult mesenchymal stem cells |
IV | >6 months | Safety (various parameters) |
United States |
Recruiting |
| NCT01287936 | I/IIA | Modified stromal cells (SB623) |
IC | 6-60 months |
Safety (various parameters) |
United States |
Recruiting |
| NCT01453829 | I/II | Autologous adipose-derived stromal Cells |
IV/IA | Unclear | NIHSS, Adverse events, Barthel index |
Mexico | Recruiting |
| NCT01678534 | II | Allogenic mesenchymal stem cells from adipose tissue |
IV | <12 hours | Adverse events, complications and tumour development |
Spain | Not yet recruiting |
| NCT01310114 | IIA | Human placenta-derived cells, PDA001 |
IV | Subacute | Adverse events and mRS |
United States |
Ongoing, not recruiting |
| NCT01673932 | I | Umbilical cord blood mononuclear cells |
IC | 6-60 months |
NIHSS and safety (various parameters) |
China | Recruiting |
| NCT01438593 | I | Umbilical Cord Blood CD34+ Stem Cell |
IV | 6-60 months |
NIHSS | Taiwan | Not yet recruiting |
| NCT01700166 | I | Autologous human cord blood derived stem cell |
IV | Children only: 6 weeks - 6 years |
Functional, physiological and anatomic outcome |
United states |
Recruiting |
| NCT01151124 | I | CTX0E03 neural stem cells | IC | 6-months - 5 years |
Adverse events |
United Kingdom |
Ongoing, not recruiting |
| NCT01327768 | I | Olfactory ensheathing cells | IC | 6-60 months |
NIHSS | Taiwan | Recruiting |
|
| |||||||
| TBI | |||||||
| NCT01575470 | I/II | Autologous bone marrow mononuclear cells |
IV | < 36 hours | Neurological events (incl. CVA) |
United States |
Recruiting |
| NCT01451528 | ? | Allogeneic umbilical cord blood |
IV/IA | < 6 months |
Short-term memory |
Republic of Korea |
Not yet recruiting |
| NCT00254722 | I | Autologous bone marrow precursor cell |
IV | Children only: < 36 hours |
Adverse events |
United States |
Completed |
| NCT01251003 | I/II | Autologous umbilical cord blood |
IV | Cildren only: 6-18 months |
Safety | United States |
Recruiting |
| NCT01851083 | I/II | Autologous bone marrow mononuclear cells |
IV | Children only: <36 hours |
Brain white matter and gray matter structural preservation |
United States |
Not yet recruiting |
IA: Intra-arterial; IC: Intracerebral; IT: Intrathecal ;IV: Intravenous
Although the beneficial effects of mesenchymal stem cells have been demonstrated in various experimental designs, as mentioned earlier, the mechanism of action of human bone-marrow-derived mononuclear cells in animal models remains uncertain. Future preclinical data will hopefully provide answers about optimal timing, route of administration and cell numbers. Ideally the most successful experimental design from rigorous preclinical studies should be translated to human clinical trials, although generalizing from animal models to humans is always difficult. Many transplantation strategies use a heterogeneous population of cells and which subtype in this heterogeneous population is of importance for the therapeutic benefit has to be clarified. Initially neuronal replacement by NSCs was thought to be the mechanism of action for stroke recovery. More recently it is thought to involve enhancement of endogenous plasticity by modulation of inflammation, promotion of angiogenesis, increased native axonal and dendritic outgrowth, neurotrophic effects and neuroprotection[79]. However, integration of transplanted cells in the brain needs further study in order to fully explore the potential of neuronal replacement. Characterization of transplanted cell differentiation in relation to functional improvement is required to determine which cells and cell subtypes are therapeutically effective and to understand the processes of integration into existing neural circuitries.
Elucidating mechanisms of action is important for selection of both cells and patients. Considering the relatively small sample sizes in the currently enrolling clinical trials it is unlikely that the full therapeutic potential of stem cells in stroke and TBI will be revealed in the near future. Trial results will hopefully provide insight into patient selection in relation to cell type used. One can hypothesize that lesion location and size might determine the optimal approach; for example functional recovery in smaller lesions would not require replacement strategies with higher procedural risks, but might benefit from techniques that increase endogenous repair mechanisms. Noninvasive imaging of transplanted cells will be essential to evaluate their survival, distribution, function and effects in patients.[80] Novel findings in experimental models and clinical trials should be combined for optimal phase III trial designs.
Conclusion
Several stem cell therapies tested in animal models for acute cerebral injury have provided preclinical evidence of improved functional recovery. The mechanism of action for many of these strategies, however, is not yet completely elucidated and clinical translation requires additional time. A ‘one size fits all’ therapy is unlikely, considering the heterogeneity of the cells used and the clinical diseases (both stroke and TBI encompass a wide range of acute cerebral insults). However, we have gained significant knowledge from preclinical studies about the mechanisms underlying recovery of neurologic function, and from early clinical trials about the safety and feasibility of cellular therapy for acute cerebral injury. The next decade of stem cell research in stroke and TBI will need to focus on additional basic and clinical research to fully explore the potential of this therapeutic avenue: From bench-to-bedside and just as importantly, from bedside-to-bench.
Key points.
Preclinical research of cell-based approaches after acute cerebral injury to ameliorate recovery is promising
Future basic research strategies should be aimed at identifying optimal cell type and numbers to use as well as comparing optimal routes and timing of delivery
Elucidating mechanisms of action of various stem cells in brain repair is crucial for the further development of cell-based therapies in stroke
Clinical translation is emerging, which is focused on safety, feasibility and functional improvement
Ongoing feedback between clinicians and experimental researchers is necessary to improve future clinical phase III trial design
Acknowledgements
This work was supported in part by NIH NINDS grant 2R01NS058784, Russell and Elizabeth Siegelman, and Bernard and Ronni Lacroute (to GKS). RL is Senior Clinical Investigator of FWO Flanders.
References
- 1.Roger VL, Go AS, Lloyd-Jones DM, et al. Executive Summary: Heart Disease and Stroke Statistics--2012 Update: A Report From the American Heart Association. Circulation. 2012;125(1):188–197. doi: 10.1161/CIR.0b013e3182456d46. [DOI] [PubMed] [Google Scholar]
- 2.Lansberg MG, Schrooten M, Bluhmki E, et al. Treatment time-specific number needed to treat estimates for tissue plasminogen activator therapy in acute stroke based on shifts over the entire range of the modified Rankin Scale. Stroke. 2009;40(6):2079–2084. doi: 10.1161/STROKEAHA.108.540708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Bouckaert M, Lemmens R, Thijs V. Reducing prehospital delay in acute stroke. Nat Rev Neurol. 2009;5(9):477–483. doi: 10.1038/nrneurol.2009.116. [DOI] [PubMed] [Google Scholar]
- 4.Taylor TN, Davis PH, Torner JC, et al. Lifetime cost of stroke in the United States. Stroke. 1996;27(9):1459–1466. doi: 10.1161/01.str.27.9.1459. [DOI] [PubMed] [Google Scholar]
- 5.Maas AI, Stocchetti N, Bullock R. Moderate and severe traumatic brain injury in adults. Lancet Neurol. 2008;7(8):728–741. doi: 10.1016/S1474-4422(08)70164-9. [DOI] [PubMed] [Google Scholar]
- 6.Lindvall O, Kokaia Z. Stem cells for the treatment of neurological disorders. Nature. 2006;441(7097):1094–1096. doi: 10.1038/nature04960. [DOI] [PubMed] [Google Scholar]
- 7.Stabenfeldt SE, Irons HR, Laplaca MC. Stem cells and bioactive scaffolds as a treatment for traumatic brain injury. Curr Stem Cell Res Ther. 2011;6(3):208–220. doi: 10.2174/157488811796575396. [DOI] [PubMed] [Google Scholar]
- 8.Bliss T, Guzman R, Daadi M, et al. Cell transplantation therapy for stroke. Stroke. 2007;38(2 Suppl):817–826. doi: 10.1161/01.STR.0000247888.25985.62. [DOI] [PubMed] [Google Scholar]
- 9.Andres RH, Horie N, Slikker W, et al. Human neural stem cells enhance structural plasticity and axonal transport in the ischaemic brain. Brain. 2011;134(Pt 6):1777–1789. doi: 10.1093/brain/awr094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.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]
- 11.Hicks AU, Lappalainen RS, Narkilahti S, 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. Eur J Neurosci. 2009;29(3):562–574. doi: 10.1111/j.1460-9568.2008.06599.x. [DOI] [PubMed] [Google Scholar]
- 12.Ishibashi S, Sakaguchi M, Kuroiwa T, et al. Human neural stem/progenitor cells, expanded in long-term neurosphere culture, promote functional recovery after focal ischemia in Mongolian gerbils. J Neurosci Res. 2004;78(2):215–223. doi: 10.1002/jnr.20246. [DOI] [PubMed] [Google Scholar]
- 13.Seminatore C, Polentes J, Ellman D, et al. The postischemic environment differentially impacts teratoma or tumor formation after transplantation of human embryonic stem cell-derived neural progenitors. Stroke. 2010;41(1):153–159. doi: 10.1161/STROKEAHA.109.563015. [DOI] [PubMed] [Google Scholar]
- 14.Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126(4):663–676. doi: 10.1016/j.cell.2006.07.024. [DOI] [PubMed] [Google Scholar]
- 15*.Oki K, Tatarishvili J, Wood J, et al. Human-induced pluripotent stem cells form functional neurons and improve recovery after grafting in stroke-damaged brain. Stem Cells. 2012;30(6):1120–1133. doi: 10.1002/stem.1104. First study to provide evidence that transplantation of human iPSC-derived cells is safe and efficacious for promoting recovery after stroke.
- 16.Buhnemann C, Scholz A, Bernreuther C, et al. Neuronal differentiation of transplanted embryonic stem cell-derived precursors in stroke lesions of adult rats. Brain. 2006;129(Pt 12):3238–3248. doi: 10.1093/brain/awl261. [DOI] [PubMed] [Google Scholar]
- 17.Daadi MM, Li Z, Arac A, et al. Molecular and magnetic resonance imaging of human embryonic stem cell-derived neural stem cell grafts in ischemic rat brain. Mol Ther. 2009;17(7):1282–1291. doi: 10.1038/mt.2009.104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18**.Espuny-Camacho I, Michelsen KA, Gall D, et al. Pyramidal neurons derived from human pluripotent stem cells integrate efficiently into mouse brain circuits in vivo. Neuron. 2013;77(3):440–456. doi: 10.1016/j.neuron.2012.12.011. In this article transplantation of in vitro generated human cortical neurons from iPSCs are shown to develop complex properties characteristic of the cerebral cortex in vivo.
- 19.Moshayedi P, Carmichael ST. Hyaluronan, neural stem cells and tissue reconstruction after acute ischemic stroke. Biomatter. 2013;3(1) doi: 10.4161/biom.23863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Horie N, Pereira MP, Niizuma K, et al. Transplanted stem cell-secreted vascular endothelial growth factor effects poststroke recovery, inflammation, and vascular repair. Stem Cells. 2011;29(2):274–285. doi: 10.1002/stem.584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Lee HJ, Kim KS, Park IH, et al. Human neural stem cells over-expressing VEGF provide neuroprotection, angiogenesis and functional recovery in mouse stroke model. PLoS One. 2007;2(1):e156. doi: 10.1371/journal.pone.0000156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Lee ST, Chu K, Jung KH, et al. Anti-inflammatory mechanism of intravascular neural stem cell transplantation in haemorrhagic stroke. Brain. 2008;131(Pt 3):616–629. doi: 10.1093/brain/awm306. [DOI] [PubMed] [Google Scholar]
- 23.Stroemer P, Patel S, Hope A, et al. The neural stem cell line CTX0E03 promotes behavioral recovery and endogenous neurogenesis after experimental stroke in a dose-dependent fashion. Neurorehabil Neural Repair. 2009;23(9):895–909. doi: 10.1177/1545968309335978. [DOI] [PubMed] [Google Scholar]
- 24**.Kokaia Z, Martino G, Schwartz M, et al. Cross-talk between neural stem cells and immune cells: the key to better brain repair? Nat Neurosci. 2012;15(8):1078–1087. doi: 10.1038/nn.3163. This review reflects on the current knowledge concerning the interaction between neural stem cells and cells belonging to the immune system.
- 25.Andrews EM, Tsai SY, Johnson SC, et al. Human adult bone marrow-derived somatic cell therapy results in functional recovery and axonal plasticity following stroke in the rat. Exp Neurol. 2008;211(2):588–592. doi: 10.1016/j.expneurol.2008.02.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Bao X, Wei J, Feng M, et al. Transplantation of human bone marrow-derived mesenchymal stem cells promotes behavioral recovery and endogenous neurogenesis after cerebral ischemia in rats. Brain Res. 2011;1367:103–113. doi: 10.1016/j.brainres.2010.10.063. [DOI] [PubMed] [Google Scholar]
- 27.Chen J, Zhang ZG, Li Y, et al. Intravenous administration of human bone marrow stromal cells induces angiogenesis in the ischemic boundary zone after stroke in rats. Circ Res. 2003;92(6):692–699. doi: 10.1161/01.RES.0000063425.51108.8D. [DOI] [PubMed] [Google Scholar]
- 28.Li Y, Chen J, Chen XG, et al. Human marrow stromal cell therapy for stroke in rat: neurotrophins and functional recovery. Neurology. 2002;59(4):514–523. doi: 10.1212/wnl.59.4.514. [DOI] [PubMed] [Google Scholar]
- 29.Vendrame M, Cassady J, Newcomb J, et al. Infusion of human umbilical cord blood cells in a rat model of stroke dose-dependently rescues behavioral deficits and reduces infarct volume. Stroke. 2004;35(10):2390–2395. doi: 10.1161/01.STR.0000141681.06735.9b. [DOI] [PubMed] [Google Scholar]
- 30.Wakabayashi K, Nagai A, Sheikh AM, et al. Transplantation of human mesenchymal stem cells promotes functional improvement and increased expression of neurotrophic factors in a rat focal cerebral ischemia model. J Neurosci Res. 2010;88(5):1017–1025. doi: 10.1002/jnr.22279. [DOI] [PubMed] [Google Scholar]
- 31.Zacharek A, Chen J, Cui X, et al. Angiopoietin1/Tie2 and VEGF/Flk1 induced by MSC treatment amplifies angiogenesis and vascular stabilization after stroke. J Cereb Blood Flow Metab. 2007;27(10):1684–1691. doi: 10.1038/sj.jcbfm.9600475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Zhang L, Li Y, Zhang C, et al. Delayed administration of human umbilical tissue-derived cells improved neurological functional recovery in a rodent model of focal ischemia. Stroke. 2011;42(5):1437–1444. doi: 10.1161/STROKEAHA.110.593129. [DOI] [PubMed] [Google Scholar]
- 33.Mora-Lee S, Sirerol-Piquer MS, Gutierrez-Perez M, et al. Therapeutic effects of hMAPC and hMSC transplantation after stroke in mice. PLoS One. 2012;7(8):e43683. doi: 10.1371/journal.pone.0043683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Liu YP, Seckin H, Izci Y, et al. Neuroprotective effects of mesenchymal stem cells derived from human embryonic stem cells in transient focal cerebral ischemia in rats. J Cereb Blood Flow Metab. 2009;29(4):780–791. doi: 10.1038/jcbfm.2009.1. [DOI] [PubMed] [Google Scholar]
- 35.Kranz A, Wagner DC, Kamprad M, et al. Transplantation of placenta-derived mesenchymal stromal cells upon experimental stroke in rats. Brain Res. 2010;1315:128–136. doi: 10.1016/j.brainres.2009.12.001. [DOI] [PubMed] [Google Scholar]
- 36.Skvortsova VI, Gubskiy LV, Tairova RT, et al. Use of bone marrow mesenchymal (stromal) stem cells in experimental ischemic stroke in rats. Bull Exp Biol Med. 2008;145(1):122–128. doi: 10.1007/s10517-008-0032-7. [DOI] [PubMed] [Google Scholar]
- 37.Borlongan CV, Tajima Y, Trojanowski JQ, et al. Transplantation of cryopreserved human embryonal carcinoma-derived neurons (NT2N cells) promotes functional recovery in ischemic rats. Exp Neurol. 1998;149(2):310–321. doi: 10.1006/exnr.1997.6730. [DOI] [PubMed] [Google Scholar]
- 38.Stroemer P, Hope A, Patel S, et al. Development of a human neural stem cell line for use in recovery from disability after stroke. Front Biosci. 2008;13:2290–2292. doi: 10.2741/2842. [DOI] [PubMed] [Google Scholar]
- 39.Gao J, Prough DS, McAdoo DJ, et al. Transplantation of primed human fetal neural stem cells improves cognitive function in rats after traumatic brain injury. Exp Neurol. 2006;201(2):281–292. doi: 10.1016/j.expneurol.2006.04.039. [DOI] [PubMed] [Google Scholar]
- 40.Lundberg J, Sodersten E, Sundstrom E, et al. Targeted intra-arterial transplantation of stem cells to the injured CNS is more effective than intravenous administration: engraftment is dependent on cell type and adhesion molecule expression. Cell Transplant. 2012;21(1):333–343. doi: 10.3727/096368911X576036. [DOI] [PubMed] [Google Scholar]
- 41.Skardelly M, Gaber K, Burdack S, et al. Long-term benefit of human fetal neuronal progenitor cell transplantation in a clinically adapted model after traumatic brain injury. J Neurotrauma. 2011;28(3):401–414. doi: 10.1089/neu.2010.1526. [DOI] [PubMed] [Google Scholar]
- 42.Wennersten A, Meier X, Holmin S, et al. Proliferation, migration, and differentiation of human neural stem/progenitor cells after transplantation into a rat model of traumatic brain injury. J Neurosurg. 2004;100(1):88–96. doi: 10.3171/jns.2004.100.1.0088. [DOI] [PubMed] [Google Scholar]
- 43.Riess P, Zhang C, Saatman KE, et al. Transplanted neural stem cells survive, differentiate, and improve neurological motor function after experimental traumatic brain injury. Neurosurgery. 2002;51(4):1043–1052. doi: 10.1097/00006123-200210000-00035. discussion 1052-1044. [DOI] [PubMed] [Google Scholar]
- 44.Yan ZJ, Zhang P, Hu YQ, et al. Neural stem-like cells derived from human amnion tissue are effective in treating traumatic brain injury in rat. Neurochem Res. 2013;38(5):1022–1033. doi: 10.1007/s11064-013-1012-5. [DOI] [PubMed] [Google Scholar]
- 45.Chen Z, Tortella FC, Dave JR, et al. Human amnion-derived multipotent progenitor cell treatment alleviates traumatic brain injury-induced axonal degeneration. J Neurotrauma. 2009;26(11):1987–1997. doi: 10.1089/neu.2008.0863. [DOI] [PubMed] [Google Scholar]
- 46.Hong SQ, Zhang HT, You J, et al. Comparison of transdifferentiated and untransdifferentiated human umbilical mesenchymal stem cells in rats after traumatic brain injury. Neurochem Res. 2011;36(12):2391–2400. doi: 10.1007/s11064-011-0567-2. [DOI] [PubMed] [Google Scholar]
- 47.Kim HJ, Lee JH, Kim SH. Therapeutic effects of human mesenchymal stem cells on traumatic brain injury in rats: secretion of neurotrophic factors and inhibition of apoptosis. J Neurotrauma. 2010;27(1):131–138. doi: 10.1089/neu.2008.0818. [DOI] [PubMed] [Google Scholar]
- 48.Lu D, Sanberg PR, Mahmood A, et al. Intravenous administration of human umbilical cord blood reduces neurological deficit in the rat after traumatic brain injury. Cell Transplant. 2002;11(3):275–281. [PubMed] [Google Scholar]
- 49.Nichols JE, Niles JA, Dewitt D, et al. Neurogenic and neuro-protective potential of a novel subpopulation of peripheral blood-derived CD133+ ABCG2+CXCR4+ mesenchymal stem cells: development of autologous cell-based therapeutics for traumatic brain injury. Stem Cell Res Ther. 2013;4(1):3. doi: 10.1186/scrt151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Watson DJ, Longhi L, Lee EB, et al. Genetically modified NT2N human neuronal cells mediate long-term gene expression as CNS grafts in vivo and improve functional cognitive outcome following experimental traumatic brain injury. J Neuropathol Exp Neurol. 2003;62(4):368–380. doi: 10.1093/jnen/62.4.368. [DOI] [PubMed] [Google Scholar]
- 51.Honma T, Honmou O, Iihoshi S, et al. Intravenous infusion of immortalized human mesenchymal stem cells protects against injury in a cerebral ischemia model in adult rat. Exp Neurol. 2006;199(1):56–66. doi: 10.1016/j.expneurol.2005.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52*.Lees JS, Sena ES, Egan KJ, et al. Stem cell-based therapy for experimental stroke: a systematic review and meta-analysis. Int J Stroke. 2012;7(7):582–588. doi: 10.1111/j.1747-4949.2012.00797.x. Systematic meta-analysis of studies reporting on the efficacy of stem cell-based therapies in animal models of stroke.
- 53.Song M, Kim YJ, Kim YH, et al. Effects of duplicate administration of human neural stem cell after focal cerebral ischemia in the rat. Int J Neurosci. 2011;121(8):457–461. doi: 10.3109/00207454.2011.576792. [DOI] [PubMed] [Google Scholar]
- 54.Yang M, Wei X, Li J, et al. Changes in host blood factors and brain glia accompanying the functional recovery after systemic administration of bone marrow stem cells in ischemic stroke rats. Cell Transplant. 2010;19(9):1073–1084. doi: 10.3727/096368910X503415. [DOI] [PubMed] [Google Scholar]
- 55.Han EY, Chun MH, Kim ST, et al. Injection time-dependent effect of adult human bone marrow stromal cell transplantation in a rat model of severe traumatic brain injury. Curr Stem Cell Res Ther. 2013;8(2):172–181. doi: 10.2174/1574888x11308020008. [DOI] [PubMed] [Google Scholar]
- 56*.Krakauer JW, Carmichael ST, Corbett D, et al. Getting neurorehabilitation right: what can be learned from animal models? Neurorehabil Neural Repair. 2012;26(8):923–931. doi: 10.1177/1545968312440745. The authors provide a view on what is required to improve the translation from animal models to clinical neurorehabilitation.
- 57.Jin K, Sun Y, Xie L, et al. Comparison of ischemia-directed migration of neural precursor cells after intrastriatal, intraventricular, or intravenous transplantation in the rat. Neurobiol Dis. 2005;18(2):366–374. doi: 10.1016/j.nbd.2004.10.010. [DOI] [PubMed] [Google Scholar]
- 58.Vasconcelos-dos-Santos A, Rosado-de-Castro PH, Lopes de Souza SA, et al. Intravenous and intra-arterial administration of bone marrow mononuclear cells after focal cerebral ischemia: Is there a difference in biodistribution and efficacy? Stem Cell Res. 2012;9(1):1–8. doi: 10.1016/j.scr.2012.02.002. [DOI] [PubMed] [Google Scholar]
- 59.Kondziolka D, Wechsler L, Goldstein S, et al. Transplantation of cultured human neuronal cells for patients with stroke. Neurology. 2000;55(4):565–569. doi: 10.1212/wnl.55.4.565. [DOI] [PubMed] [Google Scholar]
- 60.Kondziolka D, Steinberg GK, Wechsler L, et al. Neurotransplantation for patients with subcortical motor stroke: a phase 2 randomized trial. J Neurosurg. 2005;103(1):38–45. doi: 10.3171/jns.2005.103.1.0038. [DOI] [PubMed] [Google Scholar]
- 61.Savitz SI, Dinsmore J, Wu J, et al. Neurotransplantation of fetal porcine cells in patients with basal ganglia infarcts: a preliminary safety and feasibility study. Cerebrovasc Dis. 2005;20(2):101–107. doi: 10.1159/000086518. [DOI] [PubMed] [Google Scholar]
- 62.Rabinovich SS, Seledtsov VI, Banul NV, et al. Cell therapy of brain stroke. Bull Exp Biol Med. 2005;139(1):126–128. doi: 10.1007/s10517-005-0229-y. [DOI] [PubMed] [Google Scholar]
- 63.Suarez-Monteagudo C, Hernandez-Ramirez P, Alvarez-Gonzalez L, et al. Autologous bone marrow stem cell neurotransplantation in stroke patients. An open study. Restor Neurol Neurosci. 2009;27(3):151–161. doi: 10.3233/RNN-2009-0483. [DOI] [PubMed] [Google Scholar]
- 64.Barbosa da Fonseca LM, Gutfilen B, Rosado de Castro PH, et al. Migration and homing of bone-marrow mononuclear cells in chronic ischemic stroke after intra-arterial injection. Exp Neurol. 2010;221(1):122–128. doi: 10.1016/j.expneurol.2009.10.010. [DOI] [PubMed] [Google Scholar]
- 65.Bang OY, Lee JS, Lee PH, et al. Autologous mesenchymal stem cell transplantation in stroke patients. Ann Neurol. 2005;57(6):874–882. doi: 10.1002/ana.20501. [DOI] [PubMed] [Google Scholar]
- 66.Lee JS, Hong JM, Moon GJ, 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):1099–1106. doi: 10.1002/stem.430. [DOI] [PubMed] [Google Scholar]
- 67.Battistella V, de Freitas GR, da Fonseca LM, et al. Safety of autologous bone marrow mononuclear cell transplantation in patients with nonacute ischemic stroke. Regen Med. 2011;6(1):45–52. doi: 10.2217/rme.10.97. [DOI] [PubMed] [Google Scholar]
- 68.Savitz SI, Misra V, Kasam M, et al. Intravenous autologous bone marrow mononuclear cells for ischemic stroke. Ann Neurol. 2011;70(1):59–69. doi: 10.1002/ana.22458. [DOI] [PubMed] [Google Scholar]
- 69.Honmou O, Houkin K, Matsunaga T, et al. Intravenous administration of auto serum-expanded autologous mesenchymal stem cells in stroke. Brain. 2011;134(Pt 6):1790–1807. doi: 10.1093/brain/awr063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Bhasin A, Srivastava MV, Kumaran SS, 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]
- 71*.Bhasin A, Padma Srivastava MV, Mohanty S, et al. Stem cell therapy: A clinical trial of stroke. Clin Neurol Neurosurg. 2012 doi: 10.1016/j.clineuro.2012.10.015. Phase I/IIA clinical trial reporting on the transplantation of autologous bone marrow mononuclear and mesenchymal stem cells to be safe and feasible and potentially to improve recovery.
- 72.Prasad K, Mohanty S, Bhatia R, et al. Autologous intravenous bone marrow mononuclear cell therapy for patients with subacute ischaemic stroke: a pilot study. Indian J Med Res. 2012;136(2):221–228. [PMC free article] [PubMed] [Google Scholar]
- 73*.Moniche F, Gonzalez A, Gonzalez-Marcos JR, et al. Intra-arterial bone marrow mononuclear cells in ischemic stroke: a pilot clinical trial. Stroke. 2012;43(8):2242–2244. doi: 10.1161/STROKEAHA.112.659409. Article reporting on phaseI/IIA clinical trial with intra-arterial delivery of autologous bone marrow mononuclear cell in subacute ischemic stroke to be feasible and safe.
- 74*.Tian C, Wang X, Wang L, et al. Autologous bone marrow mesenchymal stem cell therapy in the subacute stage of traumatic brain injury by lumbar puncture. Exp Clin Transplant. 2013;11(2):176–181. doi: 10.6002/ect.2012.0053. First clinical trial in TBI patients delivering bone marrow stem cells by lumbar puncture. The therapy was shown to be safe and effective.
- 75.Stilley CS, Ryan CM, Kondziolka D, et al. Changes in cognitive function after neuronal cell transplantation for basal ganglia stroke. Neurology. 2004;63(7):1320–1322. doi: 10.1212/01.wnl.0000140700.44904.53. [DOI] [PubMed] [Google Scholar]
- 76.Minnerup J, Kim JB, Schmidt A, et al. Effects of neural progenitor cells on sensorimotor recovery and endogenous repair mechanisms after photothrombotic stroke. Stroke. 2011;42(6):1757–1763. doi: 10.1161/STROKEAHA.110.599282. [DOI] [PubMed] [Google Scholar]
- 77. http://www.reneuron.com/press-release/interim-data-from-clinical-trial-of-reneuron-s-stem-cell-therapy-for-stroke-to-be-presented-at-leading-stroke-conference-longer-term-data-continue-to-show-good-safety-profile-and-evidence-of-sustained-reductions-in-neurologica.
- 78.Chollet F, Tardy J, Albucher JF, et al. Fluoxetine for motor recovery after acute ischaemic stroke (FLAME): a randomised placebo-controlled trial. Lancet Neurol. 2011;10(2):123–130. doi: 10.1016/S1474-4422(10)70314-8. [DOI] [PubMed] [Google Scholar]
- 79.Bliss TM, Andres RH, Steinberg GK. Optimizing the success of cell transplantation therapy for stroke. Neurobiol Dis. 2010;37(2):275–283. doi: 10.1016/j.nbd.2009.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Manley NC, Steinberg GK. Tracking stem cells for cellular therapy in stroke. Curr Pharm Des. 2012;18(25):3685–3693. doi: 10.2174/138161212802002643. [DOI] [PMC free article] [PubMed] [Google Scholar]
