Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Review Article

Stem Cell Implants: Emerging Innovation for Stroke Recovery


Reid Colliander1, Kaitlyn Alleman1, Michael Diaz2, Med Jimenez1, Patrick King1, Pranav Mirpuri1, Christopher Cutler1,
Brandon Lucke-Wold3*
1Chicago
Medical School, Rosalind Franklin University, North Chicago, IL, USA
2College
of Medicine, University of Florida, Gainesville, FL, USA
3Department of Neurosurgery, University of Florida, Gainesville, FL, USA

*Correspondence author: Brandon Lucke-Wold, MD, PhD, MCTs, Department of Neurosurgery, University of Florida, Gainesville, FL, USA;
Email: Brandon.Lucke-Wold@neurosurgery.ufl.edu

Abstract
Citation: Lucke-Wold B, et al. Stem Stroke is a debilitating neurovascular injury that those effects hundreds of thousands of
Cell Implants: Emerging Innovation
Americans each year. Despite the high prevalence, disease morbidity and mortality, options for
for Stroke Recovery. J Neuro Onco
stroke intervention and rehabilitation are still limited. Stem cells have shown promise in stroke
Res. 2023;3(1):1-14.
treatment due to their ability to self-renew and differentiate into different cell types. The
https://doi.org/10.46889/JNOR.2023.
3102
primary sources of stem cells used today are bone marrow and fetal brain tissue, with
mesenchymal stem cells, bone marrow stem cells and neural stem cells being particularly well-
studied. By secreting therapeutic and neurogenic substances they are hypothesized to help
Received Date: 03-04-2023
foster recovery at the site of injury. Delivery mechanisms for stem cell therapy include
Accepted Date: 23-04-2023
intracerebral, intra-arterial, intraperitoneal, intravenous, intraventricular and intranasal routes
Published Date: 30-04-2023
with radiographic imaging now being used to monitor the progress of stem cell therapies. Stem
cell implants have been found to be safe but optimal treatment strategies are still being
established with several promising studies underway. Future efforts should continue to focus
on improving efficacy, exploring alternative stem cell sources, enhancing migration capability
Copyright: © 2023 by the authors.
and survival and educating stroke patients on the benefits and risks of stem cell therapy.
Submitted for possible open access
publication under the terms and
Abbreviations
conditions of the Creative Commons
Attribution (CCBY) AART: Action Research Arm Test; BI: Barthel Index; BMSC: Bone Marrow Stem Cell; CNS:
license
(https://creativecommons.org/li
Central Nervous System; CSF: Cerebrospinal Fluid; ESS: European Stroke Scale; EV:
censes/by/4.0/). Extracellular Vesicles; FMA: Fugl-Meyer Assessment; MAPC: Multipotent Adult Progenitor
Cell; MNC: Mononuclear Cell; MRI: Magnetic Resonance Imaging; mRS: Modified Rankin Scale;
MSC: Mesenchymal Stem Cell; MUSE: Multilineage-Differentiating Stress-Enduring Cell;
NIHSS: National Institute of Health Stroke Scale; NSC: Neural Stem Cell; PET: Positron
Emission Tomography; SCT: Stem Cell Therapy; tPA: tissue Plasminogen Activator; UC-MSC:
Umbilical Cord Derived Mesenchymal Stem Cells

Keywords: Stem Cell; Stroke; Exosomes; Neuroregenerative

Introduction
More than 795,000 strokes occur in the United States every year, with nearly 1 in 4 happening in those with a prior history of
stroke [1]. It is estimated that the stroke-related healthcare costs were approximately $53 billion between 2017 and 2018 [1].
Furthermore, stroke is a devastating disease in which survivors frequently experience motor and neurological deficits along with
a decreased quality of life [2,3]. Due to its high prevalence and disease morbidity, the treatment and management of stroke
remains an area of great therapeutic consequence.

https://doi.org/10.46889/JNOR.2023.3102 https://athenaeumpub.com/journal-of-neuro-and-oncology-research/
2

The many varying etiologies of stroke correspond to different treatments which depend largely on the type and timing of the
infarctive event. There are two different subtypes of stroke: ischemic and hemorrhagic. Ischemic strokes result from an occlusion
of the blood supply whereas hemorrhagic stroke is defined by bleeding from a vessel [4]. Ischemic strokes account for
approximately 87% of all strokes and uniquely can be treated with an intravenous injection of tissue Plasminogen Activator (tPA)
[1,5]. This promotes the dissolution of clots and restores patency to the occluded vessel. However, this intervention has a limited
time window and must be administered within 3-4.5 hours of insult [5,6]. Additional therapeutic options include antiplatelet
therapy, which has been shown to be effective at preventing a stroke recurrence within 48 hours but cannot ameliorate the
ischemia that has already occurred [6]. Surgery and other minimally invasive interventions are also used in the setting of ischemic
stroke and include procedures such as mechanical thrombectomy, ventriculostomy and decompressive craniectomy [7,8].
Mechanical thrombectomy is not indicated in all patients and additionally suffers from time constraints as well as it is only
effective up to 24 hours after initial presentation [9]. Unfortunately, there are few treatment options available to address
hemorrhagic strokes. These involve the management of hypertension and raised intracranial pressure, hemostatic therapy and
surgical intervention [10,11]. All of these treatments are largely focused on damage control and preventing spread or worsening
of damage from the inciting event. However, once Central Nervous System (CNS) neurons are damaged, they are for the most
part, unable to regenerate, with no current treatments available intended to renew the damaged neural networks [12]. New
research is hoping to bridge this gap in therapy, with stem cells offering a promise in neuroprotection and neuroregeneration in
the management of stroke.

Research into stem cells is still very much in its infancy. Less than 30 years ago the first primate embryonic stem cell line was
derived from rhesus monkeys and it was only in 1998 that the first human embryonic stem cells were derived by Thomson, et
al., [13,14]. Since then, the field has exploded with a variety of applications scoping many different domains of medicine [15].
There are three classes of stem cells that are being explored in the treatment of stroke. These include neural stem cells, bone
marrow stem cells and mesenchymal stem cells [16].

Mechanism of Action
Stem cells are cells that uniquely have the ability to self-renew and create functional tissues. More specifically, they can
differentiate into nearly any cell type present in the human body, which is what drew scientific interest to their therapeutic
potential in the first place. There are several major categories of stem cells: (1) Totipotent, (2) Pluripotent and (3) Multipotent
[17]. Totipotent cells can form all cell types within the human body, plus the extraembryonic cells that are crucial for early
development of the fertilized embryo. Pluripotent stem cells are commonly used in therapies as they can give rise to all cell types
within the human body. Multipotent stem cells are similar, but more limited in their differentiation potential- these cells include
the adult stem cell and the neural stem cell. Stem cells are further distinct from precursor cells, which are limited to differentiating
into a single cell type and progenitor cells, which cannot divide indefinitely [18,19].

Several mechanisms for the action of stem cells have been proposed over the years. Initially, it was thought that grafted cells
could replace dead sections of the brain and become integrated in their place. This hypothesis has given way to the modern
theory of the bystander effect, wherein implanted cells secrete therapeutic and neurogenic substances that ameliorate injury and
foster regeneration [20,21]. Ever since ischemic stroke was also recognized to impact non-neuronal cell components of the brain
such as glia and vasculature, stem cells are also thought to contribute to angiogenesis, vasculogenesis, anti-apoptosis and anti-
inflammation [22].

Primarily due to its well-established safety in the literature, the bone marrow is the primary source of stem cells in use today
[23,24]. Bone marrow derived populations include Mesenchymal Stem Cells (MSCs), bone marrow stem cells (BMSCs),
Mononuclear Cells (MNCs), endothelial progenitor cells (SB623), Multipotent Adult Progenitor Cells (MAPCs) and Multilineage-
differentiating Stress-Enduring cells (MUSE) [18]. Another primary source of stem cells is human brain fetal tissue, which has
been used to derive important cell lines of Neural Stem Cells (NSCs) [25].

Several initial studies determined the various neuroprotective and neuroregenerative effects of these cells in preclinical and later
clinical settings [26-28]. Based on these trials, MSCs, BMSCs and NSCs are particularly well-studied and promising. For example,
as an autologous cell with determined population purity and well-defined viability and multilineage differentiating potential,

https://doi.org/10.46889/JNOR.2023.3102 https://athenaeumpub.com/journal-of-neuro-and-oncology-research/
3

MSCs have been shown to promote neurogenesis and anti-inflammation, possibly mediated through the secretion of factors
including IL-1, TNF-alpha, IL-11 and TGF-β [20,29,30]. Studies on murine stroke models have also demonstrated the ability of
MSCs to decrease levels of axon growth inhibitors, leading to an increased density of functional axons in the ischemic area [31,32].
BMSCs exert similar mechanisms of action as secreted factors play roles in neurogenesis rather than neuronal replacement.
Unfortunately, neither MSCs or BMSCs studies have led to significant and replicable improvements in neurological outcomes of
stroke patients, but efforts continue. NSCs working through neuroregenerative effects too have shown promise in preclinical
models and there is some data to suggest that implantation of the cell line CTX0E03 has resulted in valid improvements in
functional recovery [33]. Notably, to some extent, all three cell lines have shown the ability to migrate to damaged areas and
even to guide the migration of endogenous cells via “biobridges” of matrix metalloproteinases in-vivo models [34,35]. However,
these clinical studies are very much in early stages of research; efficacious and generalizable criteria for stem cell administration
are still lacking.

The future of stem cell therapies is multifaceted. Naturally, clinical trials with different lineages of cells are underway, but beyond
this classic approach, it has been proposed recently to use derivatives of stem cells such as vesicles, mitochondria, exosomes and
micro-RNAs to foster regeneration in the ischemic area (Fig. 1). These cell components have been shown to exert similar paracrine
effects as compared to stem cells, resulting in neurogenesis and angiogenesis [36,37]. In a similar vein, it has been proposed that
if stem cell therapies are considered as biologics with regards to their paracrine effects, it may be easier to optimize delivery
routes, classify the stem-cell secreted factors and eventually design drugs that mimic these effects [38].

Figure 1: Mechanisms of stem cell therapy include (1) delivery of stem cell derivatives such as extracellular vesicles,
mitochondria and exosomes, (2) direct cell differentiation with replacement and induced migration with "biobridges" and (3)
paracrine secretion of various neurotrophic factors.

Delivery Mechanisms
Unsurprisingly, several different approaches for stem cell delivery exist in the laboratory setting and many have been translated
to clinical settings. Different administration routes have included intracerebral, intra-arterial, intraperitoneal, intravenous,

https://doi.org/10.46889/JNOR.2023.3102 https://athenaeumpub.com/journal-of-neuro-and-oncology-research/
4

intraventricular and intranasal [39]. Stem cells delivered intravenously and intrathecally in stroke patients has led to
improvements in terms of aphasia, motor deficits and spasticity [40]. The advantage of more invasive routes such as intracerebral
is that they allow for a larger number of implanted cells to reach target destinations, but suffer from limited generalizability,
immune rejection and increased risk for infection. On the other hand, less invasive routes such as intravenous do not suffer from
these disadvantages to the same extent and have even shown to have similar effectiveness compared to other delivery methods
to the point that most modern clinical trials prefer to utilize this technique [41,42]. However, one of the major drawbacks of
intravenous stem cell therapy is the pulmonary first-pass effect, meaning the total amount of stem cells injected does not reach
the site of injury. Depending on the type of stem cell, very few reach the target, thus diminishing the therapeutic effect [43]. There
have been recent strides in developing more targeted stem cell delivery methods, not only in terms of reaching the location of
injury but also addressing specific cellular defects following a stroke (Fig. 2).

Extracellular Vesicles (EVs) or exosomes are extracellular membrane-bound vesicles that transmit cargo without direct cellular
contact as a form of intercellular communication [44-46]. EVs have regenerative properties when derived from mesenchymal
stem cells [47]. Different exosome treatments that involve altering genetic properties have improved recovery of the damaged
tissue. For example, miR-542-3p is decreased following stroke in mice models and is thought to play a role in inflammation along
with TLR-4 [48]. An exosome treatment in which miR-542-3p was overexpressed and injected into a mouse stroke model paracele
led to decreased brain injury as well as decreased inflammation [48]. In a similar vein, neural stem cell exosomes treated with
interferon gamma exhibited stronger therapeutic effects in rats when stereotactically transplanted into the area of infarct when
compared to the group receiving the untreated neural stem cell exosomes. Interferon gamma was used to condition the exosomes
due to its ability to improve cell survival in conditions of oxidative stress [49]. Next, rat stroke models administered miR-17-92
cluster-enriched exosomes from mesenchymal stem cells intravenously had increased neurogenesis and neuroplasticity along
with oligodendrogenesis [50]. Further investigation regarding various ways to induce multiple genetic modifications to pre-
condition exosomes may enhance neurorestorative effects.

Much of the current stroke therapy literature turns to EVs as personalized targeted delivery vehicles especially in the application
of stroke where multipotent mesenchymal stromal cells have shown therapeutic effects [51,52]. An important aspect of NSC
therapy is the regulation of NSCs to differentiate into the desired cell fate after transplantation on neural tissue [53-56]. It is
known that many of the stem cells transplanted to the recipient host will commit to the glial progenitor lineage which limits the
therapeutic effects of NSCs in the setting of post-stroke and reperfusion therapy. Although delivery via exosomes may serve to
improve some of the effects of stroke, they are not able to target the site of injury and are therefore limited in terms of their
benefits. Nanoparticle delivery systems are one strategy developed to overcome this. One study involved using iron oxide
nanoparticles-harboring mesenchymal stem cells to enhance delivery to the ischemic lesion in rats. Following systemic injection
into the tail vein, the rats were fitted with a 3D-printed helmet containing a magnet which localized the nanoparticles to the area
of the lesion. This group had enhanced blood vessel density and decreased damage to the neurons in the location of the lesion
[57]. Lin, et al., demonstrated that the utilization of theranostic nanomedicine leads to a 3-4-fold increase in neuronal
differentiation of stem cells that can be detected in vivo with Magnetic Resonance Imaging (MRI) [53]. These findings suggest
the important role of nanomedicine in clinical therapy. Additionally, there is evidence to suggest that mitochondria may serve
as regulators of NSCs which can additionally play a role in targeted gene therapy [58].

Another obstacle has been the differentiation of neural stem cells into astrocytes rather than neurons. The use of
superparamagnetic iron oxide nanoparticles and small interfering RNA/antisense oligonucleotides against Pnky lncRNA in
combination with neural stem cells has shown promise in guiding differentiation into neurons as Pnky lncRNA inhibits neuronal
differentiation [59]. When this system was employed in mice by injecting the stem cells directly into the area of infarct, the infarct
volume was lower than that of mice given neural stem cells without this genetic alteration [53]. Another method for increased
therapeutic effect includes the use of biodegradable polymeric nanoparticles containing adipose stem cells genetically modified
to overexpress vascular endothelial growth factor [60].

https://doi.org/10.46889/JNOR.2023.3102 https://athenaeumpub.com/journal-of-neuro-and-oncology-research/
5

Figure 2: Depiction of two stem cell preparation methods and three delivery routes. The top portion of the figure shows a
method which involves genetically treating exosomes from stem cells prior to delivery. The bottom portion depicts packaging
the stem cells into nanoparticles and the right-hand side of the figure displays three major modes of delivery used in rodent
experiments: intracerebral injection, stereotactic injection and venous injection.

Outcomes Thus Far


The safety of stem cell implants in stroke patients has been well established in several phase 1 and 2 clinical trials [29,30,33,61-
82]. No severe adverse events attributable to stem cell therapy have been reported. Adverse events that have been attributed to
treatment have been minor, transient and primarily due to route of delivery (e.g., headache from intracranial injection)
[29,30,33,61-82].

While no trial has found significant safety concerns, efficacy has been harder to establish. Multiple single arm studies have shown
significant improvements from baseline in a number of measures of stroke burden, including modified Rankin Scale (mRS),
Barthel Index (BI), National Institute of Health Stroke Scale (NIHSS), European Stroke Scale (ESS), Fugl-Meyer Assessment
(FMA), Action Research Arm Test (ARAT) and lesion size [30,33,64,67-70,73,74,79]. For example, Steinberg, et al., followed 18
patients with chronic stroke symptoms (6 months to 5 years) for 2 years after intracranial injection with mesenchymal stem cells
and found improvement from baseline in ESS, NIHSS and FMA that plateaued at 12 months but had not relapsed by the end of
the study [30,70].

Improvement relative to placebos, however, have not been consistent in randomized controlled trials [29,61-63,65,66,71,72,75-
78,80-82]. For example, the ACTIsSIMA trial studied 163 patients using the same cell type and in the same clinical setting as
Steinberg et al., failed to find an improvement in treatment relative to controls (the results are as yet unpublished, but available
on clinicaltrials.gov, trial number: NCT02448641). Other RCTs using intravenous mesenchymal cells in the acute/subacute setting
have had mixed results with improvement in some measures of stroke burden but not others and inconsistency between studies
in which measures show improvement [61,63,66,75,76,78]. Studies investigating CD34+ stem cells have similarly shown mixed
results, with improvement in NIHSS, ESS and mRS with intracranial injection in the chronic setting, but not with intravenous
and subcutaneous injections in the acute/subacute setting [62,65,71,81].

https://doi.org/10.46889/JNOR.2023.3102 https://athenaeumpub.com/journal-of-neuro-and-oncology-research/
6

A recent meta-analysis of randomized trials with scores on the mRS, NIHSS and BI at 6, 12 and 24 months found significant
differences in NIHSS at 6 months and mRS at 12 months that favored treatment [83]. Although differences in the other measures
were not statistically significant, the authors were only able to pool data from four studies (with each analysis containing no
more than three), resulting in only between 50 and 81 patients being analyzed for each outcome. This small overall sample along
with significant heterogeneity between studies (in part due to inclusion regardless of cell type, route and clinical setting) resulted
in wide confidence intervals that, while unable to exclude the null, were still consistent with substantial functional improvement
with treatment [83].

Given the large number of parameters that can vary in this space (cell type, dose, route of delivery, rounds of treatment, time
since stroke onset, etc.), the number of possible best use cases is vast. Different methods may be better suited in the acute versus
the chronic setting and stacking the optimal treatment in each setting may have significant synergistic effects. The established
safety of the treatment, along with the exciting possibilities warrants further study and a number of ongoing trials are doing just
that Table 1 shows trials currently ongoing in this space along with cell type, route of delivery, clinical setting, trial phase, sample
size, trial design and estimated completion date.

Trial/location/acronym Cell Route Setting Phase n Design Estimated Completion Date

NCT05008588 UC-MSC IC acute 1/2 15 RCT-OL December 2023


Indonesia

NCT04811651 UC-MSC IV <6 2 200 RCT-B October 1, 2023


China months
“UMSIS”

NCT05292625 UC-MSC IV/IT <24 1/2 48 RCT-B June 2, 2023


Vietnam months

NCT04631406 NSC IC 6-60 1/2 30 Single Arm December 31, 2024


USA/Canada months

NCT05158101 UC-MSC IV N/A 1 15 Single Arm February 2026


Argentina

NCT04280003 A-MSC IV <4 days 2 30 RCT-B July 15, 2023


Spain
“AMASCIS-02”

NCT01151124 NSC IC 6-60 1 12 Single Arm March 2023


UK months
“PISCES”

NCT04434768 UC-MSC IV/IV+I <36 1 14 Single Arm December 31, 2023


Taiwan A hours

NCT04097652 UC-MSC IV 4-7 days 1 9 Single Arm December 31, 2025


Taiwan

NCT03384433 MSC-CfE IV <24 1/2 5 Single Arm December 17, 2021


Iran hours

https://doi.org/10.46889/JNOR.2023.3102 https://athenaeumpub.com/journal-of-neuro-and-oncology-research/
7

NCT04953663 BM-MSC IV >6 1/2 60 RCT-B January 1, 2023


China months

NCT03545607 APC IV 18-36 3 300 RCT-B June 2023


USA hours

NCT04590118 BM-MSC IV >6 1/2 60 RCT-B August 1, 2023


China months

NCT02795052 BM-MSC IV+IN >6 N/A 500 Single Arm July 2024
UAE months

NCT04093336 BM-MSC IV <7 days 1/2 120 RCT-B August 31, 2024
China

USA = United States of America, UAE = United Arab Emirates, UC-MSC = Umbilical Cord Derived Mesenchymal Stem Cells,
NSC = Neural Stem Cells, A-MSC = Adipose Derived Mesenchymal Stem Cells, MSC-CfE = Mesenchymal Stem Cell Derived
Cell-free Exomes, BM-MSC = Bone Marrow Derived Mesenchymal Stem Cells, APC = Multipotent Adult Progenitor Cells, IC =
Intracerebral, IV = Intravenous, IA = Intraarterial, IN = Intranasal, RCT-OL = Open Label Randomized Controlled Trial, RCT-B =
Blinded Randomized Controlled Trial
Table 1: Ongoing trials.

Radiographic Findings
The promise of stem cell therapies has mandated the study of how cells are trafficked and utilized in the body. In the preclinical
setting, much of this information has come from dissection and analysis of frozen specimens. Imaging as a noninvasive method
of monitoring therapeutic efficacy has been explored more recently. Classically there are two different approaches to tracking
stem cells: direct and indirect labeling with markers such as super paramagnetic iron oxides, perfluoropolyether and
perfluorocarbon which can be picked up by MRI or Positron Emission Tomography (PET) scanning [84]. Several studies have
investigated these techniques, but challenges have included the limitations of spatial resolution of current imaging modalities
and the dynamics of labeling, such as the quantification of tracer uptake and the concentration of tagged cells in a sample [85-
88]. One recent development aimed at managing these challenges is the introduction of reporter genes in stem cells, which when
expressed form molecules that can bind to specific injected radiotracers allowing for easier visualization of stem cell migration
[89].

Neuron-derived EVs regulate the Blood-Brain Barrier (BBB) in both physiological and pathological states [90]. Because EVs can
cross the BBB, they may serve a role when looking for stroke biomarkers or radiological imaging. Some studies already utilize
CD9 or CD63 as a specific marker for EVs that can be fluorescently tagged with GFP in neuroglia and tracked for radiographic
studies [91,92]. Fig. 3 highlights the main functions of exosomes which are being utilized in research as drug therapy vehicles
such as in the setting of stroke. In animal models, Neural Stem Cells (NSCs) in EVs were shown to improve functional outcomes
post-thromboembolic stroke [93]. Exosomes have promising roles in clinical therapy while also allowing for the visualization of
using biomarkers to understand their intercellular interactions and therapeutic effects [45,46].

Radiographic assessment is important for localizing neurological defects and understanding the sequelae of stroke. In acute
settings of suspected stroke, Computed Tomography (CT) can be utilized due to fast imaging speed, wide availability and
production of good contrast between a bright clot and surrounding lower attenuating cortical tissue and Cerebrospinal Fluid
(CSF) [94-96]. MRI can also be utilized but takes longer to obtain imaging [94-96]. In one study, radiographic results administering
EV NSCs in the context of stroke showed decreased infarct volume [93]. This decrease was shown in settings where the EV NSCs
were administered both within and outside the tPA clinical therapeutic window [93]. To elaborate on this finding, other studies
show coupling of angiogenesis and neurogenesis in settings of neurovascular ischemia [97,98]. It is known that when embryonic
stem cells are introduced into rat brains during times of focal cerebral ischemia, these cells can be tracked in-vivo migrating along

https://doi.org/10.46889/JNOR.2023.3102 https://athenaeumpub.com/journal-of-neuro-and-oncology-research/
8

the corpus callosum to reach these ischemic areas from distances as far as the opposite hemisphere [99]. Porcine studies utilizing
EV NSCs also show significant neuroprotection from induced ischemic stroke [100]. MRI results comparing the NSC treatment
group from placebo show at the 84-day mark significant tissue level recovery, decreased cerebral lesion volume and preservation
of white matter integrity [100]. The current research utilizing animal models shows evidence of potential clinical use of NSCs in
the setting of acute ischemic stroke which can be monitored radiographically for spatial and temporal changes.

As discussed in the previous section, initial data regarding clinical Phase I/II trials of the utilization of mesenchymal stem cells
in chronic stroke shows promise. In two of these studies, MRI results showed no progression or new ischemic lesions in the brain
between the delivery of NSCs and prior to discharge [64,101]. Interestingly, one trial showed a reduction of mean lesion volume
>20% at the one-week mark post mesenchymal stem cell infusion as evident on MRI [64]. These early clinical findings show much
promise in the therapeutic utilization of NSCs in the treatment of ischemic stroke.

Figure 3: Exosome function and utilization as nanomedicine. Exosomes are nano-sized (30-150 nm) extracellular vesicles that
transmit cargo to other cells. Much of the recent research utilizes this function to insert drugs and proteins as a form of
regenerative medicine.

Areas Needed for Further Discovery


In the present study, we critically appraised and critiqued the wealth of emerging cell therapy research for stroke recovery.
Though much has been achieved, several areas for focused inquiry and effort have been identified. The most pressing is to
establish the efficacy of Stem Cell Therapy (SCT) as a therapeutic tool, as discussed earlier. However, additional areas for
exploration include developing alternative stem cell sources and wider patient education on SCT.

Intracerebral transplantation of BMSC and MSC implants has demonstrated early promise in promoting the neurogenesis
process post-stroke, but both remain steadily inefficacious relative to NSC implants [18,102]. Current evidence suggests that
further exploration of BMSC and MSC, as well as Umbilical Cord Derived Mesenchymal Stem Cells (UC-MSC), represents a

https://doi.org/10.46889/JNOR.2023.3102 https://athenaeumpub.com/journal-of-neuro-and-oncology-research/
9

worthwhile pursuit in light of their immunomodulatory properties, which may prove clinically valuable for tackling prolonged
post-stroke inflammation states [18,103]. The endogenous repair mechanisms promoted by MSCs have been specifically
attributed to recruitment of immune mediators IL-6 and PGE2 plus responsiveness to IL-1β, which enhances monocyte and
granulocyte recruitment for functional reduction of infarct volume [42,104]. However, despite evidence indicating the safe
implantation of such stem cell populations in human replacement trials, establishing efficacy and best use cases with narrowing
indications is the biggest challenge for future research [18,42,103]. There also exists a surge of early data proposing new
expandable cell lines and methodologies with marked pre-clinical potential for neuropathologies [105-107]. These and the above-
described findings taken together, future stroke research should concentrate efforts on enhancement of stem cell migration
capability and survival within the ischemic environment.

Moreover, patient attitudes and understanding surrounding SCT also represent a much-desired focus area. The available
literature suggests that stroke patients are stratified in their attitudes towards SCT, which may conceivably limit their willingness
to participate in clinical trials or, perhaps worse yet, motivate participation without consideration of the associated risks [108-
110]. Akid and colleagues reported that in a controlled sample of 84 ischemic stroke patients, 12% (10/84) had prior knowledge
of SCT but upwards of 36% (30) reported a willingness to participate in SCT clinical trials after receiving information [108].
Further, they observed that male gender was significantly correlated with positive SCT attitude (OR: 3.74, 95% CI: 1.45-9.61)
[108]. Unsworth, et al., additionally identified younger age, greater perceived caregiver burden and poorer physical functioning,
among others, as strong predictors for considering experimental SCT [110]. It is therefore clear that clinicians must place due
emphasis on educating their SCT candidate populations to appreciate patient perspectives and ensure better-informed consent
protocols in the high-risk neurosurgical arena.

Conclusion
Stroke remains a debilitating and costly sector of the healthcare system with limited options for intervention and rehabilitation.
Due to their unique ability of selective differentiation, stem cells provide a great opportunity to bridge the gap in treatment for
these patients. In-vitro and in-vivo studies have already demonstrated promise in the use of MSC, BMSC and NSC with greater
studies on the horizon. Through the use of exosomes and nanoparticles, intravenous and intrathecal delivery of stem cells may
soon provide therapeutic benefit to those suffering from cognitive and neurological impairments following a stroke.

Acknowledgements
We would like to acknowledge BioRender.Com for allowing us to use their software to generate all of the images and figures
displayed in this review.

Conflict of Interest
The authors have no conflict of interest to declare.

References
1. Tsao CW, Aday AW, Almarzooq ZI. Heart disease and stroke statistics-2022 update: a report from the american heart
association. Circulation. 2022;145(8):e153-e639.
2. Baumann M, Le Bihan E, Chau K, Chau N. Associations between quality of life and socioeconomic factors, functional
impairments and dissatisfaction with received information and home-care services among survivors living at home two
years after stroke onset. BMC Neurol. 2014;14:92.
3. Adamson J, Beswick A, Ebrahim S. Is stroke the most common cause of disability? J Stroke Cerebrovasc Dis. 2004;13(4):171-
7.
4. Chang JC. Stroke classification: critical role of unusually large von Willebrand factor multimers and tissue factor on clinical
phenotypes based on novel "two-path unifying theory" of hemostasis. Clin Appl Thromb Hemost. 2020;26:1076029620913634.
5. Tissue plasminogen activator for acute ischemic stroke. N Engl J Med. 1995;333(24):1581-7.
6. Bansal S, Sangha KS, Khatri P. Drug treatment of acute ischemic stroke. Am J Cardiovasc Drugs. 2013;13(1):57-69.
7. Shah A, Almenawer S, Hawryluk G. Timing of decompressive craniectomy for ischemic stroke and traumatic brain injury: a
review. Front Neurol. 2019;10:11.

https://doi.org/10.46889/JNOR.2023.3102 https://athenaeumpub.com/journal-of-neuro-and-oncology-research/
10

8. Dzierwa K, Knapik M, Tekieli Ł. Clinical outcomes of extracranial carotid artery-related stroke eligible for mechanical
reperfusion on top of per-guidelines thrombolytic therapy: analysis from a 6-month consecutive patient sample in 2 centers.
Med Sci Monit. 2022;28:e938549.
9. Inoue M, Yoshimoto T, Tanaka K. Mechanical thrombectomy up to 24 hours in large vessel occlusions and infarct velocity
assessment. J Am Heart Assoc. 2021;10(24):e022880.
10. De Oliveira Manoel AL. Surgery for spontaneous intracerebral hemorrhage. Crit Care. 2020;24(1):45.
11. Dastur CK, Yu W. Current management of spontaneous intracerebral haemorrhage. Stroke Vasc Neurol. 2017;2(1):21-9.
12. Huebner EA, Strittmatter SM. Axon regeneration in the peripheral and central nervous systems. Results Probl Cell Differ.
2009;48:339-51.
13. Eguizabal C, Aran B, Chuva de Sousa Lopes SM. Two decades of embryonic stem cells: a historical overview. Hum Reprod
Open. 2019;2019(1):hoy024.
14. Thomson JA, Itskovitz-Eldor J, Shapiro SS. Embryonic stem cell lines derived from human blastocysts. Science.
1998;282(5391):1145-7.
15. Zakrzewski W, Dobrzyński M, Szymonowicz M, Rybak Z. Stem cells: past, present and future. Stem Cell Res Ther.
2019;10(1):68.
16. Nistor-Cseppentö DC, Jurcău MC, Jurcău A, Andronie-Cioară FL, Marcu F. Stem cell- and cell-based therapies for ischemic
stroke. Bioengineering (Basel). 2022;9(11).
17. Lajtha LG. Stem cell concepts. Differentiation. 1979;14(1-2):23-34.
18. Chrostek MR, Fellows EG, Crane AT, Grande AW, Low WC. Efficacy of stem cell-based therapies for stroke. Brain Res.
2019;1722:146362.
19. Alvarez CV, Garcia-Lavandeira M, Garcia-Rendueles ME. Defining stem cell types: understanding the therapeutic potential
of ESCs, ASCs and iPS cells. J Mol Endocrinol. 2012;49(2):R89-111.
20. Savitz SI, Chopp M, Deans R, Carmichael T, Phinney D, Wechsler L. Stem cell therapy as an emerging paradigm for Stroke
(STEPS) II. Stroke. 2011;42(3):825-9.
21. Lindvall O, Kokaia Z. Stem cell research in stroke: how far from the clinic? Stroke. 2011;42(8):2369-75.
22. Borlongan CV. Concise review: stem cell therapy for stroke patients: are we there yet? Stem Cells Transl Med. 2019;8(9):983-
8.
23. Napoli E, Borlongan CV. Recent advances in stem cell-based therapeutics for stroke. Transl Stroke Res. 2016;7(6):452-7.
24. Napoli E, Lippert T, Borlongan CV. Stem cell therapy: repurposing cell-based regenerative medicine beyond cell
replacement. Adv Exp Med Biol. 2018;1079:87-91.
25. Pollock K, Stroemer P, Patel S. A conditionally immortal clonal stem cell line from human cortical neuroepithelium for the
treatment of ischemic stroke. Exp Neurol. 2006;199(1):143-55.
26. Kondziolka D, Wechsler L, Goldstein S. Transplantation of cultured human neuronal cells for patients with stroke. Neurol.
2000;55(4):565-9.
27. Bateman ME, Strong AL, Gimble JM, Bunnell BA. Concise Review: Using fat to fight disease: a systematic review of
nonhomologous adipose-derived stromal/stem cell therapies. Stem Cells. 2018;36(9):1311-28.
28. Napoli E, Borlongan CV. Stem cell recipes of bone marrow and fish: just what the stroke doctors ordered. Stem Cell Rev Rep.
2017;13(2):192-7.
29. Prasad K, Sharma A, Garg A. Intravenous autologous bone marrow mononuclear stem cell therapy for ischemic stroke: a
multicentric, randomized trial. Stroke. 2014;45(12):3618-24.
30. Steinberg GK, Kondziolka D, Wechsler LR. Two-year safety and clinical outcomes in chronic ischemic stroke patients after
implantation of modified bone marrow-derived mesenchymal stem cells (SB623): a phase 1/2a study. J Neurosurg. 2018:1-
11.
31. Liu Z, Li Y, Qu R. Axonal sprouting into the denervated spinal cord and synaptic and postsynaptic protein expression in the
spinal cord after transplantation of bone marrow stromal cell in stroke rats. Brain Res. 2007;1149:172-80.
32. Shen LH, Li Y, Gao Q, Savant-Bhonsale S, Chopp M. Down-regulation of neurocan expression in reactive astrocytes promotes
axonal regeneration and facilitates the neurorestorative effects of bone marrow stromal cells in the ischemic rat brain. Glia.
2008;56(16):1747-54.
33. Kalladka D, Sinden J, Pollock K. Human neural stem cells in Patients with Chronic Ischaemic Stroke (PISCES): a phase 1,

https://doi.org/10.46889/JNOR.2023.3102 https://athenaeumpub.com/journal-of-neuro-and-oncology-research/
11

first-in-man study. Lancet. 2016;388(10046):787-96.


34. Tajiri N, Kaneko Y, Shinozuka K. Stem cell recruitment of newly formed host cells via a successful seduction? Filling the gap
between neurogenic niche and injured brain site. PLoS One. 2013;8(9):e74857.
35. Sullivan R, Duncan K, Dailey T, Kaneko Y, Tajiri N, Borlongan CV. A possible new focus for stroke treatment - migrating
stem cells. Expert Opin Biol Ther. 2015;15(7):949-58.
36. Rong Y, Liu W, Wang J. Neural stem cell-derived small extracellular vesicles attenuate apoptosis and neuroinflammation
after traumatic spinal cord injury by activating autophagy. Cell Death Dis. 2019;10(5):340.
37. Zhang Y, Liu Y, Liu H, Tang WH. Exosomes: biogenesis, biologic function and clinical potential. Cell Biosci. 2019;9:19.
38. Tuazon JP, Castelli V, Borlongan CV. Drug-like delivery methods of stem cells as biologics for stroke. Expert Opin Drug
Deliv. 2019;16(8):823-33.
39. Liu X, Ye R, Yan T. Cell based therapies for ischemic stroke: from basic science to bedside. Prog Neurobiol. 2014;115:92-115.
40. Stancioiu F, Papadakis GZ, Lazopoulos G. CD271(+) stem cell treatment of patients with chronic stroke. Exp Ther Med.
2020;20(3):2055-62.
41. Schreiber R, Hollands R, Blokland A. A mechanistic rationale for PDE-4 inhibitors to treat residual cognitive deficits in
acquired brain injury. Curr Neuropharmacol. 2020;18(3):188-201.
42. Berlet R, Anthony S, Brooks B. Combination of stem cells and rehabilitation therapies for ischemic stroke. Biomolecules.
2021;11(9).
43. Fischer UM, Harting MT, Jimenez F. Pulmonary passage is a major obstacle for intravenous stem cell delivery: the pulmonary
first-pass effect. Stem Cells Dev. 2009;18(5):683-92.
44. Lai CP, Breakefield XO. Role of exosomes/microvesicles in the nervous system and use in emerging therapies. Front Physiol.
2012;3:228.
45. Rak J. Extracellular vesicles - biomarkers and effectors of the cellular interactome in cancer. Front Pharmacol. 2013;4:21.
46. Chen J, Chopp M. Exosome therapy for stroke. Stroke. 2018;49(5):1083-90.
47. Zhang ZG, Buller B, Chopp M. Exosomes - beyond stem cells for restorative therapy in stroke and neurological injury. Nat
Rev Neurol. 2019;15(4):193-203.
48. Cai G, Cai G, Zhou H. Mesenchymal stem cell-derived exosome miR-542-3p suppresses inflammation and prevents cerebral
infarction. Stem Cell Res Ther. 2021;12(1):2.
49. Zhang G, Zhu Z, Wang H. Exosomes derived from human neural stem cells stimulated by interferon gamma improve
therapeutic ability in ischemic stroke model. J Adv Res. 2020;24:435-45.
50. Xin H, Katakowski M, Wang F. MicroRNA cluster miR-17-92 cluster in exosomes enhance neuroplasticity and functional
recovery after stroke in rats. Stroke. 2017;48(3):747-53.
51. Luarte A, Bátiz LF, Wyneken U, Lafourcade C. Potential therapies by stem cell-derived exosomes in CNS diseases: focusing
on the neurogenic niche. Stem Cells Int. 2016;2016:5736059.
52. Xin H, Li Y, Cui Y, Yang JJ, Zhang ZG, Chopp M. Systemic administration of exosomes released from mesenchymal stromal
cells promote functional recovery and neurovascular plasticity after stroke in rats. J Cereb Blood Flow Metab.
2013;33(11):1711-5.
53. Lin B, Lu L, Wang Y. Nanomedicine Directs neuronal differentiation of neural stem cells via silencing long noncoding RNA
for stroke therapy. Nano Lett. 2021;21(1):806-15.
54. Darsalia V, Allison SJ, Cusulin C. Cell number and timing of transplantation determine survival of human neural stem cell
grafts in stroke-damaged rat brain. J Cereb Blood Flow Metab. 2011;31(1):235-42.
55. Riemens RJM, Van Den Hove DLA, Esteller M, Delgado-Morales R. Directing neuronal cell fate in-vitro: achievements and
challenges. Prog Neurobiol. 2018;168:42-68.
56. Wilson KL, Pérez SCL, Naffaa MM, Kelly SH, Segura T. Stoichiometric post-modification of hydrogel microparticles dictates
neural stem cell fate in microporous annealed particle scaffolds. Adv Mater. 2022;34(33):e2201921.
57. Kim HY, Kim TJ, Kang L. Mesenchymal stem cell-derived magnetic extracellular nanovesicles for targeting and treatment of
ischemic stroke. Biomaterials. 2020;243:119942.
58. Khacho M, Harris R, Slack RS. Mitochondria as central regulators of neural stem cell fate and cognitive function. Nat Rev
Neurosci. 2019;20(1):34-48.
59. Ramos AD, Andersen RE, Liu SJ. The long noncoding RNA Pnky regulates neuronal differentiation of embryonic and

https://doi.org/10.46889/JNOR.2023.3102 https://athenaeumpub.com/journal-of-neuro-and-oncology-research/
12

postnatal neural stem cells. Cell Stem Cell. 2015;16(4):439-47.


60. Keeney M, Deveza L, Yang F. Programming stem cells for therapeutic angiogenesis using biodegradable polymeric
nanoparticles. J Vis Exp. 2013(79):e50736.
61. Jaillard A, Hommel M, Moisan A. Autologous Mesenchymal Stem Cells Improve Motor Recovery in Subacute Ischemic
Stroke: a Randomized Clinical Trial. Transl Stroke Res. 2020;11(5):910-23.
62. Sprigg N, Bath PM, Zhao L. Granulocyte-colony-stimulating factor mobilizes bone marrow stem cells in patients with
subacute ischemic stroke: the Stem cell Trial of recovery EnhanceMent after Stroke (STEMS) pilot randomized, controlled
trial (ISRCTN 16784092). Stroke. 2006;37(12):2979-83.
63. Lee JS, Hong JM, Moon GJ, Lee PH, Ahn YH, Bang OY. 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.
64. Honmou O, Houkin K, Matsunaga T. Intravenous administration of auto serum-expanded autologous mesenchymal stem
cells in stroke. Brain. 2011;134(Pt 6):1790-807.
65. England TJ, Abaei M, Auer DP. Granulocyte-colony stimulating factor for mobilizing bone marrow stem cells in subacute
stroke: the stem cell trial of recovery enhancement after stroke 2 randomized controlled trial. Stroke. 2012;43(2):405-11.
66. Bang OY, Lee JS, Lee PH, Lee G. Autologous mesenchymal stem cell transplantation in stroke patients. Ann Neurol.
2005;57(6):874-82.
67. Banerjee S, Bentley P, Hamady M. Intra-arterial immunoselected CD34+ stem cells for acute ischemic stroke. Stem Cells
Transl Med. 2014;3(11):1322-30.
68. Vahidy FS, Haque ME, Rahbar MH. Intravenous bone marrow mononuclear cells for acute ischemic stroke: safety, feasibility
and effect size from a phase I clinical trial. Stem Cells. 2019;37(11):1481-91.
69. Taguchi A, Sakai C, Soma T. Intravenous autologous bone marrow mononuclear cell transplantation for stroke: phase1/2a
clinical trial in a homogeneous group of stroke patients. Stem Cells Dev. 2015;24(19):2207-18.
70. Steinberg GK, Kondziolka D, Wechsler LR. Clinical outcomes of transplanted modified bone marrow-derived mesenchymal
stem cells in stroke: a phase 1/2a study. Stroke. 2016;47(7):1817-24.
71. Sprigg N, O'Connor R, Woodhouse L. Granulocyte colony stimulating factor and physiotherapy after stroke: results of a
feasibility randomised controlled trial: stem cell trial of recovery enhancement after Stroke-3 (STEMS-3 ISRCTN16714730).
PLoS One. 2016;11(9):e0161359.
72. Savitz SI, Yavagal D, Rappard G. A Phase 2 randomized, sham-controlled trial of internal carotid artery infusion of
autologous bone marrow-derived ALD-401 cells in patients with recent stable ischemic Stroke (RECOVER-Stroke).
Circulation. 2019;139(2):192-205.
73. Muir KW, Bulters D, Willmot M. Intracerebral implantation of human neural stem cells and motor recovery after stroke:
multicentre Prospective Single-Arm Study (PISCES-2). J Neurol Neurosurg Psychiatry. 2020;91(4):396-401.
74. Levy ML, Crawford JR, Dib N, Verkh L, Tankovich N, Cramer SC. Phase I/II study of safety and preliminary efficacy of
intravenous allogeneic mesenchymal stem cells in chronic stroke. Stroke. 2019;50(10):2835-41.
75. Lee J, Chang WH, Chung JW. Efficacy of intravenous mesenchymal stem cells for motor recovery after ischemic stroke: a
neuroimaging study. Stroke. 2022;53(1):20-8.
76. Law ZK, Tan HJ, Chin SP. The effects of intravenous infusion of autologous mesenchymal stromal cells in patients with
subacute middle cerebral artery infarct: a phase 2 randomized controlled trial on safety, tolerability and efficacy.
Cytotherapy. 2021;23(9):833-40.
77. Hess DC, Wechsler LR, Clark WM. 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.
78. Chung JW, Chang WH, Bang OY. Efficacy and safety of intravenous mesenchymal stem cells for ischemic stroke. Neurology.
2021;96(7):e1012-23.
79. Sharma A, Sane H, Gokulchandran N. Autologous bone marrow mononuclear cells intrathecal transplantation in chronic
stroke. Stroke Res Treat. 2014;2014:234095.
80. Bhasin A, Srivastava MVP, Mohanty S. Paracrine mechanisms of intravenous bone marrow-derived mononuclear stem cells
in chronic ischemic stroke. Cerebrovasc Dis Extra. 2016;6(3):107-19.
81. Chen DC, Lin SZ, Fan JR. Intracerebral implantation of autologous peripheral blood stem cells in stroke patients: a
randomized phase II study. Cell Transplant. 2014;23(12):1599-612.

https://doi.org/10.46889/JNOR.2023.3102 https://athenaeumpub.com/journal-of-neuro-and-oncology-research/
13

82. Fang J, Guo Y, Tan S. Autologous endothelial progenitor cells transplantation for acute ischemic stroke: a 4-year follow-up
study. Stem Cells Transl Med. 2019;8(1):14-21.
83. Permana AT, Bajamal AH, Parenrengi MA, Suroto NS, Lestari P, Fauzi AA. Clinical outcome and safety of stem cell therapy
for ischemic stroke: A systematic review and meta-analysis. Surg Neurol Int. 2022;13:206.
84. Wu CX, Wang D, Cai Y, Luo AR, Sun H. Effect of autologous bone marrow stem cell therapy in patients with liver cirrhosis:
a meta-analysis. J Clin Transl Hepatol. 2019;7(3):238-48.
85. Walczak P, Kedziorek DA, Gilad AA, Lin S, Bulte JW. Instant MR labeling of stem cells using magneto electroporation. Magn
Reson Med. 2005;54(4):769-74.
86. Kalish H, Arbab AS, Miller BR. Combination of transfection agents and magnetic resonance contrast agents for cellular
imaging: relationship between relaxivities, electrostatic forces and chemical composition. Magn Reson Med. 2003;50(2):275-
82.
87. Gaudet JM, Ribot EJ, Chen Y, Gilbert KM, Foster PJ. Tracking the fate of stem cell implants with fluorine-19 MRI. PLoS One.
2015;10(3):e0118544.
88. Ashmore-Harris C, Iafrate M, Saleem A, Fruhwirth GO. Non-invasive reporter gene imaging of cell therapies, including t
cells and stem cells. Mol Ther. 2020;28(6):1392-416.
89. Mathiasen AB, Kastrup J. Non-invasive in-vivo imaging of stem cells after transplantation in cardiovascular tissue.
Theranostics. 2013;3(8):561-72.
90. Xu B, Zhang Y, Du XF. Neurons secrete miR-132-containing exosomes to regulate brain vascular integrity. Cell Res.
2017;27(7):882-97.
91. Otero-Ortega L, Laso-García F, Gómez-de Frutos MD. White Matter repair after extracellular vesicles administration in an
experimental animal model of subcortical stroke. Sci Rep. 2017;7:44433.
92. Xiong Y, Mahmood A, Chopp M. Emerging potential of exosomes for treatment of traumatic brain injury. Neural Regen Res.
2017;12(1):19-22.
93. Webb RL, Kaiser EE, Scoville SL. Human neural stem cell extracellular vesicles improve tissue and functional recovery in
the murine thromboembolic stroke model. Transl Stroke Res. 2018;9(5):530-9.
94. Birenbaum D, Bancroft LW, Felsberg GJ. Imaging in acute stroke. West J Emerg Med. 2011;12(1):67-76.
95. Chalela JA, Kidwell CS, Nentwich LM. Magnetic resonance imaging and computed tomography in emergency assessment
of patients with suspected acute stroke: a prospective comparison. Lancet. 2007;369(9558):293-8.
96. Muir KW, Buchan A, von Kummer R, Rother J, Baron JC. Imaging of acute stroke. Lancet Neurol. 2006;5(9):755-68.
97. Yin KJ, Hamblin M, Chen YE. Angiogenesis-regulating microRNAs and ischemic stroke. Curr Vasc Pharmacol.
2015;13(3):352-65.
98. Teng H, Zhang ZG, Wang L. Coupling of angiogenesis and neurogenesis in cultured endothelial cells and neural progenitor
cells after stroke. J Cereb Blood Flow Metab. 2008;28(4):764-71.
99. Hoehn M, Küstermann E, Blunk J. Monitoring of implanted stem cell migration in-vivo: a highly resolved in-vivo magnetic
resonance imaging investigation of experimental stroke in rat. Proc Natl Acad Sci U S A. 2002;99(25):16267-72.
100. Webb RL, Kaiser EE, Jurgielewicz BJ. Human neural stem cell extracellular vesicles improve recovery in a porcine model of
ischemic stroke. Stroke. 2018;49(5):1248-56.
101. Jiang Y, Zhu W, Zhu J, Wu L, Xu G, Liu X. Feasibility of delivering mesenchymal stem cells via catheter to the proximal end
of the lesion artery in patients with stroke in the territory of the middle cerebral artery. Cell Transplant. 2013;22(12):2291-8.
102. Singh M, Pandey PK, Bhasin A, Padma MV, Mohanty S. Application of stem cells in stroke: a multifactorial approach. Front
Neurosci. 2020;14:473.
103. Incontri Abraham D, Gonzales M, Ibarra A, Borlongan CV. Stand alone or join forces? Stem cell therapy for stroke. Expert
Opin Biol Ther. 2019;19(1):25-33.
104. Carrero R, Cerrada I, Lledó E. IL1β induces mesenchymal stem cells migration and leucocyte chemotaxis through NF-κB.
Stem Cell Rev Rep. 2012;8(3):905-16.
105. Bohaciakova D, Hruska-Plochan M, Tsunemoto R. A scalable solution for isolating human multipotent clinical-grade neural
stem cells from ES precursors. Stem Cell Res Ther. 2019;10(1):83.
106. Rust R, Weber RZ, Generali M. Xeno-free induced pluripotent stem cell-derived neural progenitor cells for in-vivo
applications. J Transl Med. 2022;20(1):421.

https://doi.org/10.46889/JNOR.2023.3102 https://athenaeumpub.com/journal-of-neuro-and-oncology-research/
14

107. Barak M, Fedorova V, Pospisilova V. Human iPSC-derived neural models for studying Alzheimer’s disease: from neural
stem cells to cerebral organoids. Stem Cell Rev Rep. 2022;18(2):792-820.
108. Aked J, Delavaran H, Lindvall O, Norrving B, Kokaia Z, Lindgren A. Attitudes to stem cell therapy among ischemic stroke
survivors in the lund stroke recovery study. Stem Cells Dev. 2017;26(8):566-72.
109. Unsworth DJ, Mathias JL, Dorstyn DS, Koblar SA. Are patient educational resources effective at deterring stroke survivors
from considering experimental stem cell treatments? A randomized controlled trial. Patient Educ Couns. 2020;103(7):1373-
81.
110. Unsworth DJ, Mathias JL, Dorstyn DS, Koblar SA. Stroke survivor attitudes toward and motivations for, considering
experimental stem cell treatments. Disabil Rehabil. 2020;42(8):1122-30.

Publish your work in this journal


Journal of Neuro and Oncology Research is an international, peer-reviewed, open access journal publishing original research, reports, editorials, reviews
and commentaries. All aspects of neurology and oncology health maintenance, preventative measures and disease treatment interventions are addressed
within the journal. Neurologist or oncologist and other researchers are invited to submit their work in the journal. The manuscript submission system is
online and journal follows a fair peer-review practices.

Submit your manuscript here: https://athenaeumpub.com/submit-manuscript/

https://doi.org/10.46889/JNOR.2023.3102 https://athenaeumpub.com/journal-of-neuro-and-oncology-research/

You might also like