Next Article in Journal
Endoscopic Ultrasound-Guided Biliary Drainage
Previous Article in Journal
Artificial Intelligence in Cardiology: Why So Many Great Promises and Expectations, but Still a Limited Clinical Impact?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Comparison of the Administration Route of Stem Cell Therapy for Ischemic Stroke: A Systematic Review and Meta-Analysis of the Clinical Outcomes and Safety

by
Asra Al Fauzi
1,*,
Ahmad Muslim Hidayat Thamrin
1,
Andhika Tomy Permana
1,
I. G. M. Aswin R. Ranuh
1,
Hanik Badriyah Hidayati
2,
Muhammad Hamdan
2,
Joni Wahyuhadi
1,
Nur Setiawan Suroto
1,
Pudji Lestari
3 and
Poodipedi Sarat Chandra
4
1
Department of Neurosurgery, Faculty of Medicine, Universitas Airlangga, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
2
Department of Neurology, Faculty of Medicine, Universitas Airlangga, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
3
Department of Public Health and Preventive Medicine, Faculty of Medicine, Universitas Airlangga, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
4
Department of Neurosurgery, All India Institute of Medical Sciences, New Delhi 110608, India
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2023, 12(7), 2735; https://doi.org/10.3390/jcm12072735
Submission received: 13 February 2023 / Revised: 6 March 2023 / Accepted: 4 April 2023 / Published: 6 April 2023
(This article belongs to the Section Pharmacology)

Abstract

:
Stem cell treatment is emerging as an appealing alternative for stroke patients, but there still needs to be an agreement on the protocols in place, including the route of administration. This systematic review aimed to assess the efficacy and safety of the administration routes of stem cell treatment for ischemic stroke. A systematic review was performed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines. A comprehensive literature search was undertaken using the PubMed, Scopus, and Cochrane databases. A total of 21 publications on stem cell therapy for ischemic stroke were included. Efficacy outcomes were measured using the National Institutes of Health Stroke Scale (NIHSS), the modified Rankin Scale (mRS), and the Barthel index (BI). Intracerebral administration showed a better outcome than other routes, but a greater number of adverse events followed due to its invasiveness. Adverse events were shown to be related to the natural history of stroke not to the treatment. However, further investigation is required, since studies have yet to compare the different administration methods directly.

1. Introduction

Stroke, one of the top causes of death worldwide, continues to be a significant costly drain on global health resources [1]. Stroke is estimated to be responsible for around 140,000 fatalities yearly in the United States [2]. Most strokes are ischemic in nature, representing about 87% of all instances in the US, making it the main focus of stroke research [2]. Even though 80% of stroke patients survive for one year after an incident, more than 10% of patients have long-term disabilities [3].
The increase in survival and reduction in sequelae following ischemic stroke may be partially explained by the acute-phase delivery of thrombolytic therapies [4]. The only approved therapy for acute stroke is intravenous recombinant tissue Plasminogen Activator (tPA), which has a limited time window of only 4.5 h. Additionally, there is a strict patient criteria for receiving urgent endovascular therapy and the benefits are uncertain [5]. Due to this failure to achieve the anticipated outcomes, it is only natural that new therapeutic tactics with a longer time frame and less invasive approaches are urgently needed.
Numerous studies have demonstrated the efficacy of stem cell treatment in restoring functional ability in stroke patients [6]. Aside from its potential to stimulate endogenous reparative mechanisms without replacing the injured cerebral tissue [7], it has also been shown to promote immunomodulation and neuronal, vascular, and glial remodeling. Intravenous injection is one option for the administration of neurotrophic substances, including vascular endothelial growth factor (VEGF), insulin-like growth factor (IGF), and brain-derived neurotrophic factor [8]. The type and source of stem cell to be administered (e.g., mesenchymal stem cell, bone marrow mononuclear cell, or neural stem cell), the route of administration (e.g., intravenous, intra-arterial, or intracerebral), and the time interval between stroke onset and administration (days to months), are all said to explain the varying results [9]. Clinical trials of stem cell therapy in stroke patients have demonstrated that the therapy is feasible, safe, and promotes recovery in ischemic strokes [10]. Since only a few studies have examined this treatment’s clinical outcome, effectiveness, and safety, as related to its route of administration, this review is mainly to determine the treatment’s clinical benefits and adverse events according to the treatment’s safety [11,12,13,14].

2. Materials and Methods

2.1. Eligibility Criteria

This review includes full-text cohort studies and clinical trials on adult ischemic stroke patients (acute, subacute, or chronic) who received stem cell therapy via intracerebral, intraventricular, subarachnoid, intra-arterial, intravenous, intraperitoneal, or intranasal administration. Reviews, unpublished articles, letters to the editor, abstracts, and studies not written in English were excluded.

2.2. Type of Outcome Measurements

Clinical outcomes were measured using the modified Rankin scale (mRS), the National Institute of Health Stroke Scale (NIHSS), and the Barthel index (BI). The adverse events (AE) and severe adverse events (SAE) of stem cell administration routes were analyzed to identify the safety of each treatment.

2.3. Search Methods and Identification of Studies

2.3.1. Information Sources

This systematic review was conducted based on Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines [15]. The articles, which were published between 2010 and 2022, were acquired by searching the PubMed, Scopus, Cochrane, and other electronic database sources (Google Scholar) on 31 March 2022. We applied language restrictions to our search so that only articles written in English were selected (Figure 1).

2.3.2. Search Protocol

The study questions were formed using the patient/population, intervention, comparison, outcomes, and study design (PICOS) model (Table 1). The following search string was used to search all trial registers and databases: stem cell therapy AND (ischemic stroke OR ischemic brain) AND (intracerebral OR intraventricular OR subarachnoid OR intra-arterial OR intravenous OR intraperitoneal OR intranasal) AND (functional outcome OR mRS OR NIHSS OR BI).

2.4. Data Collection and Analysis

The included studies were screened based on their titles and abstracts. Full-text articles, including RCTs and cohort studies, fulfilling the eligibility criteria were then assessed by each author. The details regarding the causes of exclusion were noted and reported. The included studies are summarized in Table 2.

2.5. Data Extraction and Management

Four authors independently extracted data, including each article’s patient characteristics, treatments, research quality, and therapeutic results. Details regarding the author, year of publication, study design, treatment details, and functional outcome based on the predetermined parameters were summarized for the qualitative analysis. We extracted the mean difference of outcome in each arm for continuous outcomes (mean difference of NIHSS, mRS, and BI after 6, 12, and 24 months). The four review authors entered all data into Review Manager (RevMan) software, version 5.4 [15]. Unfortunately, the literature search was carried out before we were registered to any of the systematic review registries.

2.6. Risk of Bias Analysis

Each author independently assessed the risk of bias in each study using Risk of Bias in Nonrandomized Intervention Studies (ROBINS-I) for nonrandomized studies and Risk of Bias 2 (RoB 2) for randomized studies [16]. The results of each interpreter’s assessment were then discussed by all of the authors. A risk-of-bias table and a summary of the bias were used. These showed normal distribution results with some acceptable deviations, and thus the eligibility of the literature was high [17,18].
Table 2. Summary of the Studies Included in the Systematic Review.
Table 2. Summary of the Studies Included in the Systematic Review.
Author(s)Study DesignAge (years)Stroke TerritorySample SizeType of GraftNumber of Transplanted CellsRoute of AdministrationFunctional OutcomeNotes
Baseline6 Months12 Months24 Months
NIHSSmRSBINIHSSmRSBINIHSSmRSBINIHSSmRSBI
Lee et al. (2010) [19]RCT64.6 ± 13.652 MCA36 controlAutologous bone marrow MSC50 × 106Intravenous10.63 ± 3.004.80 ± 0.50n/an/an/an/an/an/an/an/an/an/aImprovement in the early post-treatment phase, no improvement later on
16 treatment10.17 ± 3.604.40 ± 0.90n/an/an/an/an/an/an/an/an/an/a
Honmou et al. (2011) [20]Non-RCT60.5 (41–73)4 MCA
6 ICA
2 Lacunar
12 treatmentAutologous bone marrow-derived MSC120 (60–160) × 106Intravenous8.25 ± 5.55n/an/an/an/an/a1.58 ± 2.02n/an/an/an/an/aSignificant improvement
Bhasin et al. (2012) [21]Non-RCT46.58 ± 10.9924 MCA12 controlAutologous bone marrow–derived MSC55 (50–58) × 106Intravenousn/an/a49.92 ± 10.03n/an/a78.67 ± 11.35n/an/an/an/an/an/aSignificant improvement on modified BI only
47.08 ± 9.9012 treatmentn/an/a48.75 ± 10.57n/an/a69.75 ± 9.90n/an/an/an/an/an/a
Prasad et al. (2012) [22]Non-RCT54 (38–70)11 MCA11 treatmentAutologous bone marrow-derived MSC40 × 106Intravenous12.27 ± 5.163.45 ± 1.0434.09 ± 22.234.80 ± 5.472.09 ± 1.3079.09 ± 20.23n/an/an/an/an/an/aSignificant neurological outcome in the subacute stroke
Jiang et al. (2013) [23]Prospective cohort48.5 (40–59)4 MCA4 treatmentUmbilical cord-derived MSC20 × 106Intra-arterialn/a4.00 ± 0.816n/an/a3.25 ± 0.50n/an/an/an/an/an/an/aSmall size of samples; significant improvement
Banerjee et al. (2014) [24]Non-RCT57 (45–75)4 MCA
1 PCA
5 treatmentCD34 ± stem cell2.42 (1.2–2.79) × 106Intra-arterial10.40± 5.133.80 ± 0.84n/a2.20 ± 1.921.60 ± 1.14n/an/an/an/an/an/an/aSignificant improvement
Chen et al. (2014) [25]RCT52.8 ± 9.030 MCA15 controlPBSC(3–8) × 106Intracerebral9.60 ± 1.302.80 ± 0.40n/a9.40 ± 1.202.70 ± 0.50n/a8.70 ± 1.902.70 ± 0.50n/an/an/an/aSignificant clinical outcome improvement
50.1 ± 7.715 treatment9.30 ± 0.502.90 ± 0.30n/a6.70 ± 1.702.50 ± 0.50n/a5.50 ± 1.802.10 ± 0.30n/an/an/an/a
Prasad et al. (2014) [26]RCT18–7560 controlAutologous bone marrow mononuclear stem cell280.75 × 106Intravenous11.00 ± 4.44n/an/an/an/an/an/an/an/an/an/an/aInsignificant results
108 MCA
5 ACA
7 MCA+ACA
60 treatment13.00 ± 4.44n/an/an/an/an/an/an/an/an/an/an/a
Qiao et al. (2014) [27]Prospective cohort61.5 (45–85)5 MCA
1 ACA
6 treatmentUmbilical cord mesenchymal stem cell0.5 × 106/kgbwIntravenous8.17 ± 5.844.00 ± 1.1040.83 ± 33.38n/an/an/an/an/an/an/an/an/aImproved neurological function
Kalladka et al. (2016) [28]Non-RCT78 (68–82)9 MCA
1 MCA+ACA
1 PCA
3 (2 m) *Human neural stem cell (CTX0E03)2 × 106Intracerebral7.67 ± 1.534.00 ± 0.0011.00 ± 1.005.67 ± 2.08n/an/a4.33 ± 2.08n/a12.67 ± 1.155.67 ± 1.15n/a12.00 ± 1.73Improved neurological function; no controls
69 (61–75)3 (5 m) *5 × 1068.00 ± 2.003.67 ± 0.5811.67 ± 2.526.33 ± 3.06n/an/a6.33 ± 2.89n/a14.67 ± 2.085.67 ± 4.04n/a14.33 ± 3.51
64 (60–68)3 (10 m) *10 × 1067.33 ± 0.582.67 ± 0.5814.33 ± 1.534.33 ± 0.58n/an/a4.67 ± 1.15n/a14.67 ±3.214.00 ± 1.73n/a13.33 ± 1.53
66 (61–71)2 (20 m) *20 × 1066.50 ± 0.713.00 ± 0.0013.50 ± 2.123.50 ± 2.12n/an/a3.50 ± 3.53n/a16 ± 2.834.00 ± 0.00n/a17.5 ± 3.53
Hess et al. (2017) [29]RCT18–33n/a63 controlMultipotent adult progenitor cell(400–1200) × 106Intravenous13.40 ± 3.70n/an/an/an/an/an/an/an/an/an/an/aInsignificant results
71 treatment13.30 ± 3.50n/an/an/an/an/an/an/an/an/an/an/a
Jin et al. (2017) [30]RCT53.10 ± 13.0713 anterior circulation
7 posterior circulation
10 controlAutologous bone marrow–derived MSC10 × 106Intracerebral10.70 ± 3.714.10 ± 0.9915.00 ± 8.508.20 ± 3.493.90 ± 1.1029.00 ± 12.876.50 ± 3.343.40 ± 0.9741.5 ± 17.655.70 ± 3.123.10 ± 1.1047.00 ± 24.06Lumbar subarachnoid injection; significant neurological improvement
50.80 ± 17.4310 treatment12.30 ± 3.954.60 ± 0.7014.50 ± 13.019.40 ± 3.814.00 ± 0.8226.00 ± 16.808.80 ± 3.713.60 ± 0.7037.5 ± 15.868.60 ± 3.693.00 ± 1.33351.50 ± 26.15
Laskowitz et al. (2018) [31]Non-RCT65.5 (45–79)10 MCA10 treatmentUmbilical cord blood stem cell1680 (840–2920) × 106Intravenous11.20 ± 1.624.40 ± 0.5218.80 ± 12.26n/an/an/an/an/an/an/an/an/aSignificant improvement
Jailllard et al. (2019) [12]RCT53n/a15 controlAutologous bone marrow–derived MSC(100–300) × 106Intravenous12.75 ± 1.504.00 ± 0.0042.50 ± 14.519.40 ± 4.703.00 ± 0.6677.86 ± 25.40n/an/an/a8.43 ± 4.963.07 ± 1.1085.00 ± 20.48Insignificant clinical outcome, except for the motoric score
16 treatment13.5 ± 2.463.875 ± 0.1645.00 ± 18.828.94 ± 5.203.00 ± 0.6380.63 ± 30.87n/an/an/a7.73 ± 5.782.75 ± 0.9382.00 ± 27.83
Savitz et al. (2019) [14]RCT60.7 ± 10.4n/a19 controlAutologous bone marrow–derived ALD-4013.8 × 106Intra-arterialn/an/an/an/an/an/an/an/an/an/an/an/aNo significant improvement of the neurofunctional outcome between groups
20 treatment
Steinberg et al. (2019) [3]Non-RCT64 (33–75)n/a6 (2.5 m) *Modified bone marrow MSC (SB623)2.5 × 106Intracerebral9.30 ± 1.703.22 ± 0.43n/an/an/an/an/an/an/an/an/an/aSignificant improvement of NIHSS score;
insignificant result of mRS
6 (5 m) *5 × 106
6 (10 m) *10 × 106
Vahidy et al. (2019) [13]Non-RCT63.7 ± 12.5n/a185 controlAutologous bone marrow–derived MSC10 × 106/kgbwIntravenousn/a0.40 ± 0.85n/an/an/an/an/an/an/an/an/an/aFavorable safety
60.7 ± 13.325 treatmentn/a0.08 ± 0.40n/an/an/an/an/an/an/an/an/an/a
Zhang et al. (2019) [11]Prospective cohort42 (30–49)n/a3 (12 m) *Neural stem cell (NSI-566)12 × 106Intracerebral5.33 ± 3.51n/an/an/an/an/an/an/an/an/an/an/aSignificant improvement; imaging revealed new neural tissue formation
43 (41–45)3 (24 m) *24 × 1067.67 ± 2.08n/an/an/an/an/an/an/an/an/an/an/a
48 (37–54)3 (72 m) *72 × 1066.00 ± 1.00n/an/an/an/an/an/an/an/an/an/an/a
Chung et al. (2021) [32]RCT64.27 ± 13.25n/a15 controlAutologous bone marrow–derived MSC1 × 106/kgbwIntravenous14.47 ± 5.324.47 ± 0.8319.80 ± 25.5n/an/an/an/an/an/an/an/an/aInsignificant overall results;
significant improvement in lower extremity motor function
63.03 ± 14.3639 treatment11.36 ± 5.204.26 ± 0.7528.28 ± 26.63n/an/an/an/an/an/an/an/an/a
Kang Law et al. (2021) [33]RCT64.0 ± 13.917 MCA9 controlAutologous bone marrow–derived MSC2 × 106/kgbwIntravenousn/an/an/an/an/an/an/an/an/an/an/an/aSignificant BI improvement compared with the control group
54.60 ± 13.28 treatmentn/an/an/an/an/an/an/an/an/an/an/an/a
Ruiz et al. (2022) [34]RCT76 (69–80)19 MCA10
control
Adipose-derived MSC1 × 106/kgbwIntravenousn/an/an/an/an/an/an/an/an/an/an/an/aNo significant neurological improvement between the treatment groups
78 (70–82)9 treatmentn/an/an/an/an/an/an/an/an/an/an/an/a
RCT, randomized controlled trial; MSC, mesenchymal stem cell; PBSC, peripheral blood stem cell; NIHSS, National Institutes of Health Stroke Scale; mRS, modified Rankin scale; BI, Barthel index; * in a million cells.

3. Results

A total of 175 studies were identified and screened. Of these, thirty-three were assessed for eligibility and twenty-one (eleven PubMed studies, seven Scopus studies, and three Google Scholar studies) were included in the meta-analysis (Table 2).

3.1. Demographics, Timing of Intervention, Territory of Stroke, and Type of Stem Cell

The included studies were conducted in Asia, the United States, and Europe. Most of the studies were from Asia (12 out of 21; 57.1%), mainly from China (5 out of 21; 23.8%). Five (23.8%) studies were conducted in the United States, while the studies in Europe (5 out of 21; 23.8%) were undertaken primarily in the United Kingdom (3 out of 21; 14.2%).
Across the 21 studies, 836 patients were included, with a median age of 60.6 years (range 30–85 years). The stem cell therapy group consisted of 406 patients; 247 (60.83%) participants were male and 159 (39.16%) were female. Two of twenty-one studies defined intravascular tPA and endovascular thrombectomy as the intervention for the participants [14,29]. In contrast, other studies described a general supportive therapy for stroke (e.g., antiplatelet, antihypertensive, and rehabilitation) [12,25,30]. One study described the premedications given before stem cell administration (diphenhydramine, hydrocortisone, acetaminophen) but not specifically for the stroke [31].
A total of 390 comorbidities were reported. Hypertension was the most common, in 118 (29.06%) out of the 390 participants; 80 (19.7%) participants were smokers; and diabetes was reported in 68 (16.74%) participants, dyslipidemia in 70 (17.24%), and cardiac problems in 54 (13.3%).
The duration from the onset of stroke and stem cell administration was reported in 333 (82.02%) out of the 406 participants. Most participants (35.22%) had stem cell therapy in the subacute phase, followed by chronic stroke, with 108 (26.6%) participants, while the remaining had acute stroke (20.2%).
The area of vascularization of strokes was reported for 339 (83.49%) out of 406 participants. Most strokes had middle cerebral artery (MCA) involvement (87.6%), followed by anterior cerebral artery (ACA) and MCA involvement (2.4%), while posterior circulation and ACA strokes each occurred in 2.09% and 1.8%, respectively.
Six different stem cell types and sources were administered, namely bone marrow, peripheral blood, umbilical blood, adult progenitor cells, and human neural stem cells. Most of the participants had bone marrow-derived stem cell therapy (265 participants, 65.27%), followed by multipotent stem cells (71, 17.49%), and neural-derived peripheral and umbilical blood-derived stem cells were each administered to 20 (4.92%) participants. In the most recent study, adipose-derived stem cells were used in nine (2.21%) participants (Table 3).

3.2. Route of Administration: Clinical Outcomes and Adverse Events

The intravenous group were treated with the most used route of administration and consisted of 304 (78.87%) participants. The intracerebral group consisted of 64 (15.76%) participants, while the intra-arterial group consisted of 38 (9.35%) participants.

3.2.1. Clinical Outcome

The clinical outcome data that we extracted were 6 months (6 of 21 studies), 12 months (4 of 21 studies), and 24 months NIHSS (3 of 21 studies); 6 months, 12 months, and 24 months mRS (2 of 21 studies each); 6 months, 12 months, and 24 months BI (4 of 21, 2 of 21, and 3 of 21 studies, respectively). The mean baseline NIHSS score showed wide variation, namely 9.03 ± 1.76 in the intracerebral group, 10.4 ± 5.13 in the intra-arterial group, and 12.20 ± 4.10 in the intravenous group. We failed to extract some of the data due to the reported measures in some articles only being reported as median values. Baseline mRS scores were not used as a variable, with 3.36 ± 0.38 in the intracerebral group, 3.89 ± 0.83 in the intra-arterial group, and 3.30 ± 0.65 in the intravenous group. The baseline BI score was 13.48 ± 7.12 in the intracerebral group, with no available data for processing in the intra-arterial group, and 34.21 ± 21.64 in the intravenous group.
The NIHSS clinical outcomes after 6 months showed a decrease of 6.96 ± 2.36 in the intracerebral group, 2.2 ± 1.92 in the intra-arterial group, and 7.25 ± 5.31 in the intravenous group. The mRS score after 6 months also decreased by 3.34 ± 0.63 in the intracerebral group, 2.33 ± 0.86 in the intra-arterial group, and 2.63 ± 0.90 in the intravenous group. The improvement in BI score was marked by an increase after 6 months, with 26 ± 16.80 in the intracerebral group and 76.85 ± 21.41 in the intravenous group, while there were no available data in the intra-arterial group.
There was a tendency of decreasing NIHSS and mRS scores and increasing BI scores, indicating better clinical outcomes, after 12 and 24 months in the stem cell therapy group (Table 4).

3.2.2. Adverse Events (Based on Route of Administration)

AEs and SAEs were defined by the terms of the Common Terminology Criteria for Adverse Events (CTCAE) version 5.0. The grading of AEs and SAEs also followed CTCAE version 5.0. An SAE was defined as CTCAE grade 3 or more [35]. AEs were reported to have occurred 431 times in 21 studies, while SAEs were reported 101 (26.09%) times. The numbers varied across the different routes of administration.
AEs in the intravenous group occurred in 282 (65.42%) patients, with 68 (24.11%) classified as severe, comprising 23.05% of the reported patients with AEs in the intravenous group.
AEs in the intracerebral group occurred in 123 (28.53%) patients, with 28 (22.76%) considered severe, comprising 44.44% of the reported patients with AEs in the intracerebral group.
In the intra-arterial group, AEs occurred in 26 (6.03%) patients, with 5 (19.23%) classified as severe, accounting for 17.24% of the reported patients with AEs in the intra-arterial group. Table 5 and Table 6 provide a complete list of adverse occurrences and a summary of them categorized by route of administration.
Using risk ratio analysis, we compared the stem cell and control groups for AE and SAE risk. This analysis involved eight studies with available data. Hence, the subgroup analysis population was heterogenous (I2 = 72%). The results showed the stem cell groups had less risk of AE, although it was not statistically significant (RR = 0.99, 95% CI 0.89–1.09, p = 0.82, I2 = 72%) (Figure 2).
We also compared the stem cell and control groups for SAE risk. This analysis involved eight studies with available data. The results showed the stem cell groups had less risk of SAE, but it was not statistically significant (RR = 0.98, 95% CI 0.77–1.26, p = 0.90, I2 = 0%). The sub-group analysis population was homogenous (I2 = 0%) (Figure 3).
We also provide the results of the quality assessment which can be accessed in Supplementary Materials in order to help the reader in the analysis of the results of the manuscript.

4. Discussion

4.1. Demographics, Timing of Intervention, Territory of Stroke, and Type of Stem Cell

Men outnumbered women by a ratio of 1.65:1, given that men have a greater age-adjusted stroke incidence; however, women are known to have a higher risk of stroke due to their longer life expectancy [36,37]. These findings may have been affected by the choice of therapy for different genders. Some studies reported worse clinical outcomes with mechanical thrombectomy in women [38,39,40].
Hypertension, a risk factor for ischemic and hemorrhagic stroke [41], was the most prevalent comorbidity, followed by diabetes, dyslipidemia, and cardiac issues with a significant role on the stroke outcome [42].
By the time stem cell therapy was administered, most individuals had had subacute strokes. The subacute period of stroke is considered the ideal time for the brain to self-repair [14]. Chronic strokes, on the other hand, necessitate the opening of the blood–brain barrier (BBB) to facilitate a more successful treatment strategy. Previous research has shown that manipulating the BBB with IV mannitol prior to stem cell treatment results in an increase in trophic factors in the brain following infarction; however, the mRS score was not observed [32,43]. It should be noted that the increased permeability of the BBB could worsen the ischemic process through increased inflammatory factors at the injury site [44]. On the other hand, stem cell treatment for acute stroke has been shown to lower the inflammatory response, enhancing the tissue repair and neuroprotection processes [45,46].
According to the available data, the MCA accounts for the most stroke pathology [47]. Meanwhile, isolated ACA territory strokes are rare, reported to only account for 0.5–3% of all ischemic strokes [48,49]. Only 1.8% of patients in our research experienced an isolated ACA stroke, whereas 2.4% had both MCA and ACA stroke. Only two trials in this evaluation reported on patients who had received stem cell treatment for posterior circulation strokes [30]. Histological differences also play a role. The brainstem is composed more of white than gray matter [50], and microglia, along with the precursors of oligodendrocytes, are known to help secrete trophic factors [51].
Most studies utilized autologous bone marrow–derived stem cells [3,12,13,14,19,20,21,22,26,30,32,46]. The use of these stem cells to mend neural tissue was described with encouraging results [52,53,54,55,56] and reportedly showed only motoric improvement [12]; in addition, Prasad observed a slight improvement in outcomes [26]. Lee observed considerable improvement in the early post-therapeutic period but no improvement at later stages [19]. Kalladka displayed improved neurological functions, although no comparative controls were employed in that study [28].

4.2. Route of Administration: Clinical Outcome and Adverse Events

The route of administration is one of the most contentious issues, as it relates to the efficacy and safety of the procedure on patients [46,57]. This is discussed in more depth in our study.

4.2.1. Intravenous Route

Despite being the most frequently utilized approach in the literature analyzed in this investigation, only four out of eleven studies using the intravenous route for stem cell delivery reported favorable clinical results, and only one out of four was an RCT. The other three were not controlled studies, although they all revealed improved neurological outcomes following therapy for varying lengths of time. Apart from the trial by Lee et al. (2018), all previous RCTs failed to demonstrate substantially better neurological results [12,26,29,32].
The intravenous route is regarded as the most straightforward and the least invasive technique available [58], compared with its intravascular counterpart, the intra-arterial route. The most recent literature revealed that intravenous stem cell treatment for chronic ischemic stroke had unsatisfactory results [32].
On the other hand, intravenous stem cell treatment causes stem cells to migrate to organs other than the brain due to their systemic nature. Research has indicated that time is critical in stroke therapies, particularly for acute strokes [59], and the fact that brain tissues are very vulnerable to hypoxia [60]. Hess reported that the early administration of allogeneic multipotent adult stem cells (<36 h) in cases of acute stroke is associated with better clinical outcomes [29].
The impact of graft time and route on the survival and functional advantages of CD133+ human bone marrow cells in a stroke model might help personalize transplantation methods for individual instances [61]. Localized graft survival with improvement in motor deficits was observed in both immediate and delayed intracerebral transplantation; however, compared with graft survival, behavioral improvement was only observed in immediate intravenous transplantation [61].
As previously noted, stroke is accompanied by severe inflammation and an immune response early in the disease process; therefore, employing an invasive procedure in the acute stage may not be advised [62]. Consequently, in the early stage, pursuing intravenous transplantation, a minimally invasive treatment, is more advisable.

4.2.2. Intra-Arterial Route

One RCT trial conducted using the intra-arterial route was found to have unsatisfactory results [14], while the other two studies lacked controls [23,24].
Intra-arterial stem cell administration for stroke facilitates a smaller dose and more concentrated delivery of cells to the cerebral lesion [60] Savitz mentioned that the therapy’s dosage has no correlation with the neurofunctional outcome [14]. Moreover, intra-arterial administration requires a smaller number of cell grafts, compared with the intravenous route [63], and Zhang et al. (2018) reported better neurologic outcomes in the intra-arterial group compared with the intravenous and intracerebral routes using an animal model [58].
In general, stem cell delivery via the intra-arterial route has a mechanism of action similar to stem cell administration via the intravenous route [8]. The primary method of healing following stem cell administration via the intra-arterial route is via the stimulation of injected stem cell to produce growth factors, cytokines, and chemokines by paracrine stem cells that contribute to anti-apoptotic effects, angiogenesis, and neurogenesis [64].

4.2.3. Intracerebral Route

All five studies reported a better clinical outcome regarding intracerebral administration [25,30], and one reported insignificant improvement in the 24-month mRS, while there were improvements in the alternative scoring systems [3].
Intracerebral stem cell treatment in instances of cerebral ischemia provides the benefit of minimizing concern for biodistribution and focused migration of cells into infarcted neural tissue [58]. Additionally, it has the advantage of direct inoculation and targeted therapy to the infarcted cerebral tissue without the disadvantages of cell dispersion and ineffective localization associated with the intravenous route and microembolization and bubble formation associated with the intra-arterial route [65].
The mechanism of the action of stem cells administered via the intracerebral route may differ from the intravascular one, as it directs the stem cell delivery [61]. Since the survival rate and total number of new neurons are exceedingly low, the intracerebral route may provide neuronal healing. Since the BBB is disturbed in stroke, transplanted stem cells easily traverse the BBB, congregating in infarcted brain regions and re-establishing the BBB’s integrity [10,66,67,68,69].
Some reports demonstrated that the intracerebral route may decrease apoptosis in the ischemia border area and be related to remarkable neurological recovery in animal models [10,66,70]. It was reported that human bone marrow mesenchymal stem cell (hMSCs) could reduce the apoptosis in neuronal cell death on cerebral ischemia [68] by secreting a wide range of anti-inflammatory cytokines [67].
However, the invasive nature of this intracerebral delivery technique requires further attention and patient selection. Several studies reported procedure-related adverse events. Kalladka reported one symptomatic procedure-related extradural hematoma and one symptomatic anticoagulant-related subdural hemorrhage event [28]. Steinberg reported one asymptomatic procedure-related subdural hemorrhage event with good recovery [3]. Zhang reported one asymptomatic microcerebral hemorrhage without sequelae [58]. The intracerebral route can be performed using less invasive techniques, such as intraventricular and subarachnoid administration [69]. The subarachnoid route was reported in an RCT and had remarkable clinical outcomes in the experimental group [30].
To the authors’ knowledge, there is no research comparing the efficacy of various modalities of stem cell treatments for ischemic strokes in humans. However, Zhang et al. (2018) reported that the intra-arterial route had superior efficacy compared with other routes (intravenous and intracerebral) in an animal model of cerebral ischemia [58].
Other routes worth noting are intraperitoneal and intranasal administration [69]. These two routes still require further experimental studies to demonstrate their feasibility, safety, and effectiveness. The intranasal route could offer a less invasive approach and is promising as it bypasses the BBB [71].

4.2.4. Adverse Events

The majority of AEs (65.42%) occurred in the intravenous group, while 28.53% occurred in the intracerebral group and 6.03% in the intra-arterial group. The authors recognize that these data should be treated with caution, as they do not consider the number of participants and are only a rudimentary representation of all occurrences. As mentioned above, Laskowitz reported that most AE occurrence was not related to the treatment group (99.1%) [31]. Consistent with this, Vahidy also reported that there were no study-related SAEs in the therapy group [13].
The ratio of all reported SAEs to all reported AEs was 26.09%; however, this figure should be regarded cautiously because not all studies reported on all types of AE [19,28]. The intracerebral group had the highest rate of SAEs (44.44%), followed by the intravenous group (23.05%), while the intra-arterial group had the lowest rate of SAEs (17.24%). These findings might be due to the invasiveness of the intracerebral route and the limited sample in the intra-arterial group [28].
The comparison of AEs and SAEs between the stem cell and control groups was also studied using forest plot. Although not statistically significant, AEs and SAEs occurred less in the stem cell group. These findings might have been caused by patients’ comorbidities.

5. Conclusions

In conclusion, although stem cell treatment demonstrated superior results over standard conservative therapy alone in stroke patients, our data show that several factors (e.g., patient’s comorbidities, treatment’s timing, administration route) might blur the treatment’s benefits and safety. To the best of our knowledge, this review is the first study that determines the functional outcome and the treatment’s adverse events based on the delivery route. The findings of trials utilizing various delivery methods demonstrated positive effectiveness and safety. Although intracerebral injection resulted in better neurological outcomes than other routes, it was associated with a higher rate of AE because of its intrusive nature. On the other hand, the intra-arterial and intravenous routes had unsatisfactory outcomes but the highest degrees of safety, although the most AE occurrences were not related to the treatment protocols. Since we found that the studies’ outcomes and follow ups are both varied and limited in most of the included studies in this review, a more extensive and focused investigation is required to evaluate the effectiveness and safety of this future treatment strategy.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/jcm12072735/s1.

Author Contributions

Conceptualization, A.A.F. and P.S.C.; methodology, A.M.H.T. and A.T.P.; software, I.G.M.A.R.R.; validation, H.B.H., M.H., and J.W.; formal analysis, N.S.S.; investigation, P.L.; resources, A.T.P.; data curation, I.G.M.A.R.R.; writing—original draft preparation, A.M.H.T.; writing—review and editing, I.G.M.A.R.R.; visualization, A.M.H.T., I.G.M.A.R.R., and A.T.P.; supervision, A.A.F. and P.S.C.; project administration, A.A.F.; funding acquisition, A.A.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

The authors would like to thank all those who contributed to the conceptualization, production, and publication of this study.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bhatia, V.; Gupta, V.; Khurana, D.; Sharma, R.R.; Khandelwal, N. Randomized Assessment of the Safety and Efficacy of Intra-Arterial Infusion of Autologous Stem Cells in Subacute Ischemic Stroke. Am. J. Neuroradiol. 2018, 39, 899–904. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Barthels, D.; Das, H. Current Advances in Ischemic Stroke Research and Therapies. Biochim. Biophys. Acta Mol. Basis Dis. 2020, 1866, 165620. [Google Scholar] [CrossRef]
  3. Steinberg, G.K.; Kondziolka, D.; Wechsler, L.R.; Lunsford, L.D.; Kim, A.S.; Johnson, J.N.; Bates, D.; Poggio, G.; Case, C.; McGrogan, M.; et al. 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. 2019, 131, 1462–1472. [Google Scholar] [CrossRef] [PubMed]
  4. Wardlaw, J.M.; Murray, V.; Berge, E.; del Zoppo, G.J. Thrombolysis for Acute Ischaemic Stroke. Cochrane Database Syst. Rev. 2014, 2014, CD000213. [Google Scholar] [CrossRef] [PubMed]
  5. Powers, W.J.; Derdeyn, C.P.; Biller, J.; Coffey, C.S.; Hoh, B.L.; Jauch, E.C.; Johnston, K.C.; Johnston, S.C.; Khalessi, A.A.; Kidwell, C.S.; et al. 2015 American Heart Association/American Stroke Association Focused Update of the 2013 Guidelines for the Early Management of Patients With Acute Ischemic Stroke Regarding Endovascular Treatment: A Guideline for Healthcare Professionals From the American. Stroke 2015, 46, 3020–3035. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Boltze, J.; Ayata, C. Challenges and Controversies in Translational Stroke Research-an Introduction. Transl. Stroke Res. 2016, 7, 355–357. [Google Scholar] [CrossRef] [Green Version]
  7. Singh, M.; Pandey, P.K.; Bhasin, A.; Padma, M.V.; Mohanty, S. Application of Stem Cells in Stroke: A Multifactorial Approach. Front. Neurosci. 2020, 14, 437. [Google Scholar] [CrossRef]
  8. Detante, O.; Jaillard, A.; Moisan, A.; Barbieux, M.; Favre, I.M.; Garambois, K.; Hommel, M.; Remy, C. Biotherapies in Stroke. Rev. Neurol. 2014, 170, 779–798. [Google Scholar] [CrossRef]
  9. Janowski, M.; Wagner, D.-C.; Boltze, J. Stem Cell-Based Tissue Replacement After Stroke: Factual Necessity or Notorious Fiction? Stroke 2015, 46, 2354–2363. [Google Scholar] [CrossRef] [Green Version]
  10. Bang, O.Y. Clinical Trials of Adult Stem Cell Therapy in Patients with Ischemic Stroke. J. Clin. Neurol. 2016, 12, 14–20. [Google Scholar] [CrossRef] [Green Version]
  11. Zhang, G.; Li, Y.; Reuss, J.L.; Liu, N.; Wu, C.; Li, J.; Xu, S.; Wang, F.; Hazel, T.G.; Cunningham, M.; et al. Stable Intracerebral Transplantation of Neural Stem Cells for the Treatment of Paralysis Due to Ischemic Stroke. Stem Cells Transl. Med. 2019, 8, 999–1007. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Jaillard, A.; Hommel, M.; Moisan, A.; Zeffiro, T.A.; Favre-Wiki, I.M.; Barbieux-Guillot, M.; Vadot, W.; Marcel, S.; Lamalle, L.; Grand, S.; et al. Autologous Mesenchymal Stem Cells Improve Motor Recovery in Subacute Ischemic Stroke: A Randomized Clinical Trial. Transl. Stroke Res. 2020, 11, 910–923. [Google Scholar] [CrossRef] [PubMed]
  13. Vahidy, F.S.; Haque, M.E.; Rahbar, M.H.; Zhu, H.; Rowan, P.; Aisiku, I.P.; Lee, D.A.; Juneja, H.S.; Alderman, S.; Barreto, A.D.; et al. Intravenous Bone Marrow Mononuclear Cells for Acute Ischemic Stroke: Safety, Feasibility, and Effect Size from a Phase I Clinical Trial. Stem Cells 2019, 37, 1481–1491. [Google Scholar] [CrossRef]
  14. Savitz, S.I.; Yavagal, D.; Rappard, G.; Likosky, W.; Rutledge, N.; Graffagnino, C.; Alderazi, Y.; Elder, J.A.; Chen, P.R.; Budzik, R.F.; et al. 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, 192–205. [Google Scholar] [CrossRef]
  15. The Cochrane Collaboration Review Manager (RevMan); Version 5.4; John Wiley & Sons: Chichester, UK, 2020.
  16. Higgins, J.; Thomas, J.; Chandler, J.; Cumpston, M.; Li, T.; Page, M.; Welch, V. Cochrane Handbook for Systematic Reviews of Interventions; Version 6.1.; Higgins, J., Thomas, J., Chandler, J., Cumpston, M., Li, T., Page, M., Welch, V., Eds.; Wiley & Sons: Chichester, UK, 2020. [Google Scholar]
  17. Sterne, J.A.; Hernán, M.A.; Reeves, B.C.; Savović, J.; Berkman, N.D.; Viswanathan, M.; Henry, D.; Altman, D.G.; Ansari, M.T.; Boutron, I.; et al. ROBINS-I: A Tool for Assessing Risk of Bias in Non-Randomised Studies of Interventions. BMJ 2016, 355, i4919. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Lo, C.K.-L.; Mertz, D.; Loeb, M. Newcastle-Ottawa Scale: Comparing Reviewers’ to Authors’ Assessments. BMC Med. Res. Methodol. 2014, 14, 45. [Google Scholar] [CrossRef] [Green Version]
  19. Lee, J.S.; Hong, J.M.; Moon, G.J.; Lee, P.H.; Ahn, Y.H.; Bang, O.Y. A Long-Term Follow-Up Study of Intravenous Autologous Mesenchymal Stem Cell Transplantation in Patients With Ischemic Stroke. Stem Cells 2010, 28, 1099–1106. [Google Scholar] [CrossRef]
  20. Honmou, O.; Houkin, K.; Matsunaga, T.; Niitsu, Y.; Ishiai, S.; Onodera, R.; Waxman, S.G.; Kocsis, J.D. Intravenous Administration of Auto Serum-Expanded Autologous Mesenchymal Stem Cells in Stroke. Brain 2011, 134, 1790–1807. [Google Scholar] [CrossRef] [Green Version]
  21. Bhasin, A.; Srivastava, M.V.; Bhatia, R.; Mohanty, S.; Kumaran, S.S.; Bose, S. Autologous Intravenous Mononuclear Stem Cell Therapy in Chronic Ischemic Stroke. J. Stem Cells Regen. Med. 2012, 8, 181–189. [Google Scholar] [CrossRef]
  22. Prasad, K.; Mohanty, S.; Bhatia, R.; Srixivastava, M.V.P.; Garg, A.; Srivastava, A.; Goyal, V.; Tripathi, M.; Kumar, A.; Bal, C.; et al. Autologous Intravenous Bone Marrow Mononuclear Cell Therapy for Patients with Subacute Ischaemic Stroke: A Pilot Study. Indian J. Med. Res. 2012, 136, 221–228. [Google Scholar]
  23. 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 Transpl. 2013, 22, 2291–2298. [Google Scholar] [CrossRef]
  24. Banerjee, S.; Bentley, P.; Hamady, M.; Marley, S.; Davis, J.; Shlebak, A.; Nicholls, J.; Williamson, D.A.; Jensen, S.L.; Gordon, M.; et al. Intra-Arterial Immunoselected CD34+ Stem Cells for Acute Ischemic Stroke. Stem Cells Transl. Med. 2014, 3, 1322–1330. [Google Scholar] [CrossRef]
  25. Chen, D.C.; Lin, S.Z.; Fan, J.R.; Lin, C.H.; Lee, W.; Lin, C.C.; Liu, Y.J.; Tsai, C.H.; Chen, J.C.; Cho, D.Y.; et al. Intracerebral Implantation of Autologous Peripheral Blood Stem Cells in Stroke Patients: A Randomized Phase II Study. Cell Transpl. 2014, 23, 1599–1612. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Prasad, K.; Sharma, A.; Garg, A.; Mohanty, S.; Bhatnagar, S.; Johri, S.; Singh, K.K.; Nair, V.; Sarkar, R.S.; Gorthi, S.P.; et al. Intravenous Autologous Bone Marrow Mononuclear Stem Cell Therapy for Ischemic Stroke: A Multicentric, Randomized Trial. Stroke 2014, 45, 3618–3624. [Google Scholar] [CrossRef] [Green Version]
  27. Qiao, L.-Y.; Huang, F.-J.; Zhao, M.; Xie, J.-H.; Shi, J.; Wang, J.; Lin, X.-Z.; Zuo, H.; Wang, Y.-L.; Geng, T.-C. A Two-Year Follow-Up Study of Cotransplantation with Neural Stem/Progenitor Cells and Mesenchymal Stromal Cells in Ischemic Stroke Patients. Cell Transpl. 2014, 23, 65–72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Kalladka, D.; Sinden, J.; Pollock, K.; Haig, C.; McLean, J.; Smith, W.; McConnachie, A.; Santosh, C.; Bath, P.M.; Dunn, L.; et al. Human Neural Stem Cells in Patients with Chronic Ischaemic Stroke (PISCES): A Phase 1, First-in-Man Study. Lancet 2016, 388, 787–796. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Hess, D.C.; Wechsler, L.R.; Clark, W.M.; Savitz, S.I.; Ford, G.A.; Chiu, D.; Yavagal, D.R.; Uchino, K.; Liebeskind, D.S.; Auchus, A.P.; et al. Safety and Efficacy of Multipotent Adult Progenitor Cells in Acute Ischaemic Stroke (MASTERS): A Randomised, Double-Blind, Placebo-Controlled, Phase 2 Trial. Lancet Neurol. 2017, 16, 360–368. [Google Scholar] [CrossRef] [PubMed]
  30. Jin, Y.; Ying, L.; Yu, G.; Nan, G. Analysis of the Long-Term Effect of Bone Marrow Mononuclear Cell Transplantation for the Treatment of Cerebral Infarction. Int. J. Clin. Exp. Med. 2017, 10, 3059–3068. [Google Scholar]
  31. Laskowitz, D.T.; Bennett, E.R.; Durham, R.J.; Volpi, J.J.; Wiese, J.R.; Frankel, M.; Shpall, E.; Wilson, J.M.; Troy, J.; Kurtzberg, J. Allogeneic Umbilical Cord Blood Infusion for Adults with Ischemic Stroke: Clinical Outcomes from a Phase I Safety Study. Stem Cells Transl. Med. 2018, 7, 521–529. [Google Scholar] [CrossRef]
  32. Chung, J.-W.; Chang, W.H.; Bang, O.Y.; Moon, G.J.; Kim, S.J.; Kim, S.-K.; Lee, J.S.; Sohn, S.-I.; Kim, Y.-H. Efficacy and Safety of Intravenous Mesenchymal Stem Cells for Ischemic Stroke. Neurology 2021, 96, e1012–e1023. [Google Scholar] [CrossRef]
  33. Law, Z.K.; Tan, H.J.; Chin, S.P.; Wong, C.Y.; Wan Yahya, W.N.N.; Muda, A.S.; Zakaria, R.; Ariff, M.I.; Ismail, N.A.; Cheong, S.K.; et al. 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, 833–840. [Google Scholar] [CrossRef] [PubMed]
  34. de Celis-Ruiz, E.; Fuentes, B.; Alonso de Leciñana, M.; Gutiérrez-Fernández, M.; Borobia, A.M.; Gutiérrez-Zúñiga, R.; Ruiz-Ares, G.; Otero-Ortega, L.; Laso-García, F.; Gómez-de Frutos, M.C.; et al. Final Results of Allogeneic Adipose Tissue–Derived Mesenchymal Stem Cells in Acute Ischemic Stroke (AMASCIS): A Phase II, Randomized, Double-Blind, Placebo-Controlled, Single-Center, Pilot Clinical Trial. Cell Transpl. 2022, 31, 096368972210838. [Google Scholar] [CrossRef] [PubMed]
  35. Cancer Therapy Evaluation Program (CTEP). Common Terminology Criteria for Adverse Events (CTCAE); v.5.0 [5x7]; Cancer Therapy Evaluation Program (CTEP); 2017; p. 155. Available online: https://ctep.cancer.gov/protocolDevelopment/electronic_applications/ctc.htm#ctc_50 (accessed on 12 February 2023).
  36. De Silva, D.A.; Ebinger, M.; Davis, S.M. Gender Issues in Acute Stroke Thrombolysis. J. Clin. Neurosci. 2009, 16, 501–504. [Google Scholar] [CrossRef]
  37. Reeves, M.J.; Bushnell, C.D.; Howard, G.; Gargano, J.W.; Duncan, P.W.; Lynch, G.; Khatiwoda, A.; Lisabeth, L. Sex Differences in Stroke: Epidemiology, Clinical Presentation, Medical Care, and Outcomes. Lancet Neurol. 2008, 7, 915–926. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Davison, M.A.; Ouyang, B.; Keppetipola, K.M.; Chen, M. Arterial Diameter and the Gender Disparity in Stroke Thrombectomy Outcomes. J. Neurointerv. Surg. 2018, 10, 949–952. [Google Scholar] [CrossRef]
  39. Lorenzano, S.; Ahmed, N.; Falcou, A.; Mikulik, R.; Tatlisumak, T.; Roffe, C.; Wahlgren, N.; Toni, D. Does Sex Influence the Response to Intravenous Thrombolysis in Ischemic Stroke? Stroke 2013, 44, 3401–3406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Meseguer, E.; Mazighi, M.; Labreuche, J.; Arnaiz, C.; Cabrejo, L.; Slaoui, T.; Guidoux, C.; Olivot, J.-M.; Abboud, H.; Lapergue, B.; et al. Outcomes of Intravenous Recombinant Tissue Plasminogen Activator Therapy According to Gender. Stroke 2009, 40, 2104–2110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Boehme, A.K.; Esenwa, C.; Elkind, M.S.V. Stroke Risk Factors, Genetics, and Prevention. Circ. Res. 2017, 120, 472–495. [Google Scholar] [CrossRef]
  42. Roger, V.L.; Go, A.S.; Lloyd-Jones, D.M.; Benjamin, E.J.; Berry, J.D.; Borden, W.B.; Bravata, D.M.; Dai, S.; Ford, E.S.; Fox, C.S.; et al. Executive Summary: Heart Disease and Stroke Statistics—2012 Update. Circulation 2012, 125, 188–197. [Google Scholar] [CrossRef]
  43. Borlongan, C.V.; Hadman, M.; Davis Sanberg, C.; Sanberg, P.R. Central Nervous System Entry of Peripherally Injected Umbilical Cord Blood Cells Is Not Required for Neuroprotection in Stroke. Stroke 2004, 35, 2385–2389. [Google Scholar] [CrossRef] [Green Version]
  44. Borlongan, C.V.; Glover, L.E.; Sanberg, P.R.; Hess, D.C. Permeating the Blood Brain Barrier and Abrogating the Inflammation in Stroke: Implications for Stroke Therapy. Curr. Pharm. Des. 2012, 18, 3670–3676. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Savitz, S.I. Cell Therapies: Careful Translation From Animals to Patients. Stroke 2013, 44, S107–S109. [Google Scholar] [CrossRef] [Green Version]
  46. Permana, A.T.; Bajamal, A.H.; Parenrengi, M.A.; Suroto, N.S.; Lestari, P.; Fauzi, A.A. Clinical Outcome and Safety of Stem Cell Therapy for Ischemic Stroke: A Systematic Review and Meta-Analysis. Surg. Neurol. Int. 2022, 13, 206. [Google Scholar] [CrossRef]
  47. Nogles, T.; Galuska, M. Middle Cerebral Artery Stroke; StatPearls Publishing: Treasure Island, FL, USA, 2023. [Google Scholar]
  48. Kang, S.Y.; Kim, J.S. Anterior Cerebral Artery Infarction: Stroke Mechanism and Clinical-Imaging Study in 100 Patients. Neurology 2008, 70, 2386–2393. [Google Scholar] [CrossRef] [PubMed]
  49. Sato, S.; Toyoda, K.; Matsuoka, H.; Okatsu, H.; Kasuya, J.; Takada, T.; Shimode, A.; Uehara, T.; Naritomi, H.; Minematsu, K. Isolated Anterior Cerebral Artery Territory Infarction: Dissection as an Etiological Mechanism. Cerebrovasc. Dis. 2010, 29, 170–177. [Google Scholar] [CrossRef] [PubMed]
  50. Fisher, M.; Feuerstein, G.; Howells, D.W.; Hurn, P.D.; Kent, T.A.; Savitz, S.I.; Lo, E.H. Update of the Stroke Therapy Academic Industry Roundtable Preclinical Recommendations. Stroke 2009, 40, 2244–2250. [Google Scholar] [CrossRef]
  51. Lloyd, A.F.; Miron, V.E. The Pro-Remyelination Properties of Microglia in the Central Nervous System. Nat. Rev. Neurol. 2019, 15, 447–458. [Google Scholar] [CrossRef]
  52. Zacharek, A.; Shehadah, A.; Chen, J.; Cui, X.; Roberts, C.; Lu, M.; Chopp, M. Comparison of Bone Marrow Stromal Cells Derived From Stroke and Normal Rats for Stroke Treatment. Stroke 2010, 41, 524–530. [Google Scholar] [CrossRef] [Green Version]
  53. Yang, B.; Xi, X.; Aronowski, J.; Savitz, S.I. Ischemic Stroke May Activate Bone Marrow Mononuclear Cells to Enhance Recovery After Stroke. Stem Cells Dev. 2012, 21, 3332–3340. [Google Scholar] [CrossRef] [Green Version]
  54. Fernandes, M.; Valente, S.G.; Sabongi, R.G.; Dos Santos, J.B.G.; Leite, V.M.; Ulrich, H.; Nery, A.A.; Da Silva Fernandes, M.J. Bone Marrow-Derived Mesenchymal Stem Cells versus Adipose-Derived Mesenchymal Stem Cells for Peripheral Nerve Regeneration. Neural Regen. Res. 2018, 13, 100–104. [Google Scholar] [CrossRef]
  55. Uccelli, A.; Laroni, A.; Freedman, M.S. Mesenchymal Stem Cells for the Treatment of Multiple Sclerosis and Other Neurological Diseases. Lancet Neurol. 2011, 10, 649–656. [Google Scholar] [CrossRef] [PubMed]
  56. Simard, A.R.; Rivest, S. Bone Marrow Stem Cells Have the Ability to Populate the Entire Central Nervous System into Fully Differentiated Parenchymal Microglia. FASEB J. 2004, 18, 998–1000. [Google Scholar] [CrossRef] [Green Version]
  57. Kawabori, M.; Shichinohe, H.; Kuroda, S.; Houkin, K. Clinical Trials of Stem Cell Therapy for Cerebral Ischemic Stroke. Int. J. Mol. Sci. 2020, 21, 7380. [Google Scholar] [CrossRef] [PubMed]
  58. Zhang, H.; Xie, X.; Xiong, Y.; Liu, S.; Hu, G.; Cao, W.; Wu, X. Comparisons of the Therapeutic Effects of Three Different Routes of Bone Marrow Mesenchymal Stem Cell Transplantation in Cerebral Ischemic Rats. Brain Res. 2018, 1680, 143–154. [Google Scholar] [CrossRef] [PubMed]
  59. Saver, J.L.; Goyal, M.; Van Der Lugt, A.; Menon, B.K.; Majoie, C.B.L.M.; Dippel, D.W.; Campbell, B.C.; Nogueira, R.G.; Demchuk, A.M.; Tomasello, A.; et al. Time to Treatment with Endovascular Thrombectomy and Outcomes from Ischemic Stroke: Ameta-Analysis. JAMA-J. Am. Med. Assoc. 2016, 316, 1279–1288. [Google Scholar] [CrossRef] [PubMed]
  60. Gutiérrez-Fernández, M.; Rodríguez-Frutos, B.; Álvarez-Grech, J.; Vallejo-Cremades, M.T.; Expósito-Alcaide, M.; Merino, J.; Roda, J.M.; Díez-Tejedor, E. Functional Recovery after Hematic Administration of Allogenic Mesenchymal Stem Cells in Acute Ischemic Stroke in Rats. Neuroscience 2011, 175, 394–405. [Google Scholar] [CrossRef]
  61. Borlongan, C.V.; Evans, A.; Yu, G.; Hess, D.C. Limitations of Intravenous Human Bone Marrow CD133+ Cell Grafts in Stroke Rats. Brain Res. 2005, 1048, 116–122. [Google Scholar] [CrossRef]
  62. Neal, E.G.; Liska, M.G.; Lippert, T.; Lin, R.; Gonzalez, M.; Russo, E.; Xu, K.; Ji, X.; Vale, F.L.; van Loveren, H.; et al. An Update on Intracerebral Stem Cell Grafts. Expert Rev. Neurother. 2018, 18, 557–572. [Google Scholar] [CrossRef]
  63. Ishizaka, S.; Horie, N.; Satoh, K.; Fukuda, Y.; Nishida, N.; Nagata, I. Intra-Arterial Cell Transplantation Provides Timing-Dependent Cell Distribution and Functional Recovery after Stroke. Stroke 2013, 44, 720–726. [Google Scholar] [CrossRef] [Green Version]
  64. Balber, A.E. Concise Review: Aldehyde Dehydrogenase Bright Stem and Progenitor Cell Populations from Normal Tissues: Characteristics, Activities, and Emerging Uses in Regenerative Medicine. Stem Cells 2011, 29, 570–575. [Google Scholar] [CrossRef]
  65. Argibay, B.; Trekker, J.; Himmelreich, U.; Beiras, A.; Topete, A.; Taboada, P.; Pérez-Mato, M.; Vieites-Prado, A.; Iglesias-Rey, R.; Rivas, J.; et al. Intraarterial Route Increases the Risk of Cerebral Lesions after Mesenchymal Cell Administration in Animal Model of Ischemia. Sci. Rep. 2017, 7, 40758. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Sun, J.; Huang, Y.; Gong, J.; Wang, J.; Fan, Y.; Cai, J.; Wang, Y.; Qiu, Y.; Wei, Y.; Xiong, C.; et al. Transplantation of HPSC-Derived Pericyte-like Cells Promotes Functional Recovery in Ischemic Stroke Mice. Nat. Commun. 2020, 11, 5196. [Google Scholar] [CrossRef] [PubMed]
  67. Boshuizen, M.C.S.; Steinberg, G.K. Stem Cell–Based Immunomodulation After Stroke. Stroke 2018, 49, 1563–1570. [Google Scholar] [CrossRef] [PubMed]
  68. Zhang, Y.; Yu, S.; Tuazon, J.P.; Lee, J.-Y.; Corey, S.; Kvederis, L.; Kingsbury, C.; Kaneko, Y.; Borlongan, C.V. Neuroprotective Effects of Human Bone Marrow Mesenchymal Stem Cells against Cerebral Ischemia Are Mediated in Part by an Anti-Apoptotic Mechanism. Neural Regen. Res. 2019, 14, 597–604. [Google Scholar] [CrossRef]
  69. Rodríguez-Frutos, B.; Otero-Ortega, L.; Gutiérrez-Fernández, M.; Fuentes, B.; Ramos-Cejudo, J.; Díez-Tejedor, E. Stem Cell Therapy and Administration Routes After Stroke. Transl. Stroke Res. 2016, 7, 378–387. [Google Scholar] [CrossRef]
  70. Stonesifer, C.; Corey, S.; Ghanekar, S.; Diamandis, Z.; Acosta, S.A.; Borlongan, C.V. Stem Cell Therapy for Abrogating Stroke-Induced Neuroinflammation and Relevant Secondary Cell Death Mechanisms. Prog. Neurobiol. 2017, 158, 94–131. [Google Scholar] [CrossRef]
  71. Lioutas, V.A.; Alfaro-Martinez, F.; Bedoya, F.; Chung, C.C.; Pimentel, D.A.; Novak, V. Intranasal Insulin and Insulin-Like Growth Factor 1 as Neuroprotectants in Acute Ischemic Stroke. Transl. Stroke Res. 2015, 6, 264–275. [Google Scholar] [CrossRef]
Figure 1. Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines flowchart.
Figure 1. Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines flowchart.
Jcm 12 02735 g001
Figure 2. Forest Plot for the Risk of Adverse Events in Stem Cell Therapy [12,14,19,21,26,29,30,32].
Figure 2. Forest Plot for the Risk of Adverse Events in Stem Cell Therapy [12,14,19,21,26,29,30,32].
Jcm 12 02735 g002
Figure 3. Forest Plot for the Risk of Severe Adverse Events in Stem Cell Therapy [12,14,19,21,26,29,30,32].
Figure 3. Forest Plot for the Risk of Severe Adverse Events in Stem Cell Therapy [12,14,19,21,26,29,30,32].
Jcm 12 02735 g003
Table 1. PICOS (Population, Intervention, Comparison, Outcome Measures, and Study Design) model.
Table 1. PICOS (Population, Intervention, Comparison, Outcome Measures, and Study Design) model.
PICOS ItemInclusion CriteriaExclusion Criteria
Population
  • Adult ischemic stroke patients receiving stem cell therapy
  • Stem cell therapy via intracerebral, intraventricular, subarachnoid, intra-arterial, intravenous, intraperitoneal, or intranasal administration
  • Child ischemic stroke patients
  • Traumatic brain injury
  • Hemorrhagic stroke
InterventionStem cell therapy via intracerebral administration
Comparison
  • Stem cell therapy via intraarterial administration
  • Stem cell therapy via intravenous administration
  • Control or sham group
Outcome measures
  • Clinical outcomes were measured using the modified Rankin scale (mRS)
  • The National Institute of Health Stroke Scale (NIHSS)
  • The Barthel index (BI). The adverse events (AE)
  • Severe adverse events (SAE)
Study design
  • Randomized clinical trials
  • Non-randomized clinical trial
  • Pilot randomized trials
  • Retrospective analyses
  • Reviews
  • Letters to the editor
  • Abstracts
Publication
  • Published in English
  • Access to full text
  • Unpublished studies
  • Study protocols
Table 3. Demographic Characteristics of the Studies.
Table 3. Demographic Characteristics of the Studies.
DescriptionNumber, n (%)
Total patientsn817 participants
Median age Median (min–max)60.6 years (30–85)
Gender
Malen247 participants (60.83%)
Femalen159 participants (39.16%)
Comorbidities
Hypertensionn118 participants (29.06%)
Diabetes mellitusn68 participants (16.74%)
Dyslipidemian70 participants (17.24%)
Cardiac problemsn54 participants (13.3%)
Smokingn80 participants (19.7%)
Standard treatment modalities
Tissue Plasminogen Activator (TPA)n2 studies
Mechanical revascularizationn2 studies
Patients with stem cell therapyn406 participants (49.7%)
Based on the route of administration
Intracerebral group (5 studies)n64 participants (15.76%)
Intra-arterial group (3 studies)n38 participants (9.35%)
Intravenous group (13 studies)n304 participants (74.87%)
Based on stroke onset
Acute (1–7 days)n82 participants (20.2%)
Subacute (1–3 weeks)n143 participants (35.22%)
Chronic (>3 weeks)n108 participants (26.6%)
Based on stem cell source
Bone marrown265 participants (65.27%)
Peripheral bloodn20 participants (4.92%)
Umbilical bloodn20 participants (4.92%)
Multipotent stem celln71 participants (17.49%)
Neural stem celln20 participants (4.92%)
Adiposen9 participants (2.21%)
Based on stroke territory
Anterior Cerebral Artery (ACA) and Middle Cerebral Artery (MCA)n8 participants (2.4%)
ACAn6 participants (1.8%)
MCAn297 participants (87.6%)
Anterior circulationn13 participants (3.8%)
Posterior circulationn7 participants (2.09%)
Table 4. Clinical Outcomes.
Table 4. Clinical Outcomes.
DescriptionNumber n (%)
Clinical outcome baseline
NIHSS
Intracerebral groupMean ± SD9.03 ± 1.76
Intra-arterial group Mean ± SD10.4 ± 5.13
Intravenous group Mean ± SD12.20 ± 4.10
mRS
Intracerebral groupMean ± SD3.36 ± 0.38
Intra-arterial group Mean ± SD3.89 ± 0.83
Intravenous group Mean ± SD3.30 ± 0.65
BI
Intracerebral groupMean ± SD13.48 ± 7.12
Intra-arterial group Mean ± SDn/a
Intravenous group Mean ± SD34.21 ± 21.64
Clinical outcome after 6 months
NIHSS
Intracerebral groupMean ± SD6.96 ± 2.36
Intra-arterial group Mean ± SD2.2 ± 1.92
Intravenous group Mean ± SD7.25 ± 5.31
mRS
Intracerebral groupMean ± SD3.34 ± 0.63
Intra-arterial group Mean ± SD2.33 ± 0.86
Intravenous group Mean ± SD2.63 ± 0.90
BI
Intracerebral groupMean ± SD26 ± 16.80
Intra-arterial group Mean ± SDn/a
Intravenous group Mean ± SD76.85 ± 21.41
Clinical outcome after 12 months
NIHSS
Intracerebral groupMean ± SD6.21 ± 2.49
Intra-arterial group Mean ± SDn/a
Intravenous group Mean ± SD1.58 ± 2.02
mRS
Intracerebral groupMean ± SD2.7 ± 0.46
Intra-arterial group Mean ± SDn/a
Intravenous group Mean ± SDn/a
BI
Intracerebral groupMean ± SD25.38 ± 8.74
Intra-arterial group Mean ± SDn/a
Intravenous group Mean ± SDn/a
Clinical outcome after 24 months
NIHSS
Intracerebral groupMean ± SD6.67 ± 4.03
Intra-arterial group Mean ± SDn/a
Intravenous group Mean ± SD7.73 ± 5.78
mRS
Intracerebral groupMean ± SD3 ± 1.33
Intra-arterial group Mean ± SDn/a
Intravenous group Mean ± SD2.75 ± 0.93
BI
Intracerebral groupMean ± SD31.86 ± 13.76
Intra-arterial group Mean ± SDn/a
Intravenous group Mean ± SD82 ± 27.83
Table 5. Detailed Adverse Events for Each Included Study.
Table 5. Detailed Adverse Events for Each Included Study.
Author(s)PatientsSample SizeRoute of AdministrationAESAE (CTCAE 3 or More)SAE Details
Jailllard et al. (2019) [12]3115 controlIntravenous24141 death, 1 recurrent stroke, 2 humeral fracture, 5 epileptic, 3 pneumonia, 1 gastrostomy, 1 atrial flutter
16 treatment18122 depression, 1 humeral fracture, 6 epileptic, 1 DVT, 2 pneumonia
Lee et al. (2010) [19]5236 controlIntravenous29201 new onset stroke, 2 angina, 9 pneumonia, 1 acute kidney injury, 1 systemic cancer, 1 benign mass, 5 seizure
16 treatment18112 new onset strokes, 1 angina, 1 PAOD, 3 pneumonia, 1 benign mass, 3 seizure
Honmou et al. (2011) [20]1212 treatmentIntravenous60None
Bhasin et al. (2012) [21]2412 controlIntravenous20None
12 treatment00
Prasad et al. (2012) [22]1111 treatmentIntravenous00None
Jiang et al., 2013 [23]44 treatmentIntra-arterialn/a0None
Banerjee et al. (2014) [24]55 treatmentIntra-arterial111 pneumonia
Chen et al. (2014) [25]3015 controlIntracerebraln/a0None
15 treatmentn/a0
Prasad et al. (2014) [26]12060 controlIntravenous60151 hypotension, 1 pneumonia, 1 fracture in lower limb, 5 death, 7 CNS AE
60 treatment61181 pneumonia, 1 PAOD, 2 fractures in the lower limb, 8 death, 6 CNS AE
Qiao et al. (2014) [27]66 treatmentIntravenous504 fever, 1 dizziness
Kalladka et al. (2016) [28]113 (2 m)Intracerebraln/a161 subdural hematoma, 1 epidural hematoma, 1 stroke, 1 cystoscopy, 2 bleed on burrhole site, 1 malignant melanoma, 5 gastrointestinal AE, 1 seizure, 1 alcohol withdrawal syndrome, 1 collapse, 1 community-acquired pneumonia
3 (5 m)
3 (10 m)
Hess et al. (2017) [29]1342 (20 m)Intravenous64216 severe, 6 LT, 9 deaths
71 treatment591911 severe, 3 LT, 5 deaths
Jin et al. (2017) [30]2010 controlIntracerebral1211 death due to large infarction
10 treatment1211 pneumonia
Laskowitz et al. (2018) [31]1010 treatmentIntravenous1138112 AEs were unrelated to the treatment group
Savitz et al. (2019) [15]4819 controlIntra-arterial1421 new onset stroke, 1 astrocytoma,
20 treatment2541 muscular pain, 1 UTI, 1 embolism, 1 brain edema
Steinberg et al. (2019) [3]186 (2.5 m)Intracerebral2091 seizure, 1 stenting of the carotid artery, 1 asymptomatic subdural hygroma, 1 TIA, 1 hypesthesia, 1 dysphagia, 1 UTI, 1 sepsis, 1 pneumonia
6 (5 m)31
6 (10 m)25
Zhang et al. (2019) [11]93 (12 m)Intracerebral1822 cholecystitis
3 (24 m)20None
3 (72 m)150None
Vahidy et al. (2019) [13]210185 controlIntravenous22724No study-related SAE in the therapy group
25 treatment
Chung et al. (2021) [32]5415 controlIntravenous10None
39 treatment20None
Kang Law et al. (2021) [33]179 controlIntravenous22No study-related SAE in the therapy group
8 treatment22
Ruiz et al. (2022) [34]1910 controlIntravenous1212No study-related SAE in the therapy group
9 treatment00
CNS, central nervous system; LT, life-threatening; PAOD, peripheral artery occlusive disease; TIA, transient ischemic attack; UTI, urinary tract infection.
Table 6. Adverse Events by Stem Cell Route of Administration.
Table 6. Adverse Events by Stem Cell Route of Administration.
Route of AdministrationNumber of Patients in Studies Reporting AEsNumber of AEsNumber of SAEsSAEs/Total Patients
Intravenous2952826823.05%
Intra-arterial2926517.24%
Intracerebral631232844.44%
Total38743110126.09%
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Fauzi, A.A.; Thamrin, A.M.H.; Permana, A.T.; Ranuh, I.G.M.A.R.; Hidayati, H.B.; Hamdan, M.; Wahyuhadi, J.; Suroto, N.S.; Lestari, P.; Chandra, P.S. Comparison of the Administration Route of Stem Cell Therapy for Ischemic Stroke: A Systematic Review and Meta-Analysis of the Clinical Outcomes and Safety. J. Clin. Med. 2023, 12, 2735. https://doi.org/10.3390/jcm12072735

AMA Style

Fauzi AA, Thamrin AMH, Permana AT, Ranuh IGMAR, Hidayati HB, Hamdan M, Wahyuhadi J, Suroto NS, Lestari P, Chandra PS. Comparison of the Administration Route of Stem Cell Therapy for Ischemic Stroke: A Systematic Review and Meta-Analysis of the Clinical Outcomes and Safety. Journal of Clinical Medicine. 2023; 12(7):2735. https://doi.org/10.3390/jcm12072735

Chicago/Turabian Style

Fauzi, Asra Al, Ahmad Muslim Hidayat Thamrin, Andhika Tomy Permana, I. G. M. Aswin R. Ranuh, Hanik Badriyah Hidayati, Muhammad Hamdan, Joni Wahyuhadi, Nur Setiawan Suroto, Pudji Lestari, and Poodipedi Sarat Chandra. 2023. "Comparison of the Administration Route of Stem Cell Therapy for Ischemic Stroke: A Systematic Review and Meta-Analysis of the Clinical Outcomes and Safety" Journal of Clinical Medicine 12, no. 7: 2735. https://doi.org/10.3390/jcm12072735

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop