Therapeutic Approaches of Amyotrophic Lateral Sclerosis: History
Please note this is an old version of this entry, which may differ significantly from the current revision.

Amyotrophic lateral sclerosis (ALS) is an extremely heterogeneous disease of motor neurons that eventually leads to death. Despite impressive advances in understanding the genetic, molecular, and pathological mechanisms of the disease, the only drug approved to date by both the FDA and EMA is riluzole, with a modest effect on survival. 

  • riluzole
  • preclinical models
  • protein homeostasis inductors
  • gene targeted strategies

1. Introduction

Amyotrophic lateral sclerosis (ALS) was first described by Jean-Martin Charcot in 1869 as an inexorably progressive disease primarily associated with degeneration of upper and lower motor neurons [1]. The early 1990s was an important turning point in the study of ALS when the positive results of a large therapeutic trial of riluzole, an anti-glutamatergic agent, were published, showing a prolongation of patient survival [2]. As a result, riluzole was approved by the FDA as the first drug for ALS. To date, it is the only therapeutic agent approved in both the United States and Europe with a modest effect, prolonging patients’ lives by only a few months without significantly improving functional deterioration [3]. Edaravone (RadicavaTM) was approved by the Food and Drugs Administration (FDA) in 2017 but not by the European Medicines Agency (EMA). Approval was based on a phase III study showing that edaravone slowed the loss of physical function by 33% at 24 weeks compared with placebo on the ALSFRS-R scale [3]. However, the efficacy of the treatment remained controversial due to the short duration of the study and the strict inclusion criteria, which were limited to patients with an early stage of the disease. Only the significant improvement in the symptomatic treatment of ALS had a significant impact on patient survival and quality of life [4]. In particular, the development of non-invasive ventilators and the improvement of bronchial suction techniques have placed the management of ALS in the context of multidisciplinary care [5].

2. Therapeutic Approaches

2.1. Pharmacologic Approaches

Table 1 provides a non-exhaustive list of ongoing clinical trials to illustrate the variety of different disease mechanisms being investigated in this complex and multifactorial disease. Recently, two drugs have been approved in some countries. The IV formulation of the drug (RadicavaTM) had previously been approved by the FDA in 2017. The approval was based on a phase III study that showed edaravone slowed the loss of physical function by 33% after 24 weeks compared to placebo on the ALSFRS-R scale [3]. However, the effectiveness of the treatment remained controversial, and it was not approved by EMA. One reason for this was the short duration of the trial and the strict inclusion criteria, which were limited to patients in the early stage of the disease. The high level of care required with repeated daily IV infusions was a limitation for prescribing the drug. This led to the development of an oral form of edaravone (Radicava ORSTM) by Mitsubishi Tanabe Pharma, which was recently approved by the FDA in May 2022, and a phase III trial is currently underway to assess the long-term safety and tolerability of oral edaravone over 96 weeks (NCT04577404). The second drug is from Amylyx Pharmaceuticals, an oral, fixed-dose co-formulation of sodium phenylbutyrate and ursodoxicoltaurine (AlbriozaTM) to address both mitochondrial dysfunction and endoplasmic reticulum stress. In June 2022, the co-formulation received its first conditional approval in Canada for the treatment of (ALS) in adults. The approval was based on the results of the multicentre phase II CENTAUR trial (NCT03127514), in which the slowing of progression of ALS with the treatment compared with placebo [6].
Table 1. A non-exhaustive list of therapeutic agents in development for ALS.
Agent Targeted Mechanism Mechanism Results Phase Ref.
Sodium Phenylbutyrate-Taurursodiol endoplasmic reticulum stress, and mitochondrial dysfunction Sodium phenylbutyrate is a histone deacetylase inhibitor that has been shown to upregulate heat shock proteins and act as a small molecule chaperone, alleviating endoplasmic reticulum stress toxicity [17,18]. Taurursodiol recovers mitochondrial bioenergetic deficits through multiple mechanisms, including preventing the translocation of Bax protein into the mitochondrial membrane, thereby decreasing mitochondrial permeability and increasing the cell’s apoptotic threshold [19] Less functional deterioration measured by the ALSFRS-R score over a 24-week period. Secondary outcomes, including decreases in isometric muscle strength and vital capacity, did not differ significantly between groups II [20]
Colchicine Protein aggregates, autophagy, and neuroinflammation Colchicine could upregulate proteins involved in autophagy, including the TFEB, the TFEB-regulated adaptor protein SQSTM1/p62 and the autophagy player microtubule-associated protein 1A/1B-light chain 3 (LC3). Ongoing II [21]
Rapamycin Autophagy and neuroinflammation Rapamycin is based on the inhibition of mTORC1. mTORC1 targets regulatory proteins in cell signalling and regulates autophagy by inhibiting the unc-51-like kinase 1 complex. Ongoing II [22]
BIIB100 (KPT-350) Nucleocytoplasmic transport dysfunction Selective inhibitor of nuclear export that inhibits exportin 1 (XPO1; CRM1). Ongoing I  
Deferiprone Iron accumulation Iron Chelation Ongoing II [23]
TIRASEMTIV Muscle contractility A FSTA that selectively activates the fast skeletal muscle troponin complex by increasing its sensitivity to calcium In a phase IIb clinical trial, SVC and muscle strength were found to decline significantly more slowly in tirasemtiv-treated participants.
But no significant difference was found in the decline in functional disability as measured by the ALSFRS-R. However, no significant difference in disease progression was demonstrated in the phase III clinical trial.
II/III [24,25]
Interleukine 2 Neuroinflammation Immunomodulatory strategy by promoting Treg expansion, which attenuates neuroinflammation. A phase IIa study showed that low dose IL-2 is well tolerated and immunologically effective in subjects with ALS [26] III [26]
Masitinib Neuroinflammation Tyrosine kinase inhibitor targets microglia and mast cells through inhibiting a limited number of kinases. Masitinib blocks microglia proliferation and activation, and mast cell-mediated degranulation, the release of cytotoxic substances that might further damage the motor nerves. A randomised, placebo-controlled phase III trial has previously shown that oral masitinib (4.5 mg/kg/day) slows the rate of functional decline with acceptable safety in ALS patients with an ALSFRS-R progression rate of <1.1 points/month III [27]
Ibudilast
(MN-166)
Neuroinflammation Inhibitor of macrophage migration inhibitory factor and phosphodiesterases 3,4,10 and 11 [28,29]. Ibudilast attenuates CNS microglial activation and secretion of pro-inflammatory cytokines. Ongoing II/III [29,30]
Fasudil Neuroinflammation Rho kinase inhibitor Ongoing II [31]
Ravulizumab Neuroinflammation Humanized monoclonal antibody to complement factor 5 which acts to block complement activation The independent Data and Safety Monitoring Board monitoring committee recommended that the study be discontinued due to lack of efficacy. No new safety findings were observed. III [32]
Zilucoplan Neuroinflammation A small molecule that works aa s C5 complement inhibitor The The independent Data and Safety Monitoring Board recommended stopping the zilucoplan regimen because the likelihood of meaningfully slowing disease progression was considered low. III [33]
Anakinra Neuroinflammation The monoclonal antibody that works as a IL–1 receptor antagonist Ongoing II  
Tocilizumab Neuroinflammation The monoclonal antibody that works as a IL–1 receptor antagonist Tocilizumab is safe and tolerable and reduces C-reactive protein concentrations in the plasma and cerebrospinal fluid of ALS patients II [34]
Tofersen
(BIIB067)
Gain of function SOD1 It is an antisense oligonucleotide (ASO) targeting SOD1 In the Phase III VALOR study, the primary endpoint as measured by the ALSFRS-R did not reach statistical significance; however, signs of reduced disease progression across multiple secondary and exploratory endpoints were observed III [35]
BIIB078 Gain of function C9ORF72 It is an antisense oligonucleotide (ASO) for C9ORF72-associated ALS In a Phase I study, BIIB078 was generally well-tolerated. The adverse events were mostly mild to moderate in severity and occurred at a similar rate across BIIB078 and placebo groups.
BIIB078 did not meet any secondary efficacy endpoints and it did not demonstrate clinical benefit. Therefore, the clinical program will be discontinued
I [36]
Abbreviations: ALSFRS-R, ALS Functional Rating Scale-revised; FSTA, fast skeletal muscle troponin activator; SVC, slow vital capacity; TFEB, master regulator transcription factor EB.
Another therapeutic approach that has recently gained renewed interest is the targeting of muscle abnormalities in ALS [7]. The first rationale is neuroprotective, as changes in the muscle and neuromuscular junction may play a role in retrograde degeneration, as suggested by the researchers' recent work on the role of secretion of toxic exosomes by muscle in motor neuron degeneration [8]. A second approach is symptomatic by increasing muscle contractility, with two troponin activators in development, tirasemtiv [9][10] and reldesemtiv (NCT04944784), or improving muscle mass and strength [11].

2.2. Gene and Cell Therapy Approaches

Recently, experimental strategies targeting genes have come to the forefront of clinical research, offering the promising therapeutic potential for ALS and hope for patients with ALS. Several technologies are being tested in preclinical or clinical phases. These include antisense oligonucleotides (ASO), interfering RNAs, viral vectors, or gene editing with CRISPR/Cas9 [12]. Successful treatment with an ASO (NusinersenTM) and then with a viral vector (ZolgensmaTM) for another motor neuron disease, spinal muscular atrophy (SMA), has raised hopes that these approaches will lead to approved drugs for ALS in the short term. It should be emphasized, however, that in autosomal dominant forms of ALS, it is no longer a question of compensating for the loss of function of a deleterious gene, as is the case with SMA, but on the contrary of decreasing the expression of a mutation that leads to a toxic gain of function. Such approaches are currently being tested in clinical trials for patients with mutations in SOD1, C9ORF72, and FUS genes.
The most advanced ASO-based treatment is TofersenTM, developed by Biogen, which is designed to reduce the synthesis of the SOD1 protein [13][14]. The VALOR study enrolled 108 ALS patients with an SOD1 mutation who were treated for 28 weeks (NCT02623699). The main results were disappointing, as the main objective of slowing functional deterioration as measured by the ALSFRS-R was not achieved [14]. However, TofersenTM will apply for approval under the accelerated approval process based on data showing a marked decrease in neurofilament light (NfL) levels, a biomarker of neuronal degeneration, and a reduction in SOD1 protein associated with a trend towards less disease progression [14]. Based on the reasonable assumption that treatment is more effective at an early stage of degeneration, a clinical trial has recently been started in subjects who are carriers of the SOD1 mutation and do not yet have clinical manifestations of the disease (ATLAS study, NCT04856982). The research is investigating whether TofersenTM can delay the onset of the disease [15][16]. Subjects are eligible for intervention in this research if the follow-up of the subjects reveals an increase in NfL levels in the blood above a certain threshold that has been shown to predict the onset of symptoms within one to two years [16][17].
A study has been conducted with another ASO-based treatment, TadnersenTM (BIIB078), which selectively inhibits the mutant C9ORF72 transcripts [18][19]. Although the therapy was generally safe and well tolerated in people with C9ORF72-associated ALS, it did not result in significant clinical benefit compared with placebo. The extension study was stopped, and clinical development was discontinued. Wave Life Sciences is taking a similar approach with its investigational drug WVE-004, a stereopure ASO, targeting variants containing G4C2, a hexanucleotide repeat expansion associated with the C9ORF72 gene. This research, the phase Ib/IIa FOCUS -C9 trial (NCT04931862), was initiated in August 2021 and is evaluating WVE-004 in C9ORF72-associated ALS and frontotemporal dementia. A phase III trial of JacifusenTM (NCT04768972), an ASO designed to reduce FUS protein synthesis from FUS mRNA, is ongoing for patients with FUS gene mutations associated with aggressive juvenile forms of ALS [20][21]. In contrast to ASOs targeting inherited forms of ALS, other strategies are currently being developed that are applicable to sporadic cases and aim to modulate the expression of disease-modifying genes. A phase I trial of BIIB105, an ASO targeting the ataxin-2 gene, is currently underway in sporadic patients with ALS (NCT04494256) [22]. The first rationale is that polyglutamine expansions in ataxin-2 increase the risk of ALS in people who carry them. Secondly, work in yeast and fly models has shown that ataxin-2 promotes aggregation and toxicity of the TDP-43 protein [23].
However, recent evidence has raised awareness that while these strategies will certainly diversify, the challenge of effectiveness and safety remains significant. These risks should be considered and are clearly underscored by the failure of a trial of the ASO TominersenTM in Huntington’s disease, where the trial was stopped prematurely because the participants’ symptoms worsened [24]. A major concern with ASO treatment is that the treatment that aims to decrease the levels of the abnormal protein also affects its normal counterpart and, therefore, its physiological function. Among ALS causal mutation, this concern is important with C9ORF72 mutations; whether the mechanism is loss and/or gain of function remain controversial [25]. Strategies based on genome editing, in particular CRISPR/Cas9 technology, could specifically target the genetic mutations, such as removing the intronic position of the C9ORF72 repeat expansion by a ‘cutting-deletion-fusion’ method [26][27][28]. A second risk is the potential immunogenicity of ASO and the risk of meningitis when administered intrathecally. Serious neurological events were reported in 4.8% of ALS patients receiving TofersenTM, including two cases of myelitis (2.0%).
The therapeutic approach using stem cells has recently been promoted as a potential neuroprotective therapeutic strategy for ALS. In particular, mesenchymal stem cells (MSCs) have multiple effects, such as stimulation of intrinsic neurogenesis, the release of various neurotrophic factors, and modulation of immune-inflammatory processes, transforming the patient’s environment from a pro-inflammatory toxic state to an anti-inflammatory and neuroprotective state [29]. Several studies investigating the effect of therapeutic approaches using MSCs in mouse disease models have shown that motor neuron loss was slower in the group treated with MSCs [30][31][32][33][34][35]. Subsequently, several clinical trials were conducted to investigate the therapeutic effect of MSCs in ALS patients using intrathecal or intraspinal administration of bone marrow-derived mesenchymal or mononuclear cells or fetal neural stem cells [36][37][38][39]. However, a recent phase III trial of intrathecal administration of MSCs in ALS patients did not meet its primary endpoint of a change in ALS decline, although participants with less severe disease retained more function compared with the placebo group (NCT03280056) [40]. It shows that there is still much to be done in terms of the source of stem cells, the mode of administration, the selection of potentially better-responding patients, clinical endpoints, and safety [41][42][43][44][45][46].

This entry is adapted from the peer-reviewed paper 10.3390/jpm12101601

References

  1. van Es, M.A.; Hardiman, O.; Chio, A.; Al-Chalabi, A.; Pasterkamp, R.J.; Veldink, J.H.; van den Berg, L.H. Amyotrophic lateral sclerosis. Lancet 2017, 390, 2084–2098.
  2. Bensimon, G.; Lacomblez, L.; Meininger, V. A controlled trial of riluzole in amyotrophic lateral sclerosis. ALS/Riluzole Study Group. N. Engl. J. Med. 1994, 330, 585–591.
  3. Abe, K.; Aoki, M.; Tsuji, S.; Itoyama, Y.; Sobue, G.; Togo, M.; Hamada, C.; Tanaka, M.; Akimoto, M.; Nakamura, K.; et al. Safety and efficacy of edaravone in well defined patients with amyotrophic lateral sclerosis: A randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2017, 16, 505–512.
  4. Traynor, B.J.; Alexander, M.; Corr, B.; Frost, E.; Hardiman, O. Effect of a multidisciplinary amyotrophic lateral sclerosis (ALS) clinic on ALS survival: A population based study, 1996-2000. J. Neurol. Neurosurg. Psychiatry 2003, 74, 1258–1261.
  5. Radunovic, A.; Annane, D.; Rafiq, M.K.; Brassington, R.; Mustfa, N. Mechanical ventilation for amyotrophic lateral sclerosis/motor neuron disease. Cochrane Database Syst. Rev. 2017, 10, Cd004427.
  6. Paganoni, S.; Macklin, E.A.; Hendrix, S.; Berry, J.D.; Elliott, M.A.; Maiser, S.; Karam, C.; Caress, J.B.; Owegi, M.A.; Quick, A.; et al. Trial of Sodium Phenylbutyrate-Taurursodiol for Amyotrophic Lateral Sclerosis. N. Engl. J. Med. 2020, 383, 919–930.
  7. Scaricamazza, S.; Salvatori, I.; Giacovazzo, G.; Loeffler, J.P.; Renè, F.; Rosina, M.; Quessada, C.; Proietti, D.; Heil, C.; Rossi, S.; et al. Skeletal-Muscle Metabolic Reprogramming in ALS-SOD1(G93A) Mice Predates Disease Onset and Is A Promising Therapeutic Target. iScience 2020, 23, 101087.
  8. Le Gall, L.; Duddy, W.J.; Martinat, C.; Mariot, V.; Connolly, O.; Milla, V.; Anakor, E.; Ouandaogo, Z.G.; Millecamps, S.; Lainé, J.; et al. Muscle cells of sporadic amyotrophic lateral sclerosis patients secrete neurotoxic vesicles. J. Cachexia Sarcopenia Muscle 2022, 13, 1385–1402.
  9. Li, M.X.; Mercier, P.; Hartman, J.J.; Sykes, B.D. Structural Basis of Tirasemtiv Activation of Fast Skeletal Muscle. J. Med. Chem. 2021, 64, 3026–3034.
  10. Shefner, J.M.; Cudkowicz, M.E.; Hardiman, O.; Cockcroft, B.M.; Lee, J.H.; Malik, F.I.; Meng, L.; Rudnicki, S.A.; Wolff, A.A.; Andrews, J.A. A phase III trial of tirasemtiv as a potential treatment for amyotrophic lateral sclerosis. Amyotroph. Lateral Scler. Front. Degener. 2019, 20, 584–594.
  11. Li, J.; Fredericks, M.; Cannell, M.; Wang, K.; Sako, D.; Maguire, M.C.; Grenha, R.; Liharska, K.; Krishnan, L.; Bloom, T.; et al. ActRIIB:ALK4-Fc alleviates muscle dysfunction and comorbidities in murine models of neuromuscular disorders. J. Clin. Investig. 2021, 131.
  12. Cappella, M.; Pradat, P.F.; Querin, G.; Biferi, M.G. Beyond the Traditional Clinical Trials for Amyotrophic Lateral Sclerosis and The Future Impact of Gene Therapy. J. Neuromuscul. Dis. 2021, 8, 25–38.
  13. Biogen Announces Topline Results from the Tofersen Phase 3 Study and its Open-Label Extension in SOD1-ALS. Available online: https://www.globenewswire.com/fr/news-release/2021/10/17/2315291/0/en/Biogen-Announces-Topline-Results-from-the-Tofersen-Phase-3-Study-and-its-Open-Label-Extension-in-SOD1-ALS.html (accessed on 1 March 2022).
  14. Miller, T.; Cudkowicz, M.; Shaw, P.J.; Andersen, P.M.; Atassi, N.; Bucelli, R.C.; Genge, A.; Glass, J.; Ladha, S.; Ludolph, A.L.; et al. Phase 1-2 Trial of Antisense Oligonucleotide Tofersen for SOD1 ALS. N. Engl. J. Med. 2020, 383, 109–119.
  15. Westlund, R. Pre-Symptomatic ALS Clinical Trial Paves the Way for Early Intervention Strategies in Neurodegenerative Diseases. Available online: https://physician-news.umiamihealth.org/pre-symptomatic-als-clinical-trial-paves-the-way-for-early-intervention-strategies-in-neurodegenerative-diseases/ (accessed on 15 April 2022).
  16. ClinicalTrials.gov. A Study of BIIB067 When Initiated in Clinically Presymptomatic Adults With a Confirmed Superoxide Dismutase 1 Mutation (ATLAS). Available online: https://clinicaltrials.gov/ct2/show/NCT04856982 (accessed on 15 April 2022).
  17. Benatar, M.; Wuu, J.; Andersen, P.M.; Lombardi, V.; Malaspina, A. Neurofilament light: A candidate biomarker of presymptomatic amyotrophic lateral sclerosis and phenoconversion. Ann. Neurol. 2018, 84, 130–139.
  18. Aoki, Y.; Wood, M.J.A. Emerging Oligonucleotide Therapeutics for Rare Neuromuscular Diseases. J. Neuromuscul. Dis. 2021, 8, 869–884.
  19. Talbot, K.; Wood, M.J.A. Wrangling RNA: Antisense oligonucleotides for neurological disorders. Sci. Transl. Med. 2019, 11, eaay2069.
  20. Conte, A.; Lattante, S.; Zollino, M.; Marangi, G.; Luigetti, M.; Del Grande, A.; Servidei, S.; Trombetta, F.; Sabatelli, M. P525L FUS mutation is consistently associated with a severe form of juvenile amyotrophic lateral sclerosis. Neuromuscul. Disord. NMD 2012, 22, 73–75.
  21. Neurology, P. Phase 3 Trial of Jacifusen for Amyloid Lateral Sclerosis With Fused-in-Sarcoma Mutation. Available online: https://practicalneurology.com/news/phase-3-trial-of-jacifusen-for-amyloid-lateral-sclerosis-with-fused-in-sarcoma-mutation#:~:text=04.07.21-,Phase%203%20Trial%20of%20Jacifusen%20for%20Amyloid,With%20Fused%2Din%2DSarcoma%20Mutation&text=A%20phase%203%20clinical%20trial,%2Dsarcoma%20gene%20(FUS) (accessed on 19 April 2022).
  22. ClinicalTrials.gov. A Study to Assess the Safety, Tolerability, and Pharmacokinetics of BIIB105 in Participants With Amyotrophic Lateral Sclerosis With or Without Poly-cytosine-adenine-guanine (CAG) Expansion in the Ataxin-2 Gene. Available online: https://www.clinicaltrials.gov/ct2/show/NCT04494256?term=NCT04494256&type=Intr&cond=ALS&phase=012&draw=2&rank=1 (accessed on 19 April 2022).
  23. Elden, A.C.; Kim, H.J.; Hart, M.P.; Chen-Plotkin, A.S.; Johnson, B.S.; Fang, X.; Armakola, M.; Geser, F.; Greene, R.; Lu, M.M.; et al. Ataxin-2 intermediate-length polyglutamine expansions are associated with increased risk for ALS. Nature 2010, 466, 1069–1075.
  24. Kwon, D. Failure of genetic therapies for Huntington’s devastates community. Nature 2021, 593, 180.
  25. Mizielinska, S.; Isaacs, A.M. C9orf72 amyotrophic lateral sclerosis and frontotemporal dementia: Gain or loss of function? Curr. Opin. Neurol. 2014, 27, 515–523.
  26. Renton, A.E.; Majounie, E.; Waite, A.; Simón-Sánchez, J.; Rollinson, S.; Gibbs, J.R.; Schymick, J.C.; Laaksovirta, H.; van Swieten, J.C.; Myllykangas, L.; et al. A hexanucleotide repeat expansion in C9ORF72 is the cause of chromosome 9p21-linked ALS-FTD. Neuron 2011, 72, 257–268.
  27. Belzil, V.V.; Bauer, P.O.; Prudencio, M.; Gendron, T.F.; Stetler, C.T.; Yan, I.K.; Pregent, L.; Daughrity, L.; Baker, M.C.; Rademakers, R.; et al. Reduced C9orf72 gene expression in c9FTD/ALS is caused by histone trimethylation, an epigenetic event detectable in blood. Acta Neuropathol. 2013, 126, 895–905.
  28. Pickar-Oliver, A.; Gersbach, C.A. The next generation of CRISPR-Cas technologies and applications. Nat. Rev. Mol. Cell Biol. 2019, 20, 490–507.
  29. Gugliandolo, A.; Bramanti, P.; Mazzon, E. Mesenchymal Stem Cells: A Potential Therapeutic Approach for Amyotrophic Lateral Sclerosis? Stem Cells Int. 2019, 2019, 3675627.
  30. Zhao, C.P.; Zhang, C.; Zhou, S.N.; Xie, Y.M.; Wang, Y.H.; Huang, H.; Shang, Y.C.; Li, W.Y.; Zhou, C.; Yu, M.J.; et al. Human mesenchymal stromal cells ameliorate the phenotype of SOD1-G93A ALS mice. Cytotherapy 2007, 9, 414–426.
  31. Vercelli, A.; Mereuta, O.M.; Garbossa, D.; Muraca, G.; Mareschi, K.; Rustichelli, D.; Ferrero, I.; Mazzini, L.; Madon, E.; Fagioli, F. Human mesenchymal stem cell transplantation extends survival, improves motor performance and decreases neuroinflammation in mouse model of amyotrophic lateral sclerosis. Neurobiol. Dis. 2008, 31, 395–405.
  32. Uccelli, A.; Milanese, M.; Principato, M.C.; Morando, S.; Bonifacino, T.; Vergani, L.; Giunti, D.; Voci, A.; Carminati, E.; Giribaldi, F.; et al. Intravenous mesenchymal stem cells improve survival and motor function in experimental amyotrophic lateral sclerosis. Mol. Med. 2012, 18, 794–804.
  33. Boido, M.; Piras, A.; Valsecchi, V.; Spigolon, G.; Mareschi, K.; Ferrero, I.; Vizzini, A.; Temi, S.; Mazzini, L.; Fagioli, F.; et al. Human mesenchymal stromal cell transplantation modulates neuroinflammatory milieu in a mouse model of amyotrophic lateral sclerosis. Cytotherapy 2014, 16, 1059–1072.
  34. Forostyak, S.; Homola, A.; Turnovcova, K.; Svitil, P.; Jendelova, P.; Sykova, E. Intrathecal delivery of mesenchymal stromal cells protects the structure of altered perineuronal nets in SOD1 rats and amends the course of ALS. Stem Cells 2014, 32, 3163–3172.
  35. Marconi, S.; Bonaconsa, M.; Scambi, I.; Squintani, G.M.; Rui, W.; Turano, E.; Ungaro, D.; D’Agostino, S.; Barbieri, F.; Angiari, S.; et al. Systemic treatment with adipose-derived mesenchymal stem cells ameliorates clinical and pathological features in the amyotrophic lateral sclerosis murine model. Neuroscience 2013, 248, 333–343.
  36. Lewis, C.M.; Suzuki, M. Therapeutic applications of mesenchymal stem cells for amyotrophic lateral sclerosis. Stem Cell Res. Ther. 2014, 5, 32.
  37. Mazzini, L.; Ferrero, I.; Luparello, V.; Rustichelli, D.; Gunetti, M.; Mareschi, K.; Testa, L.; Stecco, A.; Tarletti, R.; Miglioretti, M.; et al. Mesenchymal stem cell transplantation in amyotrophic lateral sclerosis: A Phase I clinical trial. Exp. Neurol. 2010, 223, 229–237.
  38. Mazzini, L.; Gelati, M.; Profico, D.C.; Sorarù, G.; Ferrari, D.; Copetti, M.; Muzi, G.; Ricciolini, C.; Carletti, S.; Giorgi, C.; et al. Results from Phase I Clinical Trial with Intraspinal Injection of Neural Stem Cells in Amyotrophic Lateral Sclerosis: A Long-Term Outcome. Stem Cells Transl. Med. 2019, 8, 887–897.
  39. Glass, J.D.; Boulis, N.M.; Johe, K.; Rutkove, S.B.; Federici, T.; Polak, M.; Kelly, C.; Feldman, E.L. Lumbar intraspinal injection of neural stem cells in patients with amyotrophic lateral sclerosis: Results of a phase I trial in 12 patients. Stem Cells 2012, 30, 1144–1151.
  40. Cudkowicz, M.E.; Lindborg, S.R.; Goyal, N.A.; Miller, R.G.; Burford, M.J.; Berry, J.D.; Nicholson, K.A.; Mozaffar, T.; Katz, J.S.; Jenkins, L.J.; et al. A randomized placebo-controlled phase 3 study of mesenchymal stem cells induced to secrete high levels of neurotrophic factors in amyotrophic lateral sclerosis. Muscle Nerve 2022, 65, 291–302.
  41. Nabavi, S.M.; Arab, L.; Jarooghi, N.; Bolurieh, T.; Abbasi, F.; Mardpour, S.; Azimyian, V.; Moeininia, F.; Maroufizadeh, S.; Sanjari, L.; et al. Safety, Feasibility of Intravenous and Intrathecal Injection of Autologous Bone Marrow Derived Mesenchymal Stromal Cells in Patients with Amyotrophic Lateral Sclerosis: An Open Label Phase I Clinical Trial. Cell J. 2019, 20, 592–598.
  42. Oh, K.W.; Noh, M.Y.; Kwon, M.S.; Kim, H.Y.; Oh, S.I.; Park, J.; Kim, H.J.; Ki, C.S.; Kim, S.H. Repeated Intrathecal Mesenchymal Stem Cells for Amyotrophic Lateral Sclerosis. Ann. Neurol. 2018, 84, 361–373.
  43. Staff, N.P.; Madigan, N.N.; Morris, J.; Jentoft, M.; Sorenson, E.J.; Butler, G.; Gastineau, D.; Dietz, A.; Windebank, A.J. Safety of intrathecal autologous adipose-derived mesenchymal stromal cells in patients with ALS. Neurology 2016, 87, 2230–2234.
  44. Petrou, P.; Gothelf, Y.; Argov, Z.; Gotkine, M.; Levy, Y.S.; Kassis, I.; Vaknin-Dembinsky, A.; Ben-Hur, T.; Offen, D.; Abramsky, O.; et al. Safety and Clinical Effects of Mesenchymal Stem Cells Secreting Neurotrophic Factor Transplantation in Patients With Amyotrophic Lateral Sclerosis: Results of Phase 1/2 and 2a Clinical Trials. JAMA Neurol. 2016, 73, 337–344.
  45. Barczewska, M.; Grudniak, M.; Maksymowicz, S.; Siwek, T.; Ołdak, T.; Jezierska-Woźniak, K.; Gładysz, D.; Maksymowicz, W. Safety of intrathecal injection of Wharton’s jelly-derived mesenchymal stem cells in amyotrophic lateral sclerosis therapy. Neural Regen. Res. 2019, 14, 313–318.
  46. Siwek, T.; Maksymowicz, W.; Barczewska, M.; Grabarczyk, L.; Sowa, M.J.J.N.N. Mesenchymal stem cell (MSC) transplantation in patients with amyotrophic lateral sclerosis (ALS): Is there a “responder population”. J. Neurol. Neurosci. 2018, 9, 260.
More
This entry is offline, you can click here to edit this entry!
Video Production Service