Minireviews Open Access
Copyright ©The Author(s) 2021. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Stem Cells. Feb 26, 2021; 13(2): 168-176
Published online Feb 26, 2021. doi: 10.4252/wjsc.v13.i2.168
Review of operative considerations in spinal cord stem cell therapy
Pavan S Upadhyayula, Joel R Martin, Robert C Rennert, Joseph D Ciacci, Department of Neurological Surgery, University of California, San Diego, La Jolla, CA 92037, United States
ORCID number: Pavan S Upadhyayula (0000-0003-2071-6965); Joel R Martin (0000-0001-6898-0488); Robert C Rennert (0000-0003-3665-6696); Joseph D Ciacci (0000-0001-6078-532X).
Author contributions: Martin JR and Ciacci JD conceived of this manuscript; Martin JR, Upadhyayula PS and Rennert RC performed the literature review and initial data collection; Martin JR, Upadhyayula PS and Rennert RC wrote the manuscript; Martin JR, Upadhyayula PS, Rennert RC and Ciacci JD all edited and prepared the manuscript for submission.
Conflict-of-interest statement: The authors report no conflict of interest.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Pavan S Upadhyayula, BA, Academic Research, Department of Neurological Surgery, University of California, San Diego, 9300 Campus Point Drive, La Jolla, CA 92037, United States. psupadhy@health.ucsd.edu
Received: December 6, 2020
Peer-review started: December 6, 2020
First decision: December 31, 2020
Revised: January 18, 2021
Accepted: February 12, 2021
Article in press: February 12, 2021
Published online: February 26, 2021

Abstract

Spinal cord injury (SCI) can permanently impair motor and sensory function and has a devastating cost to patients and the United States healthcare system. Stem cell transplantation for treatment of SCI is a new technique aimed at creating biological functional recovery. Operative techniques in stem cell transplantation for SCI are varied. We review various clinical treatment paradigms, surgical techniques and technical considerations important in SCI treatment. The NCBI PubMed database was queried for “SCI” and “stem cell” with a filter placed for “clinical trials”. Thirty-nine articles resulted from the search and 29 were included and evaluated by study authors. A total of 10 articles were excluded (9 not SCI focused or transplantation focused, 1 canine model). Key considerations for stem cell transplantation include method of delivery (intravenous, intrathecal, intramedullary, or excision and engraftment), time course of treatment, number of treatments and time from injury until treatment. There are no phase III clinical trials yet, but decreased time from injury to treatment and a greater number of stem cell injections both seem to increase the chance of functional recovery.

Key Words: Stem cell, Spinal cord injury, Operative techniques, Stem cell transplantation, Intramedullary

Core Tip: Beyond the biological diversity of stem cell transplantation for spinal cord injury are the technical considerations in designing clinical treatment paradigms. The data suggest that time from injury to treatment, the duration and chronicity of treatment and the actual delivery method of cells are important considerations. This evidence seems to suggest that longer treatment paradigms soon after injury may be most beneficial.



INTRODUCTION

Spinal cord injury (SCI) is an acute traumatic event that impairs patients’ motor and sensory function; SCI is both debilitating for the individual patient as well as for the healthcare system as a whole. Although inconsistently reported, the global prevalence of SCI ranges between 236 to 1009 per million[1]. In the United States this corresponds to an incidence of between 12000 and 20000 cases with an annual total cost of almost 10 billion dollars[2]. The pathophysiology of SCI involves a primary traumatic insult followed by a secondary cascade characterized by immune activation, pro-inflammatory mediators, edema, ischemia, reactive oxygen species generation and loss of membrane integrity[3]. The primary and secondary cascade combine to create profound neurological deficits that impair normal function.

Accordingly, SCI treatment is aimed at functional and neurological recovery. Given that the functional deficits stem from neuronal damage, a major focus in this field is the regeneration of nerve tissue. To achieve this end many preclinical and clinical trials using stem cell-based therapies have begun[4]. All clinical trials to date have delivered stem cells via 3 routes: (1) Intrathecal/Intradural; (2) Intramedullary; or (3) Intravenous.

In this review we will highlight the operative considerations associated with SCI stem cell transplantation to identify common elements that may underlie the success of any given intervention. Special attention will be given to the injection site, method of delivery and treatment algorithms.

THE CURRENT LANDSCAPE OF CLINICAL INVESTIGATION INTO STEM CELL THERAPIES FOR SCI

Stem cell-based treatment for SCI is a topic of increasing clinical investigation. Currently, 18 clinical trials are registered as completed on clinicaltrials.gov with as many as 37 others recruiting patients. These span the gamut between Phase I and Phase III clinical trials. To date, most data show that stem cell injection into the spinal cord is safe with minimal side effects. While the first human trial in 2010 used human epithelial serous cystadenocarcinoma oligodendrocyte progenitor cells injected at the lesion site[5], current clinical investigations use a host of different stem cell types. These types generally fall into three broad groups: Embryonic stem cells, mesenchymal stem cells, or neural-derived stem cells. The most common group, mesenchymal stem cells, can be harvested from many sites including bone marrow-mesenchymal stem cells (BM-MSC), umbilical cord-MSC or adipose tissue-MSC[6]. The pros and cons of these different stem cells have been greatly debated[4].

To provide an accurate summary, the database of clinical trials, clinicaltrials.gov was examined for any SCI studies using stem cells. The search criteria: “SCI and stem cell” was used and completed studies were examined. Furthermore, the NCBI PubMed database was searched for “SCI” and “stem cell” with a filter placed for “clinical trials”. A total of 39 articles resulted, with 29 articles being relevant to the question at hand (9 articles were not focused on SCI/stem cell transplantation, 1 non-human model). The relevant data are summarized below with the articles summarized in Table 1. Where motor improvement is reported, this references changes in key muscle group function based on a five point grading scale as standard in SCI literature. Follow-up across all clinical trials reporting functional outcomes, vs safety profile, was a minimum of 10 mo.

Table 1 Summary of clinical trials using stem cell therapy in spinal cord injury.
Ref.
Patients
n
Technical description
Outcomes
Levi et al[17], 2019Chronic C-SCI patients12Perilesional intramedullary injection of human CNS-SCs using a two-hand stabilization techniqueUEMS showed an increase in treatment group compared to control untreated SCI patients (2.8 points in 9 mo)
Curtis et al[11], 2018 Chronic T-SCI4Instrumentation removal, laminectomy, durotomy and stereotactic injection-using a floating cannula of spinal cord-derived neural stem cellsISNCSCI improvement in 2 subjects with no adverse events
Levi et al[8], 2018 Chronic C/T-SCI29Free-hand intramedullary injection of human CNS-SCs13/29 patients experienced adverse events, all resolved by 3 mo
Xiao et al[12], 2018 Acute C/T-SCI2SCI injury site confirmed as complete and excised, collagen scaffold with hUC-MSCs transplanted as a bridge across injury siteBoth patients improved from ASIA A → ASIA C
Vaquero et al[27], 2018 Chronic SCI patients9Three intrathecal injections of 100 × 106 MSCs44% patients increased voluntary muscle contraction and 66% improved in bladder compliance with no adverse effects
Anderson et al[25], 2017Subacute T-SCI6U/S + MRI used for navigation. Table mount (Geron Corp) and Hamilton syringe used for intramedullary microinjection of sural nerve-derived SCsNo major adverse events and no consistent improvement in ISNCSCI
Vaquero et al[18], 2017Chronic incomplete C/T/L SCI 12 Subarachnoid administration via lumbar puncture of autologous MSCsSexual function (2/8), spasticity (3/9) and bowel/bladder function improved (8/9) improvements were noted
Vaquero et al[28], 2016Chronic complete C/T/L SCI12 Subarachnoid administration via lumbar puncture of autologous MSCsAll patients experienced improvement in sensation and sphincter control. Motor activity below the lesion obtained in 50% of patients
Satti et al[29], 2016 Chronic and subacute T-SCI 6Intrathecal injection of autologous MSCsEvaluated safety only-no adverse events
Bansal et al[22], 2016 SCI patients 8Lumbar puncture at L1/L2 with autologous BMSCs injected 3 times every 4 wkPatients with injury less than 6 mo improved-ASIA grade improvement in 6/10, walking with support restored in 8/10
Hur et al[21], 2016 Subacute to chronic C/T/L-SCI14Intrathecal injection through lumbar tap of 9 × 107 ADMSCASIA motor improved in ⅝ patients. 4 adverse events included headache and UTI
Oh et al[10], 2016 Chronic C-SCI16Laminectomy and durotomy with 1.6 × 107 BM-MSCs in 1 mL injected intramedullary with a 27 gauge needle. Fibrin glue used to prevent cell leakage. 3.2 × 107 BM-MSCs injected into the subdural space12.5% of patients with significant motor improvement
Shin et al[20], 2015 Acute/subacute C- SCI 15 Human fetal tissue-derived neural stem cell progenitor cells free hand injection 5 mm deep into lesion site5/19 in the treatment group with improved ASIA grade, compared to 1/15 in the control group with ASIA improvement
Mendonça et al[19], 2014 Chronic T/L-SCI14 BM-MSCs injected based on lesion volume. Direct injection above and below level8/14 developed lower limb functional gain in hip flexors. 7/14 improved ASIA grades to B/C 9/14 with improved urologic function
Cheng et al[30], 2014 Chronic T/L-SCI10CT-guided intramedullary injection at the lesion site using purified UC-MSCs. Two transplantations separated by 10 d, each transplantation with 3 separate injections of 2 × 107 cells7/10 patients had significant improvement in movement and muscle tension
Al-Zoubi et al[31], 2014 Chronic T-SCI19Autologous purified CD34+/CD133+ SCs injected into cyst cavity or subarachnoid space7/19 patients with segmental sensory improvement, 2/19 with motor improvement (ASIA-A → ASIA-C)
Yoon et al[23], 2014 Acute/subacute/chronic C-SCI25Intramedullary perilesional injection of 2 × 108 BMCs in 6 locations + 5 cycles of GM-CSF SubqASIA grade increased in 30.4% of acute and subacute treated patients with no improvement in chronic treatment group
Dai et al[32], 2013 Chronic complete C-SCI20BMMSC transplantation at site of injury with MIS-laminectomy, dural incision and injection at a depth of 3 mm at central dorsal aspect of the junction between the lesioned and normal spinal cord.10/20 in BMMSC transplantation group had improvement in motor, light touch and pinprick sensory with 9/20 showing ASIA improvement. No improvement in any control patients (0/20)
Park et al[9], 2012 Traumatic C-SCI 10Laminectomy and durotomy with 8 × 106 MSCs in 1 mL injected intramedullary over 10 s with a 26.5 gauge needle. Fibrin glue used to prevent cell leakage. At 4-8 wks post-op additional 5 × 107 MSCs injected via lumbar tap6/10 patients with motor power improvement of UE
Frolov et al[33], 2012 Chronic C-SCI20 Repeated intrathecal autologous HSCs (from leukapheresis) repeatedly injected over 1 yr3-4 patients with improved SEP and MEP
Karamouzian et al[24], 2012 Acute/subacute T-SCI 11Purified BM-MSC injected via standard lumbar puncture needle5/11 in BM-MSC treatment group had two grade improvement in ASIA score (i.e. A → C) compared to 3/20 in control group (P = 0.09)
Ra et al[34], 2011 Chronic SCI8IV administration of human ADMSCsSafe with no adverse events related to transplantation at 3 mo
Lima et al[14], 2009Chronic C/T SCI20Laminectomy with partial scar excision and olfactory mucosal autograft placement. Rehabilitation focused on lower extremity stepping continued post-operatively11/20 patients had ASIA improvement (6A → C, 3B → C, 2A → B) with 1/20 having ASIA decline (B → A). 15/20 with new voluntary EMG
Cristante et al[35], 2009 Chronic C/T-SCI39Apheresis for isolation of CD34+ bone marrow mononuclear stem cells-injected endovascularly via intercostal arteries or vertebral arteries26/39 patients showed recovery of SSEP to peripheral stimuli
Pal et al[36], 2009 Subacute to Chronic C/T SCI30BM-MSC expanded ex-vivo and injected via LPInjection safe with no adverse events
Mackay-Sim et al[37], 2008 Chronic complete T-SCI6Nasal biopsy for isolation of OESC, cultured for 4-10 wks. Laminectomy, durotomy and injection into damaged spinal cord and proximal/distal ends of lesion with a table mounted stereotactic injection apparatusNo adverse events, 1 of 6 patients with an improvement of 3 segments in LT/PP
Chernykh et al[38], 2007 Chronic C/T/L SCI18Purified BM-MSCs injected into the cystic lesion cavity and given intravenouslyMotor and sensory improvement was equivocal, spasticity was significantly improved by BM-MSC injection
Lima et al[13], 2006 Chronic C/T-SCI7Laminectomy, with scar excision with suturing graft loaded with olfactory tissue to meninges/superficial tissue layers2 patients went from ASIA-A to ASIA-C (out of 7 total) with return of bladder sensation/VAC
Callera et al[39], 2006 Chronic SCI10BM-MSCs injected via LPInjection safe with no adverse events
TECHNICAL CONSIDERATIONS IN STEM CELL DELIVERY
Stem cell transplant techniques

Across the many clinical trials examining the use of stem cells (SCs) in the treatment of SCI, there are a few key technical considerations. The first is the method of injection. Intravenous and intrathecal injection have relatively few technical considerations since IV injection and lumbar puncture are routine procedures. Intramedullary injection of stem cells, however, poses a greater challenge. The spinal cord generally oscillates with respiration and cardiac pulsation. Studies in animals show that the pulsatile nature of the cord is increased following increases in blood pressure and that it is not due to cerebrospinal fluid based wave transmission. While respiratory oscillation causes greatest spinal cord movement in the thoracic spine, the genuine spinal cord pulsation is thought to be driven by the blood flow through radicular arteries[7]. This spinal cord pulsation may pose an issue with intramedullary injection techniques. In general there are two canonical ways of achieving intramedullary spinal cord injections: The free hand technique, and a stereotactic technique utilizing a bed mounted frame. It is important to note that the stereotactic technique, as it is mounted to the bed, does not necessarily improve stability in relation to spinal cord pulsation. A recent study by Levi et al[8] described the use of a free hand technique. Specifically, to achieve an injection depth of 3-5 mm they marked the injection needle using a rongeur or silicone tip and had the surgeon stabilize the needle using two hands anchored at the edge of the surgical field. The injection time, which was a maximum of 3 min and 30 s per injection, required the use of two hands for stabilization[8].

Although no serious complications occurred in this trial or others that used a free-hand technique[9,10], the damage to the spinal cord may be obfuscated by the presence of preexisting pathology. Moreover, the need for stabilization may limit the maximum injection time and thereby the amount of stem cells able to be transplanted. Curtis et al[11] described a novel technique using a floating cannula that is able to address these theoretical issues. This cannula is attached to an XYZ manipulator mounted directly on a patient's vertebral column. The cannula is able to move with pulsation of the spinal cord making long term injections feasible with minimal damage to the existing spinal cord tissue[11].

Whether the preservation of existing spinal cord tissue is technically necessary is up for debate. While the previously described studies focused on chronic SCI, a study by Xiao et al[12] in acute SCI patients completely excised the necrotic spinal cord around the SCI lesion. The authors placed a collagen scaffold impregnated with human umbilical cord MSCs in this area and showed nerve conduction across the SCI lesion plus functional improvement from American Spinal Injury Association Impairment Scale (ASIA) A to ASIA C in two patients[12]. This study was built on previous work by Lima et al[13], where necrotic scar tissue was excised and an olfactory mucosal autograft was placed. Importantly, this radical technique of laminectomy, scar excision and mucosal autograft replacement in chronic cervical and thoracic SCI led to marked functional improvement. In the earlier study by Lima et al[13], 28% of patients had recovery of bladder sensation or voluntary anal contraction[13], while in the more recent study 55% of patients had ASIA improvement of at least 1 grade[14]. Importantly, this second study involved intensive rehabilitation following autograft transplantation. It is important to note that olfactory mucosa grafts have been associated with spinal masses pointing to their increased regenerative potential but also to their increased side effect profile[15,16]. As a technical consideration, excision of scar tissue may allow for increased regeneration of neural white matter tracts and may be an important technical consideration even with other injection techniques that do not use a graft substrate or scaffold.

Number of treatments

Although some studies of SCI show improvement following a single stem cell injection[11,17], studies with multiple injections spaced out over time seem to have greater improvements in outcomes[8,18]. A comparison of two trials conducted by the same group, Oh et al[10] and Park et al[9], further illustrated this point. The original study, by Park et al[9], was a Phase I single-arm study and included the injection of BM-MSCs derived from iliac crest grafts into both the intramedullary and subdural space with additional injections into the thecal space using lumbar puncture at 4 wk and 8 wk following the initial operation. Six of 10 patients showed motor improvement and 3 showed gradual improvement in activities of daily living[9]. The follow-up study, by Oh et al[10], was a Phase III clinical trial and the study authors used the same initial injection treatment paradigm with no follow-up injections. The one time injection yielded poor functional improvement (12.5% with improved motor outcomes) compared to the multiple injection protocol (60% with motor improvement or improvement on Activities of Daily Living). Although limited, these data point to the importance of optimizing chronicity of SC injection in SCI patients. Importantly, while 5/16 patients had diffusion tensor imaging magnetic resonance imaging changes showing tracts spanning the SCI level in the single injection Phase III clinical trial compared to 7/10 showing such changes in the pilot study[10]. It is difficult to associate the differences solely due to the presence of multiple treatments. Multiple other studies have shown improvements in ASIA scores, motor function and urodynamics with only a single stem-cell treatment[19,20]. Furthermore, understanding the number of cells transplanted and its effect on outcomes is more difficult. Various trials have transplanted cell numbers ranging from 1 × 105 to 40 × 107. There was no consistent relationship between cell number and outcome over the trials reported. It remains to be seen whether a multiple treatment vs single treatment protocol could improve functional recovery in a large-scale clinical trial.

Time from injury to treatment

Generally, SCI is characterized based on chronicity into acute, subacute and chronic phases. While the time course for each period is highly variable, the acute phase generally spans days post-injury, the subacute phase weeks post-injury and the chronic phase months post-injury. Although some studies on chronic SCI show improvement following stem cell injection[8,11], studies in the acute or subacute SCI population seem to show a more dramatic return of function[10,21,22]. A study by Yoon et al[23] stratified patients into acute (< 13 d), subacute (14 d to 8 wk) and chronic (> 8 wk) groups with all patients receiving intramedullary injection of BM-MSCs with systemic granulocyte macrophage colony stimulating factor treatment. Notably, 30.4% of the acute and subacute treated patients had improved ASIA grade (ASIA A to Asia B/C), while none of the chronic patients showed any improvement[23]. This was not limited to intramedullary cell transplantation. Bansal et al[22] demonstrated that lumbar puncture for delivery of BM-MSCs led to ASIA grade improvement in 6/10 patients who were less than 6 mo from injury with no patients over 6 mo from injury achieving functional improvement[22].

A study by Shin et al[20], used human fetal tissue-derived neural stem cell progenitor cells and included both a control group and an intramedullary cell transplantation group. Notably, while 26% of patients in the transplantation group had ASIA grade improvement, only 6.6% had improvement in the control group[20]. This disparity between transplantation and control group patients was also seen by Karamouzian et al[24]. These authors injected purified BM-MSCs via lumbar puncture and noted that 45 patients in the cell transplantation group had ASIA improvement compared to 15% in the control group (P = 0.09)[24]. This helps to answer the major critique that some degree of functional improvement occurs in the acute/subacute period naturally and may explain the functional improvements seen in the acute to subacute cell transplantation studies. Also, the fact that both studies with control groups used different stem cell types and different methods of injection (lumbar puncture vs intramedullary injection) also highlights the importance of the treatment window vs mechanism of treatment. This is further supported by the studies carried out by Bansal et al[22] and Yoon et al[23]. These studies had patients in multiple treatment windows, with intramedullary or lumbar puncture delivered BM-MSCs and noted that patients in the acute to subacute period from injury achieved greater functional improvement[22,23]. In fact, of the studies that focused on the acute/subacute SCI population, only one out of seven studies showed no motor or functional improvement with the rest having a subset of patients that had ASIA grade improvement. The single study with no functional improvement notably used a novel form of stem cells derived from peripheral Schwann cells and included subjects from 4-7 wk following injury-generally on the upper limit as compared to other acute/subacute studies[25].

FUTURE DIRECTIONS

In summary, clinical studies to date have highlighted a few key findings. First, that stem cell injection is generally well tolerated with a minimal side effect profile when appropriately dosed. The use of stem cell treatment in SCI may lead to functional improvement when intervention is performed in the acute to subacute treatment window and when multiple treatment injections are utilized. A few pre-clinical studies are examining devices that could facilitate intramedullary stem cell injection. One such device tested in rodents and pigs by Kutikov et al[26] injects cells in a trail creating longitudinal tracts of neural stem cells vs isolated injection sites. In doing so they demonstrated a novel technique for stem cell injections able to create new tracts that span multiple spinal cord levels[26]. Continued technological development could help facilitate intramedullary stem cell injection over longer periods of time, thereby obviating the greatest risk to this treatment, the need for surgical delivery. The aim of this manuscript is to help optimize clinical trial parameters, patient selection and cell transplantation techniques. Comparative clinical trials using different types of stem cells are necessary to determine what type of cells are most efficacious in improving functional outcomes in patients.

CONCLUSION

Stem cell treatment for SCI is a burgeoning field. While numerous studies have focused on the biological aspect of this treatment, technical challenges remain. Time from injury to treatment, the duration and chronicity of treatment and the actual delivery of cells are important considerations. Currently, the lack of phase III clinical trials directly studying these factors makes it difficult to draw conclusions. Early evidence seems to suggest that longer treatment paradigms soon after injury may be most beneficial. Finally, operative techniques and devices that can effectively target the intramedullary space could help with stem cell delivery and functional recovery following treatment.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Cell and tissue engineering

Country/Territory of origin: United States

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B, B, B, B

Grade C (Good): C, C

Grade D (Fair): D

Grade E (Poor): 0

P-Reviewer: Fessler R, Klimczak A, Sukumaran A, Zhao L S-Editor: Fan JR L-Editor: Webster JR P-Editor: Xing YX

References
1.  Singh A, Tetreault L, Kalsi-Ryan S, Nouri A, Fehlings MG. Global prevalence and incidence of traumatic spinal cord injury. Clin Epidemiol. 2014;6:309-331.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 155]  [Cited by in F6Publishing: 353]  [Article Influence: 35.3]  [Reference Citation Analysis (0)]
2.  Ma VY, Chan L, Carruthers KJ. Incidence, prevalence, costs, and impact on disability of common conditions requiring rehabilitation in the United States: stroke, spinal cord injury, traumatic brain injury, multiple sclerosis, osteoarthritis, rheumatoid arthritis, limb loss, and back pain. Arch Phys Med Rehabil 2014; 95: 986-995. e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 499]  [Cited by in F6Publishing: 534]  [Article Influence: 53.4]  [Reference Citation Analysis (0)]
3.  Alizadeh A, Dyck SM, Karimi-Abdolrezaee S. Traumatic Spinal Cord Injury: An Overview of Pathophysiology, Models and Acute Injury Mechanisms. Front Neurol. 2019;10:282.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 342]  [Cited by in F6Publishing: 539]  [Article Influence: 107.8]  [Reference Citation Analysis (0)]
4.  Jin MC, Medress ZA, Azad TD, Doulames VM, Veeravagu A. Stem cell therapies for acute spinal cord injury in humans: a review. Neurosurg Focus. 2019;46:E10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 83]  [Article Influence: 20.8]  [Reference Citation Analysis (0)]
5.  Frantz S. Embryonic stem cell pioneer Geron exits field, cuts losses. Nat Biotechnol. 2012;30:12-13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 64]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
6.  Kibat P, Stricker H. [Factors of elimination of liposome-encapsulated drug model substances after intravenous administration]. Arzneimittelforschung. 1988;38:1472-1478.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 44]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
7.  Matsuzaki H, Wakabayashi K, Ishihara K, Ishikawa H, Kawabata H, Onomura T. The origin and significance of spinal cord pulsation. Spinal Cord. 1996;34:422-426.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 30]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
8.  Levi AD, Okonkwo DO, Park P, Jenkins AL 3rd, Kurpad SN, Parr AM, Ganju A, Aarabi B, Kim D, Casha S, Fehlings MG, Harrop JS, Anderson KD, Gage A, Hsieh J, Huhn S, Curt A, Guzman R. Emerging Safety of Intramedullary Transplantation of Human Neural Stem Cells in Chronic Cervical and Thoracic Spinal Cord Injury. Neurosurgery. 2018;82:562-575.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 88]  [Article Influence: 17.6]  [Reference Citation Analysis (0)]
9.  Park JH, Kim DY, Sung IY, Choi GH, Jeon MH, Kim KK, Jeon SR. Long-term results of spinal cord injury therapy using mesenchymal stem cells derived from bone marrow in humans. Neurosurgery. 2012;70:1238-47; discussion 1247.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 128]  [Cited by in F6Publishing: 132]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
10.  Oh SK, Choi KH, Yoo JY, Kim DY, Kim SJ, Jeon SR. A Phase III Clinical Trial Showing Limited Efficacy of Autologous Mesenchymal Stem Cell Therapy for Spinal Cord Injury. Neurosurgery. 2016;78:436-47; discussion 447.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 107]  [Article Influence: 13.4]  [Reference Citation Analysis (0)]
11.  Curtis E, Martin JR, Gabel B, Sidhu N, Rzesiewicz TK, Mandeville R, Van Gorp S, Leerink M, Tadokoro T, Marsala S, Jamieson C, Marsala M, Ciacci JD. A First-in-Human, Phase I Study of Neural Stem Cell Transplantation for Chronic Spinal Cord Injury. Cell Stem Cell 2018; 22: 941-950. e6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 159]  [Cited by in F6Publishing: 194]  [Article Influence: 38.8]  [Reference Citation Analysis (0)]
12.  Xiao Z, Tang F, Zhao Y, Han G, Yin N, Li X, Chen B, Han S, Jiang X, Yun C, Zhao C, Cheng S, Zhang S, Dai J. Significant Improvement of Acute Complete Spinal Cord Injury Patients Diagnosed by a Combined Criteria Implanted with NeuroRegen Scaffolds and Mesenchymal Stem Cells. Cell Transplant. 2018;27:907-915.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 95]  [Article Influence: 15.8]  [Reference Citation Analysis (0)]
13.  Lima C, Pratas-Vital J, Escada P, Hasse-Ferreira A, Capucho C, Peduzzi JD. Olfactory mucosa autografts in human spinal cord injury: a pilot clinical study. J Spinal Cord Med. 2006;29:191-203; discussion 204.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 251]  [Cited by in F6Publishing: 258]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
14.  Lima C, Escada P, Pratas-Vital J, Branco C, Arcangeli CA, Lazzeri G, Maia CA, Capucho C, Hasse-Ferreira A, Peduzzi JD. Olfactory mucosal autografts and rehabilitation for chronic traumatic spinal cord injury. Neurorehabil Neural Repair. 2010;24:10-22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 173]  [Cited by in F6Publishing: 169]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
15.  Woodworth CF, Jenkins G, Barron J, Hache N. Intramedullary cervical spinal mass after stem cell transplantation using an olfactory mucosal cell autograft. CMAJ. 2019;191:E761-E764.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 16]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
16.  Dlouhy BJ, Awe O, Rao RC, Kirby PA, Hitchon PW. Autograft-derived spinal cord mass following olfactory mucosal cell transplantation in a spinal cord injury patient: Case report. J Neurosurg Spine. 2014;21:618-622.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 120]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
17.  Levi AD, Anderson KD, Okonkwo DO, Park P, Bryce TN, Kurpad SN, Aarabi B, Hsieh J, Gant K. Clinical Outcomes from a Multi-Center Study of Human Neural Stem Cell Transplantation in Chronic Cervical Spinal Cord Injury. J Neurotrauma. 2019;36:891-902.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 86]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
18.  Vaquero J, Zurita M, Rico MA, Bonilla C, Aguayo C, Fernández C, Tapiador N, Sevilla M, Morejón C, Montilla J, Martínez F, Marín E, Bustamante S, Vázquez D, Carballido J, Rodríguez A, Martínez P, García C, Ovejero M, Fernández MV; Neurological Cell Therapy Group. Repeated subarachnoid administrations of autologous mesenchymal stromal cells supported in autologous plasma improve quality of life in patients suffering incomplete spinal cord injury. Cytotherapy. 2017;19:349-359.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 85]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
19.  Mendonça MV, Larocca TF, de Freitas Souza BS, Villarreal CF, Silva LF, Matos AC, Novaes MA, Bahia CM, de Oliveira Melo Martinez AC, Kaneto CM, Furtado SB, Sampaio GP, Soares MB, dos Santos RR. Safety and neurological assessments after autologous transplantation of bone marrow mesenchymal stem cells in subjects with chronic spinal cord injury. Stem Cell Res Ther. 2014;5:126.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 129]  [Article Influence: 12.9]  [Reference Citation Analysis (0)]
20.  Shin JC, Kim KN, Yoo J, Kim IS, Yun S, Lee H, Jung K, Hwang K, Kim M, Lee IS, Shin JE, Park KI. Clinical Trial of Human Fetal Brain-Derived Neural Stem/Progenitor Cell Transplantation in Patients with Traumatic Cervical Spinal Cord Injury. Neural Plast. 2015;2015:630932.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 84]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
21.  Hur JW, Cho TH, Park DH, Lee JB, Park JY, Chung YG. Intrathecal transplantation of autologous adipose-derived mesenchymal stem cells for treating spinal cord injury: A human trial. J Spinal Cord Med. 2016;39:655-664.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 97]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
22.  Bansal H, Verma P, Agrawal A, Leon J, Sundell IB, Koka PS. Autologous Bone Marrow-Derived Stem Cells in Spinal Cord Injury. J Stem Cells. 2016;11:51-61.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Yoon SH, Shim YS, Park YH, Chung JK, Nam JH, Kim MO, Park HC, Park SR, Min BH, Kim EY, Choi BH, Park H, Ha Y. Complete spinal cord injury treatment using autologous bone marrow cell transplantation and bone marrow stimulation with granulocyte macrophage-colony stimulating factor: Phase I/II clinical trial. Stem Cells. 2007;25:2066-2073.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 269]  [Cited by in F6Publishing: 280]  [Article Influence: 16.5]  [Reference Citation Analysis (0)]
24.  Karamouzian S, Nematollahi-Mahani SN, Nakhaee N, Eskandary H. Clinical safety and primary efficacy of bone marrow mesenchymal cell transplantation in subacute spinal cord injured patients. Clin Neurol Neurosurg. 2012;114:935-939.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 104]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
25.  Anderson KD, Guest JD, Dietrich WD, Bartlett Bunge M, Curiel R, Dididze M, Green BA, Khan A, Pearse DD, Saraf-Lavi E, Widerström-Noga E, Wood P, Levi AD. Safety of Autologous Human Schwann Cell Transplantation in Subacute Thoracic Spinal Cord Injury. J Neurotrauma. 2017;34:2950-2963.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in F6Publishing: 162]  [Article Influence: 23.1]  [Reference Citation Analysis (0)]
26.  Kutikov AB, Moore SW, Layer RT, Podell PE, Sridhar N, Santamaria AJ, Aimetti AA, Hofstetter CP, Ulich TR, Guest JD. Method and Apparatus for the Automated Delivery of Continuous Neural Stem Cell Trails Into the Spinal Cord of Small and Large Animals. Neurosurgery. 2019;85:560-573.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 7]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
27.  Vaquero J, Zurita M, Rico MA, Aguayo C, Bonilla C, Marin E, Tapiador N, Sevilla M, Vazquez D, Carballido J, Fernandez C, Rodriguez-Boto G, Ovejero M; Neurological Cell Therapy Group from Puerta de Hierro-Majadahonda Hospital. Intrathecal administration of autologous mesenchymal stromal cells for spinal cord injury: Safety and efficacy of the 100/3 guideline. Cytotherapy. 2018;20:806-819.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 67]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
28.  Vaquero J, Zurita M, Rico MA, Bonilla C, Aguayo C, Montilla J, Bustamante S, Carballido J, Marin E, Martinez F, Parajon A, Fernandez C, Reina L; Neurological Cell Therapy Group. An approach to personalized cell therapy in chronic complete paraplegia: The Puerta de Hierro phase I/II clinical trial. Cytotherapy. 2016;18:1025-1036.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 70]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
29.  Satti HS, Waheed A, Ahmed P, Ahmed K, Akram Z, Aziz T, Satti TM, Shahbaz N, Khan MA, Malik SA. Autologous mesenchymal stromal cell transplantation for spinal cord injury: A Phase I pilot study. Cytotherapy. 2016;18:518-522.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 88]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
30.  Cheng H, Liu X, Hua R, Dai G, Wang X, Gao J, An Y. Clinical observation of umbilical cord mesenchymal stem cell transplantation in treatment for sequelae of thoracolumbar spinal cord injury. J Transl Med. 2014;12:253.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 91]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
31.  Al-Zoubi A, Jafar E, Jamous M, Al-Twal F, Al-Bakheet S, Zalloum M, Khalifeh F, Radi SA, El-Khateeb M, Al-Zoubi Z. Transplantation of purified autologous leukapheresis-derived CD34+ and CD133+ stem cells for patients with chronic spinal cord injuries: long-term evaluation of safety and efficacy. Cell Transplant. 2014;23 Suppl 1:S25-S34.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 26]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
32.  Dai G, Liu X, Zhang Z, Yang Z, Dai Y, Xu R. Transplantation of autologous bone marrow mesenchymal stem cells in the treatment of complete and chronic cervical spinal cord injury. Brain Res. 2013;1533:73-79.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 84]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
33.  Frolov AA, Bryukhovetskiy AS. Effects of hematopoietic autologous stem cell transplantation to the chronically injured human spinal cord evaluated by motor and somatosensory evoked potentials methods. Cell Transplant. 2012;21 Suppl 1:S49-S55.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 27]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
34.  Ra JC, Shin IS, Kim SH, Kang SK, Kang BC, Lee HY, Kim YJ, Jo JY, Yoon EJ, Choi HJ, Kwon E. Safety of intravenous infusion of human adipose tissue-derived mesenchymal stem cells in animals and humans. Stem Cells Dev. 2011;20:1297-1308.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 392]  [Cited by in F6Publishing: 408]  [Article Influence: 31.4]  [Reference Citation Analysis (0)]
35.  Cristante AF, Barros-Filho TE, Tatsui N, Mendrone A, Caldas JG, Camargo A, Alexandre A, Teixeira WG, Oliveira RP, Marcon RM. Stem cells in the treatment of chronic spinal cord injury: evaluation of somatosensitive evoked potentials in 39 patients. Spinal Cord. 2009;47:733-738.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 66]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
36.  Pal R, Venkataramana NK, Bansal A, Balaraju S, Jan M, Chandra R, Dixit A, Rauthan A, Murgod U, Totey S. Ex vivo-expanded autologous bone marrow-derived mesenchymal stromal cells in human spinal cord injury/paraplegia: a pilot clinical study. Cytotherapy. 2009;11:897-911.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 174]  [Cited by in F6Publishing: 186]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
37.  Mackay-Sim A, Féron F, Cochrane J, Bassingthwaighte L, Bayliss C, Davies W, Fronek P, Gray C, Kerr G, Licina P, Nowitzke A, Perry C, Silburn PA, Urquhart S, Geraghty T. Autologous olfactory ensheathing cell transplantation in human paraplegia: a 3-year clinical trial. Brain. 2008;131:2376-2386.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 299]  [Cited by in F6Publishing: 295]  [Article Influence: 18.4]  [Reference Citation Analysis (0)]
38.  Chernykh ER, Stupak VV, Muradov GM, Sizikov MY, Shevela EY, Leplina OY, Tikhonova MA, Kulagin AD, Lisukov IA, Ostanin AA, Kozlov VA. Application of autologous bone marrow stem cells in the therapy of spinal cord injury patients. Bull Exp Biol Med. 2007;143:543-547.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 47]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
39.  Callera F, do Nascimento RX. Delivery of autologous bone marrow precursor cells into the spinal cord via lumbar puncture technique in patients with spinal cord injury: a preliminary safety study. Exp Hematol. 2006;34:130-131.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 82]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]