Warning: mkdir(): Permission denied in /home/virtual/lib/view_data.php on line 81 Warning: fopen(/home/virtual/e-kjs/journal/upload/ip_log/ip_log_2024-03.txt): failed to open stream: No such file or directory in /home/virtual/lib/view_data.php on line 83 Warning: fwrite() expects parameter 1 to be resource, boolean given in /home/virtual/lib/view_data.php on line 84 Established and Emerging Therapies in Acute Spinal Cord Injury
Neurospine Search

CLOSE


Gadot, Smith, Prablek, Grochmal, Fuentes, and Ropper: Established and Emerging Therapies in Acute Spinal Cord Injury

Abstract

Acute spinal cord injury (SCI) is devastating for patients and their caretakers and has an annual incidence of 20–50 per million people. Following initial assessment with appropriate physical examination and imaging, patients who are deemed surgical candidates should undergo decompression with stabilization. Earlier intervention can improve neurological recovery in the post-operative period while allowing earlier mobilization. Optimized medical management is paramount to improve outcomes. Emerging strategies for managing SCI in the acute period stem from an evolving understanding of the pathophysiology of the injury. General areas of focus include ischemia prevention, reduction of secondary injury due to inflammation, modulation of the cytotoxic and immune response, and promotion of cellular regeneration. In this article, we review established, emerging, and novel experimental therapies. Continued translational research on these methods will improve the feasibility of bench-to-bedside innovations in treating patients with acute SCI.

INTRODUCTION

Spinal cord injuries (SCIs) are devastating traumatic events in the lives of patients, often resulting in severe and/or permanent neurologic disabilities. Nearly half a million people are living permanently disabled in the United States due to traumatic SCI, and 12,000–15,000 patients per year incur new injuries [1,2]. Further, the incidence of SCI varies by country with most developed countries reporting incidences of 20–50 per million [3,4]. The event can result in an assortment of immediate sequelae and long-term complications including loss of motor and sensory function, loss of autonomic function, and increased risk of medical complications and death [5].
As traumatic SCI more commonly occurs in young, healthy adults, the associated quality-adjusted life year and economic loss can be immense [6]. Despite recent gains in understanding of the pathogenesis and improved acute management of SCI, there remains an ongoing need to investigate novel molecular targets and therapeutics for treatment in the acute injury period, with the goal of maximizing patient recovery and preventing chronic manifestations of the injury. While the timing of surgery for decompression and stabilization of acute SCI has been well studied [7], the evidence for most medical treatments of the acute neurological injury are less well supported. This review discusses the established current management strategies and expands on emerging therapeutic strategies for acute SCI. Additionally, we discuss an updated review of experimental therapies, from bench to bedside.

MECHANISM OF INJURY IN SPINAL CORD INJURY

SCI can result either from high-energy trauma (e.g., motor vehicle accidents, diving, sports) or lower impact trauma, which often occurs in the presence of underlying degenerative or congenital spinal spondylosis. The cervical spinal cord is the most commonly affected region due to the lack of bony and muscular protection relative to thoracic and lumbar segments [8]. The mechanism of spinal cord damage is generally divided into primary and secondary processes. Primary injury encompasses the initial insult, such as compression, penetrating injury, or strain on neural tissue or vascular structures. Subsequent perfusion disruption progresses to local hypoxia and can be worsened by systemic vasogenic shock. Hypoxic cell death induces the multifaceted, progressive process of secondary injury during the following hours to weeks. Ischemia caused by primary injury activates release of vasoactive proteins and cytokines, and the resulting edema and inflammation promote cytoskeletal and mitochondrial damage and immune cell infiltration. The zone of injury then expands due to reactive oxygen species (ROS) accumulation and oxidative damage, lactic acidosis, fibrin and platelet accumulation, toxic excitatory neurotransmitter release, and axonal demyelination, leading to progressive tissue death and scarring.

CLINICAL PRESENTATION

Initial management of patients with suspected acute traumatic SCI follows established trauma guidelines to manage the airway, breathing, and circulation. Immobilization is necessary to prevent further possible damage in all patients with suspected SCI (characterized, for example, by polytrauma, neck/back spinal pain, dysesthesia, loss of consciousness, or a mechanism of injury with the potential to cause an SCI) [9-11].
Patients with SCI can present with either complete injury, defined as a lack of motor or sensory function in the anal and perineal region (representing the lowest sacral segments S4–S5), or incomplete injury with varying degrees of motor and sensory function caudal to the level of the injury [12]. The American Spinal Injury Association (ASIA) scoring system is used to stratify injury severity, ranging from complete injury (ASIA-A) to normal neurological exam (ASIA-E), to standardize assessment and treatment of SCI [2]. In addition, patients can present with spinal shock, an often-transient physiologic depression of spinal cord function secondary to cell damage which can be tracked with progressive recovery of anatomical reflexes including the bulbocavernosus, anal cutaneous, and plantar reflexes [12]. Spinal shock and medication effects may obscure accurate ASIA scoring, particularly in the first 48–72 hours after injury [13]. This process is distinct from neurogenic shock, a condition of autonomic dysregulation from injury to spinal levels C1–T6 and/or associated sympathetic ganglia, which is characterized by persistent hypotension, bradycardia, and hypothermia [12].

ESTABLISHED ACUTE MANAGEMENT STRATEGIES

Computed tomography of the entire spine is indicated in the initial evaluation of traumatic SCI patients [14]. ASIA motor scores and neurologic level of injury have the most consistent prognostic value regarding outcome [15]. Magnetic resonance imaging has utility for guiding surgical intervention and prognostication and should be performed when available but should not delay surgical treatment in a neurologically declining patient [12,15]. Standardization of magnetic resonance changes of increasing severity in acute SCI is increasing, with the BASIC score serving as one example [16]/ Surgical candidacy should be promptly determined through assessment of a worsening neurological examination along with signs and symptoms of instability and progressive compression.
Direct spinal cord compression is the most frequent mechanism of SCI and can progress following the initial injury, prompting a need for emergent surgical decompression. A meta-analysis including 16 studies involving nearly 4,000 patients found that patients who receive early spinal surgery (< 24 hours following injury) experience greater neurological recovery, shorter length of stay, lower hospitalization costs, and a lower incidence of complications than those who undergo surgery > 24 hours following injury [17]. Another recent multicenter clinical trial compared early and delayed surgical treatment (defined as ≤24 hours after injury and after ≥ 2 weeks of conservative therapy, respectively) and assessed motor recovery using the ASIA motor score, spinal cord independence measure, and independent ambulation capacity for 1 year. The study found that the 2 treatment paradigms were not significantly different; however, motor recovery at 2-week, 3-month, and 6-month follow-ups were improved with early surgical treatment, indicating accelerated recovery [18]. These results align with several large, well-documented trials and underscore the importance of early decompression to improve potential for motor recovery following SCI [19-21].
Medical management has an essential role in the optimization of patients with SCI [12,22]. Hemodynamic control is crucial in the early stages of acute SCI to reduce potential ischemic injury, with goal mean arterial pressures (MAPs) of 85–90 mmHg for 5–7 days suggested for improved functional outcomes [23]. Other crucial tenets of management include prevention of pulmonary (i.e., pneumonia), gastrointestinal (i.e., bowel obstruction), and urologic (i.e., dysautonomic uropathy) complications caused by loss of voluntary and autonomic control [12,14]. As such, current multidisciplinary clinical studies are examining, for example, the use of low versus high tidal volume mechanical ventilation in patients requiring intubation in acute SCI (NCT04912583) and the use of early epidural and sacral nerve stimulation as an adjunct for improving bladder function (NCT03083366) [24,25]. The utility of methylprednisolone (MPSS) administration has been discussed extensively in the literature and, while initially thought to improve outcomes based on the early North American Spinal Cord Injury Study (NASCIS) results, subsequent large studies have not demonstrated this benefit [26-28]. Moreover, a consistently increased risk of steroid-associated complications including infection was found. Recent animal studies have found that high-dose steroid administration may actually increase the extravasation of plasma components after SCI and can enhance tissue swelling and edema [29]. Therefore, routine administration of high-dose steroids is no longer recommended; a short course may, however, be considered in young, otherwise healthy adults presenting <8 hours from injury based on expert consensus [27,28,30]. Early physical rehabilitation, nutritional optimization, and mental health consultation are indicated, and have been shown to improve functionality and quality of life [31-33]. These therapies continue to be studied, for example, in clinical trials assessing the effects of interval versus continuous aerobic training on autonomic dysreflexia in the acute phase of SCI (NCT05061160).

EXPERIMENTAL ACUTE MANAGEMENT STRATEGIES

Emerging therapeutic strategies for acute SCI can be classified by the biological mechanism which they are designed to address, prevent, or reverse. Mechanisms largely discussed in the literature to date can be grouped into categories as described below based on the current understanding of SCI pathophysiology (Fig. 1, created with BioRender.com).

1. Preventing Ischemia

In the early stages of SCI, bleeding caused by disruption of local vasculature can lead to ischemic hypoxia within spinal grey matter. Hypoxia in these highly metabolically active structures leads to neuronal dysfunction, demyelination, and the initiation of apoptosis and necrosis [34]. Uncorrected ischemia can cause secondary damage due to the cumulative effects of cellular swelling and the initial macrophage-mediated inflammatory response which may last between 3–24 hours following the inciting event [34]. Furthermore, without proper blood flow, the highly vascular and regenerative astrocytic environment is less equipped to repair axonal and neuronal damage. Maintaining adequate perfusion systemically and within the remaining spinal vascular beds is essential for mitigating the pathological consequences of SCI.
States of neurogenic shock can further compound the ischemic burden. Prompt correction of hypotension with fluid resuscitation, transfusion, or traditional vasopressors can be difficult due to the disruption of local capillary auto-regulatory mechanisms. Agents including dopamine and midodrine have been attempted in clinical trials with varying levels of success and no high-level evidence supports the use of one pressor over another [35]. When compared with norepinephrine, dopamine maintained MAP to a similar degree but was associated with a higher intrathecal pressure (ITP) and consequentially decreased spinal cord perfusion pressure (SCPP); dopamine use was also associated with higher rates of vasopressor-associated complications [22]. A handful of case reports and small clinical trials of midodrine note successful acute maintenance of blood pressure and orthostatic symptoms with no serious adverse effects but lacked long-term follow-up [36]. Importantly, hypertension should be avoided in acute SCI to lower the risk of hyperemia and hemorrhage.
Cerebrospinal fluid (CSF) diversion has potential to alleviate ischemia caused by venous edematous leakage following SCI through the reduction of ITP. This strategy was extrapolated from the cranial trauma practice of ventriculostomy placement and the vascular surgery literature’s investigation of the use of lumbar drains in aortic dissection to support cord perfusion [37]. A preclinical study utilizing a pig SCI model previously demonstrated a significant and sustainable improvement in blood flow and SCPP with CSF drainage [38]. An early human study was undertaken with 22 patients randomized to undergo CSF drainage within 48 hours of injury versus not [39]. Lumbar drainage was not associated with significant side effects but also did not demonstrate definite neurologic benefit, though the authors cited a low power as one potential cause. Subsequent larger studies demonstrated more significant benefits with adequate safety profiles; however, a well-defined effect remains to be reproducibly demonstrated [40]. Patch duraplasty is another method of lowering ITP on the injured cord and can be performed during initial surgical decompression [41]. One study found that laminectomy with patch duraplasty resulted in lower ITP and improved spinal perfusion pressure compared with laminectomy alone in acute SCI; additionally, ASIA impairment and bowel/bladder function was improved in the duraplasty group. However, low power contributed to a lack of statistically significant differences in clinical outcomes [42]. These studies underscore the overall difficulty in performing well powered and large-scale human trials on acute SCI.
Hyperbaric oxygen (HBO), when administered early, may counteract spinal cord ischemia and hypoxia, potentially reducing apoptosis, oxidative damage, inflammation, and edema, and promoting angiogenesis and autophagy. Recent clinical studies have initiated therapy within 9–20 hours of injury with varying time-courses, demonstrating varying degrees of neurologic improvement [43]. Technical parameters (onset, duration, frequency, and pressure) and evaluation of benefits in more heterogenous samples of SCI presentations remain to be investigated. Two clinical studies are ongoing in China (NCT03112941) and Austria (NCT03101982).

2. Minimizing Inflammation-Induced Secondary Damage

The inflammatory cascade occurring hours to days following the primary of insult of SCI likely contributes to worsened neurological outcomes [34]. This secondary damage acts on the injured spinal cord through cellular and systemic factors that worsen compression, ischemia, and scarring [44]. Synthesis of proinflammatory prostaglandins and cytokines, calcium-dependent nitric oxide, opioid peptides, and necroptotic factors have been shown to augment inflammatory secondary damage in the acute stages of injury. To address this component of the injury, several commonplace pharmacological agents have demonstrated efficacy in maintaining blood flow and protecting from cellular damage in preclinical studies [34]. Cyclooxygenase inhibitors including ibuprofen and meclofenamate as well as nonselective opioid antagonists such as naloxone have been shown to improve blood flow and enhance functional results in animal spinal cord contusion models [45-48]. Still, few trials have attempted to establish the effects of these agents in human SCI. In 1990, NASCIS II found no neurologic benefit with the administration of naloxone throughout the first 24 hours after SCI compared to placebo, although those recommendations are being revisited more broadly [26]. A phase-I clinical trial involving 12 patients investigating the use of ibuprofen administration in acute SCI is in progress (NCT02096913).
Minocycline, a tetracycline antibiotic, has demonstrated multiple neuroprotective effects against secondary injury progression in SCI [34]. Minocycline impedes generation of proinflammatory cytokines (IL-1beta, tumor-necrosis factor [TNF]-alpha, cyclooxygenase-2), hinders expression of proapoptotic caspase-1/-3, and inhibits inducible nitric oxide synthase [49,50]. Successful preclinical studies showed enhanced tissue sparing and motor recovery within 3–4 weeks following SCI in mice [51,52]. A phase-II clinical trial demonstrated that patients with acute incomplete cervical SCI can benefit from early minocycline administration [53]. The study reported a 14‐point ASIA motor score recovery in patients receiving minocycline administration for 7 days following injury compared with placebo [53].
Spinal cooling is a nonpharmacologic method that has been explored for its potential to ameliorate biochemical secondary injury. One study utilized a combination of surgical decompression, steroid administration, and regional hypothermia in 20 patients and saw an improvement from initial ASIA-A impairment in 13 patients [54]. The lack of randomized comparative control arms in this study and the few other case reports/small clinical series available limit the generalizability of spinal cord cooling in acute SCI; however, the promising initial results and absence of reported adverse effects may warrant further study [12].

3. Controlling Cytotoxic Response

The acute cytotoxic response to SCI is another targetable pathomechanism. Calcium, sodium, and potassium ion dysregulation as well as release of high levels of glutamate from damaged neural tissue can cause excitotoxicity and oxidative damage. Signaling cascades that lead to ROS formation and lipid peroxidation increase the likelihood of neuronal cell death. Inhibiting free radical formation through various antioxidants including cyclosporin A, vitamins C, D, and E, selenium, lithium, polyethylene glycol-superoxide dismutase, and OXR1 gene-enhancing therapy, have all shown promise in maintaining cell viability in animal models [34]. One 16-participant, phase-I/II clinical trial of lithium in China (NCT01471613) is the first human trial to incorporate one of these agents as an adjunct in acute management strategies.
Uncontrolled activation of voltage-gated sodium channels has been hypothesized as an important step in the cytotoxic response leading to secondary spinal trauma. Riluzole is a benzothiazole that inhibits voltage-gated sodium channel-mediated glutamate release and recently gained approval for the treatment of amyotrophic lateral sclerosis. Previous animal studies have documented reduced neuronal loss and sensorimotor improvement with Riluzole administration following SCI, likely through inhibition of glutamate release and subsequent reduction of calcium-signaling induced apoptosis reduction [55]. A successful phase-I trial involving 36 patients found that 50 mg of Riluzole orally every 12 hours for 14 days enhanced motor recovery in cervical SCI at 3 months postinjury compared with a matched control group [55]. Similar motor enhancement was not found at 6-month follow-up. A transient increase in liver enzymes in the Riluzole group prompted questions of pharmacotoxicity, however its safety profile has yet to be presented in larger studies. The AO Spine North America Research Network is currently conducting the Riluzole in Acute Spinal Cord Injury Study, a multicenter randomized trial designed to evaluate the therapeutic application of Riluzole in acute SCI.
Antagonism of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid/ N-methyl-D-aspartate (NMDA) receptors has been postulated as a mechanism of reducing excitatory amino acid (glutamate) toxicity in the hyperacute phase of SCI. These neurotransmitters reach toxic levels within 15 minutes of injury and typically peak for less than 120 minutes [34]. Gacyclidine, an NMDA receptor antagonist, has been evaluated in a phase‐II double‐blind, randomized evaluation of 280 patients with SCI [56]. However, this study revealed no significant improvement in ASIA scores compared with placebo treatment. Still, other NMDA antagonists such as magnesium have been shown in preclinical studies to reduce excitotoxicity and inflammation. Organized phase-I/II clinical trials have yet to be completed examining this therapy in acute SCI.
Calcium-channel blockers (CCB) can significantly reduce the pathological influx of calcium following SCI and improve perfusion through regulation of vascular smooth muscle tone. The utility of nimodipine has been studied, most notably in a French clinical trial of 106 patients with SCI randomized to receive MPSS, nimodipine, MPSS and nimodipine, or placebo within 8 hours of injury for 7 days duration. Neither additional neurologic benefit nor significant adverse effects in the nimodipine treatment group versus control were noted [27]. Still, concerns for systemic hypotension in the context of injured auto-regulatory mechanisms in cervicothoracic SCI should prompt caution when considering CCBs during acute management. Finally, potassium channel antagonists such as the fast potassium channel blocker fampridine may have potential therapeutic benefit through improvement of axonal conduction in SCI and are currently being tested in phase-III trials [34].

4. Modulating the Immune Response

Immunomodulative therapies are currently under investigation for the treatment of acute SCI. Peripheral immune cells including macrophages, neutrophils, and T cells trigger an inflammatory response following injury that can last several days. Importantly, this response can cause cavitary lesion growth, further damaging surrounding spinal cord. Inflammatory cytokines including IL-1beta, TNF-alpha, and IL6 are produced in response to the injury and are followed by release of the neuroprotective IL-10 [34,57]. Microglia, the resident immune cells of the central nervous system, are known to have a central role in modulating this response [58]. One phase-I/II trial currently underway in China is examining the use of TNF-alpha monoclonal antibodies in acute SCI (NCT04988425). Enrollment is expected to reach 90 participants, and results are anticipated in late 2023.
Granulocyte-colony stimulating factor (G-CSF) is a cytokine glycoprotein that has increasingly been demonstrated to enhance neurogenesis and reduce TNF-alpha and IL1-beta expression levels at SCI sites [59]. Randomized phase-I/IIa studies have shown ASIA motor score improvement and a lack of significant adverse events with G-CSF therapy for acute SCI [60]. A phase-III clinical trial in Japan was recently completed with results to be published in the near future [61].

5. Nerve Regeneration Techniques

One of the most promising aims in experimental therapeutics for SCI remains establishing methods for neuronal and axonal regeneration. Current approaches focus on stem cell implantation, growth factor conditioning, neural growth inhibitor binding, tissue scaffolding, and neuromodulation. While these methods have historically been engineered to address chronic SCI, focusing regenerative efforts closer to the primary insult could theoretically expedite healing and functional recovery.

1) Stem cell implantation

The potential of stem cell therapy to induce or enhance endogenous neural regeneration and/or improve functional recovery has continued to undergo rigorous research. Stem cell therapies under investigation encompass autologous bone-marrow mononuclear cells (BM-MSCs)—including hematopoietic stem cells (HSCs) and mesenchymal stem cells—and other cell types including neural progenitor cells (NPCs), olfactory ensheathing cells (OECs), embryonic stem cells (ESCs), and induced pluripotent stem cells (iPSCs).
Bone-marrow derived mononuclear cells have the advantage of reduced immune rejection and possible synergism between HSCs and MSCs, improving angiogenesis and matrix repair [30]. On the other hand, BM-MSCs are difficult to purify and culture in vitro, and they possess limited differentiation potential [30]. Clinical trials of these therapies have shown promising but mixed results. After autologous HSC (CD-34+ and CD-133+) transplantation in 19 patients with complete SCI (ASIA-A), 37% of patients saw segmental sensory improvement (ASIA-B), 10% saw motor improvement (ASIA-C), and the remaining 53% saw no improvement (ASIA-A) within a 42–60 months follow-up period [62]. Relative to HSCs, MSCs possess higher neuronal differentiation potential, and small clinical trials have demonstrated safety and variable efficacy in improving spasticity, sensorimotor, and sphincter function in both complete and incomplete chronic SCI patients [30,63]. A meta-analysis of 328 patients demonstrated improved ASIA scores in 42% of cases after 1 year but noted the limited availability of clinical trial data and standardized methods [64]. Numerous clinical trials are ongoing, including a phase-II/III trial of MSCs in chronic ASIA-B patients by Pharmicell Co. (Seoul, Korea) and a trial of adipose-derived MSCs in ASIA-A, ASIA-B, and ASIA-C patients by Mayo Clinic. These trials were set to conclude in 2020 and 2023, respectively [65].
NPCs are native and multipotent and differentiate into neuronal-restricted precursors and glial-restricted precursors. NPC transplants readily integrate into the host milieu, induce axonal regeneration and extension, hasten remyelination, attenuate glial scar formation, and afford immunomodulation and neuroprotection [66]. A phase-I/IIa open-label nonrandomized controlled clinical trial did demonstrate AISA score improvement by at least 2 points in 5 of 19 NPC transplant patients, compared to 1 of 15 control patients [67]. Other clinical trials, such as an openlabel phase-I trial by Neuralstem (now Seneca BioPharma, Germantown, MD, USA), are ongoing. Challenges currently faced include difficult isolation of NPCs, the small number of therapy candidates, and the high cost of research [67].
OECs are glial cells that support olfactory receptor axonal growth into the olfactory bulb. Specifically, they express neurotrophins that promote neurogenesis and reduce risk of astrocyte hypertrophy. They are readily harvested autologously from the olfactory bulb and nasal mucosa and do not require patient immunosuppression. OECs are limited by inadequate source quantity and present differentiation and purification challenges similar to other regenerative options. Numerous clinical trials have been conducted, and a meta-analysis of 1,193 patients treated with OECs found no significant adverse effects, but methodological and technical concerns prevented determination of any significant efficacy as well [68]. Clinical trials in Poland (NCT03- 933072) and the United Kingdom (NCT02870426) are slated to conclude in 2023.
ESCs and iPSCs have similarly impressive differentiation potential, promising substantial efficacy; human ESCs are indeed capable of differentiation into NPCs and, subsequently, regionally specific neuronal subtypes [69]. Their pluripotency means ESCs and iPSCs do carry an associated genomic instability and risk of malignancy. The ethical stature of ESC use is also disputed. Non-human investigation of ESC- and iPSC-based treatment of SCI has demonstrated feasibility, but clinical trials are so far limited [70,71]. The Geron Corporation (Foster City, CA, USA) began a clinical trial of ESCs for SCI therapy in 2010, enrolling 5 patients before stopping the trial a year later. Not all patients enrolled had received a transplant [30]. Keio University in Tokyo, Japan, has received approval for a clinical trial of iPSC therapy for ASIA-A patients, which is ongoing [30,71].
Overall, while these data are promising in the quest to determine the best neuro-regenerative substrates in SCI, more work is required to establish efficacy and safety in the acute postinjury setting to determine long-term effects on functional recovery. Additionally, no head-to-head trials have as of yet been conducted; subsequent investigation should help to focus efforts on optimizing sourcing, delivery, and conditioning of transplanted cells.

2) Growth factor conditioning

Acidic fibroblast growth factor (aFGF) is a mitogenic, pluripotent heparin-binding protein that has been shown to enhance neuronal growth and mitigates scarring in SCI in numerous animal studies [72,73]. Human clinical trials have steadily increased in number since an initial case report demonstrating marked functional recovery of a patient with chronic SCI who was treated with 4 survival nerve grafts along with an aFGF fibrin glue and could ambulate independently with a walker at 2.5 years postinjury [74]. A different group conducted 2 more studies, 1 on 9 cervical SCI patients and a follow-up study with 60 patients (50/50 cervical and thoracolumbar SCI), and found that the use of aFGF fibrin glue resulted in significant enhancement of ASIA motor and sensory scores [75,76]. These observational studies were limited by their lack of randomization and control arms. Recombinant human aFGF (compound ES135) is currently undergoing a multicenter double blind, randomized placebo control phase-III study (NCT03229031). Hepatocyte growth factor (HGF) can enhance neuronal survival and decrease astroglial scar lesion size by increasing angiogenesis and axon regeneration [77]. Intrathecal recombinant human HGF injection has been shown to improve neurological status in rat studies [78]. Based on this, a phase-I/II trial of intrathecal recombinant HGF involving patients with acute cervical SCI found that all patients had improved Frankel scores at 72 hours [79].

3) Inhibiting neural growth inhibitors

Cethrin (VX-210) is a recombinant inhibitor of the Rho pathway which is involved in the regulation of cytoskeleton formation [30]. Inhibition of Rho has been shown in rodent thoracic contusion models to promote axonal outgrowth and improve motor function [80]. Following a phase-I/IIa clinical study that demonstrated the safety and tolerability of Cethrin for acute cervical and thoracic SCI in human patients, a double-blinded, placebo-controlled, multicenter trial (SPRING) has been developed to demonstrate therapeutic benefit, with preliminary results posted (NCT02669849).
Elezanumab is a monoclonal antibody that binds to and neutralizes repulsive guidance molecule A, an inhibitor of axonal growth and regulator of neuronal cell death [81]. A phase-II clinical trial is currently underway to evaluate the effects of Elezanumab in acute traumatic cervical SCI (NCT04295538).

4) Tissue scaffolding

Biomaterial tissue scaffolding is one promising method for facilitating SCI repair. Natural or synthetic biomaterial polymers have been shown in animal models to enhance nervous tissue regeneration by promoting survival and outgrowth of transplanted cells and providing an environment to concentrate neurotrophic growth factors [12,30]. Limited efficacy has been redemonstrated in humans. A large multicenter trial entitled INSPIRE launched in 2014 to determine the benefit of a synthetic polymer scaffold poly(lactic-co-glycolic acid)-b-poly(L-lysine) in subjects with thoracic ASIA-A impairment, has completed enrollment and expected to conclude in 2024 (NCT02138110). Multiple clinical trials led by the Chinese Academy of Sciences are investigating the application of NeuroRegen, a collagen scaffold, in concert with other experimental strategies including stem cell therapy and epidural stimulation.
Gangliosides including the glycosphingolipid ganglioside-1 (GM-1) are complex acidic glycolipids that constitute a major component of the cell membrane. Experimental evidence in animal studies has shown potential in the regeneration and growth of damaged nervous tissue leading to functional improvement [82]. One phase-II trial showed enhanced 1-year ASIA motor score improvement following daily administration of GM-1 for 18–32 days following injury [68]. However, meta-analyses evaluating potential therapeutic benefits of GM-1 across studies failed to support its widespread use for SCI [83]. Still, it has been suggested that studies with more optimized methodology (i.e., potentializing GM-1 administration with HBO and/or refining outcome measurement) may be warranted with this potential therapeutic [83].

5) Neuromodulation

Epidural stimulation (ES) is an emerging treatment strategy that has demonstrated promise in the treatment of chronic SCI in both animals and humans [84-86]. It is theorized that oscillating electrical fields may stimulate neuronal growth and remyelination and excite remaining neuronal networks at the site of the lesion [87]. Individual patients with predominantly lumbar injuries have demonstrated motor improvement with ES, including partial and full weight-bearing and overground ambulation, even with complete injuries [88,89]. Similarly promising results of upper limb motor recovery have been observed [89]. Notably, a recent study demonstrated activity-specific stimulation programs that enabled 3 individuals with complete sensorimotor paralysis to regain trunk and leg motor functions within 1 day of stimulation [90]. Several clinical trials targeting these broader functional metrics (e.g., volitional movement, autonomic function, bladder training, and sensory feedback) are underway.

FUTURE PERSPECTIVES

Table 1 summarizes the current and developing investigational therapeutic tools for treating acute SCI. Numerous methods are being developed in animal models that may soon be tested in clinical trials designed to demonstrate functional benefit in SCI. Necroptosis is a caspase-independent mechanism of programmed cell death that occurs following cellular injury in which cellular contents are not packaged into apoptotic bodies and instead leak into the extracellular space, promoting a proinflammatory state through triggering of innate and adaptive immune systems [91]. The process is dependent on receptor-interacting serine/threonine kinase 1 and 3 and studies have shown that it may be one of the primary forms of cell death following traumatic SCI [91]. Inhibition of this pathway is suspected to attenuate secondary damage and minimize cell death [92].
Autologous omental transplantation has been proposed as a method to introduce blood- and lymph-rich tissue to injured spinal cord tissue [93]. Relatively small human trials in the 1990s showed continued viability of omental grafts in chronic SCI patients but failed to demonstrate objective functional improvement, and interest in its SCI treatment potential fell flat for a time [94]. However, omental transplant therapy has progressed in other clinical settings, including flap transposition on the brain surface for neovascularization and neurotrophic effects in epileptic or ischemic brain injury [95]. Interest in omental angiogenesis as a treatment for SCI has revived in recent years, and trials in rats have improved neural preservation, reduced injury cavity, and promoted neovascularization of the injured site [93].
A newer addition to the regenerative cellular therapies discussed in the literature is peripheral nerve derived stem cells (PNSCs). Spheroid forms of PNSCs have demonstrated therapeutic potential through neurotrophic factor release and extracellular matrix expression, leading to significant functional recovery, neuronal regeneration, and neuropathic pain reduction in animal models [96].
Estrogen-based therapies are also an emerging avenue or research. Estradiol is known to provide CNS neuroprotection in animal models of SCI, traumatic brain injury, and ischemic brain injury [97]. The therapeutic capability of a third-generation selective estrogen receptor modulator, bazedoxifine, was studied in rats [98]. Bazedoxifine was shown to suppress inflammatory response and promote remyelination by inhibiting the mitogenactivated protein kinase/nuclear factor-kappa B (NF-κB) pathway and enhancing oligodendrocyte precursor cell differentiation and oligodendrocyte proliferation. There are complex and profound implications of estrogen therapy due to its natural maintenance and alteration of normal physiology. Still, there is hope that estrogen therapies may play some role in SCI treatment.
Entinostat is a class I histone deacetylase (HDAC) inhibitor directed primarily against HDAC1 and HDAC3. HDAC inhibitors have been shown to inhibit NF-κB mediated microglial activation, thus reducing SCI-induced inflammation [99]. Treatment of a mouse SCI model with Entinostat resulted in improved grip strength, Basso Mouse Scale score for locomotion, spinal edema, cell death, and local NLRP3 inflammasome activation [99].
Another method of counteracting inflammation induced by SCI may be found in IgM’s homeostatic role against IgG autoimmune response. One study found that IgM-knockout mice with induced C6–7 SCI showed significantly greater impairment in neurobehavioral recovery compared to their wild-type counterparts, including deteriorated coordination and reduced fore- and hind-limb swing speed and print-area, which serve as gait parameters [100]. IgM-knockout mice also exhibited increased lesion size, less white matter sparing, and enhanced deposition of complement-fixing IgG antibodies in the spinal cord. These newer methods will continue to be studied closely by the scientific community hoping to grow the armamentarium available to treat this multifaceted disorder.

CONCLUSION

Emerging strategies for managing SCI in the acute period stem from an evolving understanding of the pathophysiology of the disorder and focus on preventing ischemia, reducing secondary injury due to inflammation and necroptosis, modulating the cytotoxic and immune response, and promoting cellular regeneration. There is significant research ongoing in preclinical and clinical studies examining a wide array of surgical and non-surgical therapies. A major hurdle to wide acceptance of any of the emerging treatments is designing an appropriate clinical trial to test them. Continued translational research on these methods will improve the feasibility of bench-to-bedside innovations in treating patients with acute SCI.

NOTES

Conflict of Interest

The authors have nothing to disclose.

Funding/Support

This study received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.

Author Contribution

Conceptualization: RG, MP, AR; Data curation: RG, DS; Funding acquisition: AR; Methodology: RG; Project administration: AR; Visualization: RG; Writing - original draft: RG, DS; Writing - review & editing: RG, DS, MP, JG, AF, AR.

Fig. 1.
Emerging therapies for acute spinal cord injury. CSF, cerebrospinal fluid; COX, cyclooxygenase; NMDA, N-methyl-D-aspartate; AMPA, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid; TNF, tumor-necrosis factor; IL, interleukin; G-CSF, granulocyte-colony stimulating factor.
ns-2244176-088f1.jpg
Table 1.
Summary of investigational therapeutic tools and their corresponding evidence for management of acute SCI
Therapeutic goal Investigational tool Investigational status/current findings
Prevent ischemia Vasopressors Norepinephrine, dopamine, midodrine
Limited evidence supporting one pressor over another
CSF diversion Lumbar puncture & patch duraplasty may improve SCPP and ASIA scores in low-power studies
Hyperbaric oxygen Optimized technical parameters remain undefined
Minimize inflammatory damage COX-inhibitors, nonselective opioid antagonists Improved blood flow and functional status in animals.
Human clinical trials underway
Minocycline Phase-II clinical trials demonstrated improved functional status
Spinal cooling ASIA motor score improvement in small, non-RCT trials
Block cytotoxic response Free radical inhibition Few studies of antioxidant therapy in humans
Adjunctive lithium is being studied in a phase-I/II trial.
Riluzole Small, phase-I trial showed short term motor improvement.
Transaminase elevation raises concerns for pharmacotoxicity
Multicenter RCT (RISCIS) underway
AMPA/NMDA antagonists Phase-II double‐blind, randomized trial of gacyclidine showed no significant benefit versus placebo
Other antagonists (e.g., magnesium) have promising preclinical results
Ion channel modulators Nimodipine (CCB): No apparent benefit or adverse effects in RCT. Concern for systemic hypotension
Fampridine (K-channel blocker): Phase-III trial underway
Immunomodulation TNF-alpha mAb Phase-I/II clinical trial underway
G-CSF Phase-I/II trials showed ASIA motor score improvement
Phase III trial underway
Nerve regeneration Stem cell therapies Sourcing, potency, and clinical benefit vary by cell type.
Numerous clinical trials are underway
Future efforts should optimize sourcing, delivery, and conditioning of transplant cells
Growth factor conditioning aFGF: Phase I/II clinical trials demonstrate ASIA motor and sensory improvement. Phase-III trial underway
HGF: Phase I/II trials showed improvement in Frankel grade
Neural growth inhibitor blockers Cethrin: Phase-I/IIa successful; Phase-IIb/III (SPRING trial) underway
Elezanumab: Phase-II trial underway
Tissue scaffolding PLGA: Multicenter trial (INSPIRE) underway
NeuroRegen: Multiple trials underway
GM-1: Unclear benefit; clinical methodologies need optimization
Neuromodulation (ES) Small studies demonstrate promising functional recovery.
Several clinical trials underway
Future perspectives Necroptosis inhibition (RIPK-1/3 inhibition) Under preclinical investigation
Omental transplant
Peripheral nerve derived stem cell therapy
Selective estrogen receptor modulator therapy
HDAC1/3 inhibition (Entinostat)
IgM therapy

SCI, spinal cord injury; CSF, cerebrospinal fluid; SCPP, spinal cord perfusion pressure; ASIA, American Spine Injury Association; COX, cyclooxygenase; RCT, randomized clinical trial; RISCIS, Riluzole in Spinal Cord Injury Study; AMPA, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid; NMDA, N-methyl-D-aspartate; CCB, calcium-channel blocker; K, potassium; TNF, tumor-necrosis factor; mAb, monoclonal antibody; G-CSF, granulocyte-colony stimulating factor; aFGF, acidic fibroblast growth factor; HGF, hepatocyte growth factor; PLGA, poly (lactic-co-glycolic acid)-b-poly(L-lysine); RIPK, receptor-interacting protein kinase; HDAC, histone deacetylase; ES, epidural stimulation; GM-1, ganglioside-1.

REFERENCES

1. Hadley MN, Walters BC, Aarabi B, et al. Clinical assessment following acute cervical spinal cord injury. Neurosurgery 2013;72 Suppl 2:40-53.
crossref pmid pdf
2. Roberts TT, Leonard GR, Cepela DJ. Classifications In Brief: American Spinal Injury Association (ASIA) Impairment Scale. Clin Orthop Relat Res 2017;475:1499-504.
crossref pmid pmc pdf
3. Singh A, Tetreault L, Kalsi-Ryan S, et al. Global prevalence and incidence of traumatic spinal cord injury. Clin Epidemiol 2014;6:309-31.
pmid pmc
4. Ackery A, Tator C, Krassioukov A. A global perspective on spinal cord injury epidemiology. J Neurotrauma 2004;21:1355-70.
crossref pmid
5. Simpson LA, Eng JJ, Hsieh JT, et al. The health and life priorities of individuals with spinal cord injury: a systematic review. J Neurotrauma 2012;29:1548-55.
crossref pmid pmc
6. Furlan JC, Gulasingam S, Craven BC. The Health Economics of the spinal cord injury or disease among veterans of war: A systematic review. J Spinal Cord Med 2017;40:649-64.
crossref pmid pmc
7. Moghaddamjou A, Fehlings MG. The beneficial effect of early surgical decompression for acute spinal cord injury: time is spine. Neurospine 2021;18:20-2.
crossref pmid pmc pdf
8. 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.
crossref pmid pmc
9. Kumar N, Osman A, Chowdhury JR. Traumatic spinal cord injuries. J Clin Orthop Trauma 2017;8:116-24.
crossref pmid pmc
10. Hadley MN, Walters BC, Grabb PA, et al. Cervical spine immobilization before admission to the hospital. Neurosurgery 2002;50(3 Suppl):S7-17.
crossref pmid
11. Zileli M, Osorio-Fonseca E, Konovalov N, et al. Early management of cervical spine trauma: WFNS Spine Committee Recommendations. Neurospine 2020;17:710-22.
crossref pmid pmc pdf
12. Wang TY, Park C, Zhang H, et al. Management of acute traumatic spinal cord injury: a review of the literature. Front Surg 2021;8:698736.
crossref pmid pmc
13. Snyder R, Verla T, Ropper AE. Practical application of recent advances in diagnostic, prognostic, and therapeutic modalities for spinal cord injury. World Neurosurg 2020;136:330-6.
crossref pmid
14. Fehlings MG, Tetreault LA, Wilson JR, et al. A clinical practice guideline for the management of acute spinal cord injury: introduction, rationale, and scope. Global Spine J 2017;7(3 Suppl):84S-94S.
crossref pmid pmc pdf
15. Mputu Mputu P, Beauséjour M, Richard-Denis A, et al. Early predictors of neurological outcomes after traumatic spinal cord injury: a systematic review and proposal of a conceptual framework. Am J Phys Med Rehabil 2021;100:700-11.
crossref pmid
16. Parthiban J, Zileli M, Sharif SY. Outcomes of spinal cord injury: WFNS Spine Committee Recommendations. Neurospine 2020;17:809-19.
crossref pmid pmc pdf
17. Qiu Y, Chen Y, Xie Y, et al. Comparative analysis of the efficacy of early and late surgical intervention for acute spinal cord injury: a systematic review and meta-analysis based on 16 studies. Int J Surg 2021;94:106098.
crossref pmid
18. Chikuda H, Koyama Y, Matsubayashi Y, et al. Effect of early vs delayed surgical treatment on motor recovery in incomplete cervical spinal cord injury with preexisting cervical stenosis: a randomized clinical trial. JAMA Netw Open 2021;4:e2133604.
crossref pmid pmc
19. Fehlings MG, Vaccaro A, Wilson JR, et al. Early versus delayed decompression for traumatic cervical spinal cord injury: results of the Surgical Timing in Acute Spinal Cord Injury Study (STASCIS). PLoS One 2012;7:e32037.
crossref pmid pmc
20. Dvorak MF, Noonan VK, Fallah N, et al. The influence of time from injury to surgery on motor recovery and length of hospital stay in acute traumatic spinal cord injury: an observational Canadian cohort study. J Neurotrauma 2015;32:645-54.
crossref pmid pmc
21. Badhiwala JH, Wilson JR, Witiw CD, et al. The influence of timing of surgical decompression for acute spinal cord injury: a pooled analysis of individual patient data. Lancet Neurol 2021;20:117-26.
crossref pmid
22. Lee YS, Kim KT, Kwon BK. Hemodynamic management of acute spinal cord injury: a literature review. Neurospine 2021;18:7-14.
crossref pmid pmc pdf
23. Hawryluk G, Whetstone W, Saigal R, et al. Mean arterial blood pressure correlates with neurological recovery after human spinal cord injury: analysis of high frequency physiologic data. J Neurotrauma 2015;32:1958-67.
crossref pmid pmc
24. Hatton GE, Mollett PJ, Du RE, et al. High tidal volume ventilation is associated with ventilator-associated pneumonia in acute cervical spinal cord injury. J Spinal Cord Med 2021;44:775-81.
crossref pmid pmc
25. Redshaw JD, Lenherr SM, Elliott SP, et al. Protocol for a randomized clinical trial investigating early sacral nerve stimulation as an adjunct to standard neurogenic bladder management following acute spinal cord injury. BMC Urol 2018;18:72.
crossref pmid pmc pdf
26. Bracken MB, Shepard MJ, Holford TR, et al. Administration of methylprednisolone for 24 or 48 hours or tirilazad mesylate for 48 hours in the treatment of acute spinal cord injury. Results of the Third National Acute Spinal Cord Injury Randomized Controlled Trial. National Acute Spinal Cord Injury Study. JAMA 1997;277:1597-604.
crossref pmid
27. Pointillart V, Petitjean ME, Wiart L, et al. Pharmacological therapy of spinal cord injury during the acute phase. Spinal Cord 2000;38:71-6.
crossref pmid pdf
28. Matsumoto T, Tamaki T, Kawakami M, et al. Early complications of high-dose methylprednisolone sodium succinate treatment in the follow-up of acute cervical spinal cord injury. Spine (Phila Pa 1976) 2001;26:426-30.
crossref pmid
29. Cabrera-Aldana EE, Ruelas F, Aranda C, et al. Methylprednisolone administration following spinal cord injury reduces aquaporin 4 expression and exacerbates edema. Mediators Inflamm 2017;2017:4792932.
crossref pmid pmc pdf
30. Takami T, Shimokawa N, Parthiban J, et al. Pharmacologic and regenerative cell therapy for spinal cord injury: WFNS Spine Committee Recommendations. Neurospine 2020;17:785-96.
crossref pmid pmc pdf
31. Hicks AL, Martin KA, Ditor DS, et al. Long-term exercise training in persons with spinal cord injury: effects on strength, arm ergometry performance and psychological well-being. Spinal Cord 2003;41:34-43.
crossref pmid pdf
32. Post MW, van Leeuwen CM. Psychosocial issues in spinal cord injury: a review. Spinal Cord 2012;50:382-9.
crossref pmid pdf
33. Dhall SS, Hadley MN, Aarabi B, et al. Nutritional support after spinal cord injury. Neurosurgery 2013;72 Suppl 2:255-9.
crossref pmid pdf
34. Zhang Y, Al Mamun A, Yuan Y, et al. Acute spinal cord injury: pathophysiology and pharmacological intervention (review). Mol Med Rep 2021;23:417.
crossref pmid pmc
35. Ploumis A, Yadlapalli N, Fehlings MG, et al. A systematic review of the evidence supporting a role for vasopressor support in acute SCI. Spinal Cord 2010;48:356-62.
crossref pmid pdf
36. Mojtahedzadeh M, Taghvaye-Masoumi H, Najafi A, et al. Management of hypotension and bradycardia caused by spinal cord injury. The usefulness of midodrine and methylxanthines. Iran J Pharm Res 2019;18:2131-5.
pmid pmc
37. Khan NR, Smalley Z, Nesvick CL, et al. The use of lumbar drains in preventing spinal cord injury following thoracoabdominal aortic aneurysm repair: an updated systematic review and meta-analysis. J Neurosurg Spine 2016;25:383-93.
crossref pmid
38. Martirosyan NL, Kalani MY, Bichard WD, et al. Cerebrospinal fluid drainage and induced hypertension improve spinal cord perfusion after acute spinal cord injury in pigs. Neurosurgery 2015;76:461-8. discussion 468-9.
crossref pmid
39. Kwon BK, Curt A, Belanger LM, et al. Intrathecal pressure monitoring and cerebrospinal fluid drainage in acute spinal cord injury: a prospective randomized trial. J Neurosurg Spine 2009;10:181-93.
crossref pmid
40. Squair JW, Bélanger LM, Tsang A, et al. Spinal cord perfusion pressure predicts neurologic recovery in acute spinal cord injury. Neurology 2017;89:1660-7.
crossref pmid
41. Tykocki T, Poniatowski Ł, Czyż M, et al. Intraspinal pressure monitoring and extensive duroplasty in the acute phase of traumatic spinal cord injury: a systematic review. World Neurosurg 2017;105:145-52.
crossref pmid
42. Phang I, Werndle MC, Saadoun S, et al. Expansion duroplasty improves intraspinal pressure, spinal cord perfusion pressure, and vascular pressure reactivity index in patients with traumatic spinal cord injury: injured spinal cord pressure evaluation study. J Neurotrauma 2015;32:865-74.
crossref pmid pmc
43. Patel NP, Huang JH. Hyperbaric oxygen therapy of spinal cord injury. Med Gas Res 2017;7:133-43.
crossref pmid pmc
44. Schwartz G, Fehlings MG. Secondary injury mechanisms of spinal cord trauma: a novel therapeutic approach for the management of secondary pathophysiology with the sodium channel blocker riluzole. Prog Brain Res 2002;137:177-90.
pmid
45. Chaves RHF, Souza CC, Furlaneto IP, et al. Influence of tramadol on functional recovery of acute spinal cord injury in rats. Acta Cir Bras 2018;33:1087-94.
crossref pmid
46. Harada N, Taoka Y, Okajima K. Role of prostacyclin in the development of compression trauma-induced spinal cord injury in rats. J Neurotrauma 2006;23:1739-49.
crossref pmid
47. Young W, Flamm ES, Demopoulos HB, et al. Effect of naloxone on posttraumatic ischemia in experimental spinal contusion. J Neurosurg 1981;55:209-19.
crossref pmid
48. Winkler T, Sharma HS, Stålberg E, et al. Opioid receptors influence spinal cord electrical activity and edema formation following spinal cord injury: experimental observations using naloxone in the rat. Neurosci Res 1994;21:91-101.
crossref pmid
49. Scholz R, Sobotka M, Caramoy A, Stempfl T, Moehle C, Langmann T. Minocycline counter-regulates pro-inflammatory microglia responses in the retina and protects from degeneration. J Neuroinflammation 2015;12:209.
crossref pmid pmc
50. Tikka TM, Koistinaho JE. Minocycline provides neuroprotection against N-methyl-D-aspartate neurotoxicity by inhibiting microglia. J Immunol 2001;166:7527-33.
crossref pmid
51. Wells JE, Hurlbert RJ, Fehlings MG, et al. Neuroprotection by minocycline facilitates significant recovery from spinal cord injury in mice. Brain 2003;126(Pt 7):1628-37.
crossref pmid
52. Squair JW, Ruiz I, Phillips AA, et al. Minocycline reduces the severity of autonomic dysreflexia after experimental spinal cord injury. J Neurotrauma 2018;35:2861-71.
crossref pmid
53. Casha S, Zygun D, McGowan MD, et al. Results of a phase II placebo-controlled randomized trial of minocycline in acute spinal cord injury. Brain 2012;135(Pt 4):1224-36.
crossref pmid
54. Hansebout RR, Hansebout CR. Local cooling for traumatic spinal cord injury: outcomes in 20 patients and review of the literature. J Neurosurg Spine 2014;20:550-61.
crossref pmid
55. Nagoshi N, Nakashima H, Fehlings MG. Riluzole as a neuroprotective drug for spinal cord injury: from bench to bedside. Molecules 2015;20:7775-89.
crossref pmid pmc
56. Kim YH, Ha KY, Kim SI. Spinal cord injury and related clinical trials. Clin Orthop Surg 2017;9:1-9.
crossref pmid pmc pdf
57. Thompson CD, Zurko JC, Hanna BF, et al. The therapeutic role of interleukin-10 after spinal cord injury. J Neurotrauma 2013;30:1311-24.
crossref pmid
58. Brockie S, Hong J, Fehlings MG. The role of microglia in modulating neuroinflammation after spinal cord injury. Int J Mol Sci 2021;22:9706.
crossref pmid pmc
59. Nishio Y, Koda M, Kamada T, et al. Granulocyte colony-stimulating factor attenuates neuronal death and promotes functional recovery after spinal cord injury in mice. J Neuropathol Exp Neurol 2007;66:724-31.
crossref pmid
60. Derakhshanrad N, Saberi H, Yekaninejad MS, et al. Granulocyte-colony stimulating factor administration for neurological improvement in patients with postrehabilitation chronic incomplete traumatic spinal cord injuries: a double-blind randomized controlled clinical trial. J Neurosurg Spine 2018;29:97-107.
crossref pmid
61. Koda M, Hanaoka H, Fujii Y, et al. Randomized trial of granulocyte colony-stimulating factor for spinal cord injury. Brain 2021;144:789-99.
pmid pmc
62. Al-Zoubi A, Jafar E, Jamous M, et al. Transplantation of purified autologous leukapheresis-derived CD34+ and CD133+ stem cells for patients with chronic spinal cord injuries: longterm evaluation of safety and efficacy. Cell Transplant 2014;23 Suppl 1:S25-34.
crossref pmid pdf
63. Vaquero J, Zurita M, Rico MA, et al. Intrathecal administration of autologous mesenchymal stromal cells for spinal cord injury: Safety and efficacy of the 100/3 guideline. Cytotherapy 2018;20:806-19.
pmid
64. Aghayan HR, Arjmand B, Yaghoubi M, et al. Clinical outcome of autologous mononuclear cells transplantation for spinal cord injury: a systematic review and meta-analysis. Med J Islam Repub Iran 2014;28:112.
pmid pmc
65. Ahuja CS, Mothe A, Khazaei M, et al. The leading edge: Emerging neuroprotective and neuroregenerative cell-based therapies for spinal cord injury. Stem Cells Transl Med 2020;9:1509-30.
crossref pmid pmc pdf
66. Fischer I, Dulin JN, Lane MA. Transplanting neural progenitor cells to restore connectivity after spinal cord injury. Nat Rev Neurosci 2020;21:366-83.
crossref pmid pmc pdf
67. Shin JC, Kim KN, Yoo J, et al. 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.
crossref pmid pmc pdf
68. Badhiwala JH, Ahuja CS, Fehlings MG. Time is spine: a review of translational advances in spinal cord injury. J Neurosurg Spine 2018;30:1-18.
crossref pmid
69. Ronaghi M, Erceg S, Moreno-Manzano V, et al. Challenges of stem cell therapy for spinal cord injury: human embryonic stem cells, endogenous neural stem cells, or induced pluripotent stem cells? Stem Cells 2010;28:93-9.
crossref pmid pdf
70. Keirstead HS, Nistor G, Bernal G, et al. Human embryonic stem cell-derived oligodendrocyte progenitor cell transplants remyelinate and restore locomotion after spinal cord injury. J Neurosci 2005;25:4694-705.
crossref pmid pmc
71. Nagoshi N, Tsuji O, Nakamura M, et al. Cell therapy for spinal cord injury using induced pluripotent stem cells. Regen Ther 2019;11:75-80.
crossref pmid pmc
72. Ko CC, Tu TH, Wu JC, et al. Acidic fibroblast growth factor in spinal cord injury. Neurospine 2019;16:728-38.
crossref pmid pmc pdf
73. Zhou Y, Wang Z, Li J, et al. Fibroblast growth factors in the management of spinal cord injury. J Cell Mol Med 2018;22:25-37.
crossref pmid pmc pdf
74. Ko CC, Tu TH, Wu JC, et al. Functional improvement in chronic human spinal cord injury: Four years after acidic fibroblast growth factor. Sci Rep 2018;8:12691.
crossref pmid pmc pdf
75. Wu JC, Huang WC, Tsai YA, et al. Nerve repair using acidic fibroblast growth factor in human cervical spinal cord injury: a preliminary phase I clinical study. J Neurosurg Spine 2008;8:208-14.
crossref pmid
76. Wu JC, Huang WC, Chen YC, et al. Acidic fibroblast growth factor for repair of human spinal cord injury: a clinical trial. J Neurosurg Spine 2011;15:216-27.
crossref pmid
77. Kitamura K, Iwanami A, Nakamura M, et al. Hepatocyte growth factor promotes endogenous repair and functional recovery after spinal cord injury. J Neurosci Res 2007;85:2332-42.
crossref pmid
78. Kitamura K, Fujiyoshi K, Yamane J, et al. Human hepatocyte growth factor promotes functional recovery in primates after spinal cord injury. PLoS One 2011;6:e27706.
crossref pmid pmc
79. Kitamura K, Nagoshi N, Tsuji O, et al. Application of hepatocyte growth factor for acute spinal cord injury: the road from basic studies to human treatment. Int J Mol Sci 2019;20:1054.
crossref pmid pmc
80. Dergham P, Ellezam B, Essagian C, et al. Rho signaling pathway targeted to promote spinal cord repair. J Neurosci 2002;22:6570-7.
crossref pmid pmc
81. Jacobson PB, Goody R, Lawrence M, et al. Elezanumab, a human anti-RGMa monoclonal antibody, promotes neuroprotection, neuroplasticity, and neurorecovery following a thoracic hemicompression spinal cord injury in non-human primates. Neurobiol Dis 2021;155:105385.
crossref pmid
82. Geisler FH, Dorsey FC, Coleman WP. Recovery of motor function after spinal-cord injury--a randomized, placebocontrolled trial with GM-1 ganglioside. N Engl J Med 1991;324:1829-38.
crossref pmid
83. Marcon RM, Cristante AF, de Barros Filho TE, et al. Potentializing the effects of GM1 by hyperbaric oxygen therapy in acute experimental spinal cord lesion in rats. Spinal Cord 2010;48:808-13.
crossref pmid pdf
84. Angeli CA, Boakye M, Morton RA, et al. Recovery of overground walking after chronic motor complete spinal cord injury. N Engl J Med 2018;379:1244-50.
crossref pmid
85. Selfslagh A, Shokur S, Campos DSF, et al. Non-invasive, braincontrolled functional electrical stimulation for locomotion rehabilitation in individuals with paraplegia. Sci Rep 2019;9:6782.
crossref pmid pmc pdf
86. Jack AS, Hurd C, Martin J, et al. Electrical stimulation as a tool to promote plasticity of the injured spinal cord. J Neurotrauma 2020;37:1933-53.
crossref pmid pmc
87. Eisdorfer JT, Smit RD, Keefe KM, et al. Epidural electrical stimulation: a review of plasticity mechanisms that are hypothesized to underlie enhanced recovery from spinal cord injury with stimulation. Front Mol Neurosci 2020;13:163.
crossref pmid pmc
88. Krucoff MO, Gramer R, Lott D, et al. Spinal cord stimulation and rehabilitation in an individual with chronic complete L1 paraplegia due to a conus medullaris injury: motor and functional outcomes at 18 months. Spinal Cord Ser Cases 2020;6:96.
crossref pmid pmc pdf
89. Choi EH, Gattas S, Brown NJ, et al. Epidural electrical stimulation for spinal cord injury. Neural Regen Res 2021;16:2367-75.
crossref pmid pmc
90. Rowald A, Komi S, Demesmaeker R, et al. Activity-dependent spinal cord neuromodulation rapidly restores trunk and leg motor functions after complete paralysis. Nat Med 2022;28:260-71.
crossref pmid pdf
91. Fiani B, Kondilis A, Soula M, et al. Novel methods of necroptosis inhibition for spinal cord injury using translational research to limit secondary injury and enhance endogenous repair and regeneration. Neurospine 2021;18:261-70.
crossref pmid pmc pdf
92. Wang Y, Wang J, Wang H, et al. Necrosulfonamide attenuates spinal cord injury via necroptosis inhibition. World Neurosurg 2018;114:e1186-91.
crossref pmid
93. Fay LY, Lin YR, Liou DY, et al. The application of an omentum graft or flap in spinal cord injury. Int J Mol Sci 2021;22:7930.
crossref pmid pmc
94. Clifton GL, Donovan WH, Dimitrijevic MM, et al. Omental transposition in chronic spinal cord injury. Spinal Cord 1996;34:193-203.
crossref pmid pdf
95. Di Nicola V. Omentum a powerful biological source in regenerative surgery. Regen Ther 2019;11:182-91.
crossref pmid pmc
96. Lee HL, Yeum CE, Lee H, et al. Peripheral nerve-derived stem cell spheroids induce functional recovery and repair after spinal cord injury in rodents. Int J Mol Sci 2021;22:4141.
crossref pmid pmc
97. Raghava N, Das BC, Ray SK. Neuroprotective effects of estrogen in CNS injuries: insights from animal models. Neurosci Neuroecon 2017;6:15-29.
crossref pmid pmc pdf
98. Kim Y, Roh EJ, Joshi HP, et al. Bazedoxifene, a selective estrogen receptor modulator, promotes functional recovery in a spinal cord injury rat model. Int J Mol Sci 2021;22:11012.
crossref pmid pmc
99. Dai C, Liu B, Peng B, et al. Entinostat improves motor function and neuronal damage via downregulating NLRP3 inflammasome activation after spinal cord injury. Front Pharmacol 2021;12:774539.
crossref pmid pmc
100. Ulndreaj A, LVidaliu PM, Forgione N, et al. IgM immunoglobulin influences recovery after cervical spinal cord injury by modulating the IgG autoantibody response. eNeuro 2021;8:ENEURO.0491-19.2021.
crossref pmid pmc


Editorial Office
CHA University, CHA School of Medicine Bundang Medical Center
59 Yatap-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13496, Korea
Tel: +82-31-780-1924  Fax: +82-31-780-5269  E-mail: support@e-neurospine.org
The Korean Spinal Neurosurgery Society
#407, Dong-A Villate 2nd Town, 350 Seocho-daero, Seocho-gu, Seoul 06631, Korea
Tel: +82-2-585-5455  Fax: +82-2-2-523-6812  E-mail: ksns1987@gmail.com
Business License No.: 209-82-62443

Copyright © The Korean Spinal Neurosurgery Society.

Developed in M2PI

Close layer
prev next