Comprehensive Research on Past and Future Therapeutic Strategies Devoted to Treatment of Amyotrophic Lateral Sclerosis
Abstract
:1. Introduction
2. Therapeutic Strategies for ALS Targets
2.1. Therapeutic Strategies against Oxidative Stress
2.2. Therapeutic Strategies against Oxidative Stress via Protein Aggregation
2.3. Therapeutic Strategies against Mitochondrial Dysfunction
2.4. Therapeutic Strategies against Glutamate-Induced Excitotoxicity
2.5. Therapeutic Strategies for Reducing Apoptosis and/or Boosting Autophagy
2.6. Therapeutic Strategies against Neuroinflammation
2.7. Therapeutic Strategies against Axonal Degeneration
2.8. Therapeutic Strategies against Skeletal Muscle Deterioration
2.9. Therapeutic Strategies against Viruses
2.10. The Importance of Trophic Factors in ALS
2.11. Newly Synthesized and Evaluated Compounds as Anti-ALS Agents
3. Conclusions and Future Perspectives
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Liu, J.; Wang, F. Role of neuroinflammation in amyotrophic lateral sclerosis: Cellular mechanisms and therapeutic implications. Front. Immunol. 2017, 8, 1005. [Google Scholar] [CrossRef] [Green Version]
- Martinez, A.; Palomo Ruiz, M.D.; Perez, D.I.; Gil, C. Drugs in clinical development for the treatment of amyotrophic lateral sclerosis. Expert Opin. Investig. Drugs 2017, 26, 403–414. [Google Scholar] [CrossRef]
- Gordon, P.H. Amyotrophic lateral sclerosis: Pathophysiology, diagnosis and management. CNS Drugs 2011, 25, 1–15. [Google Scholar] [CrossRef]
- Harikrishnareddy, D.; Misra, S.; Upadhyay, S.; Modi, M.; Medhi, B. Roots to start research in amyotrophic lateral sclerosis: Molecular pathways and novel therapeutics for future. Rev. Neurosci. 2015, 26, 161–181. [Google Scholar] [CrossRef] [PubMed]
- Sardana, D.; Zhu, C.; Zhang, M.; Gudivada, R.C.; Yang, L.; Jegga, A.G. Drug repositioning for orphan diseases. Brief. Bioinform. 2011, 12, 346–356. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Longinetti, E.; Fang, F. Epidemiology of amyotrophic lateral sclerosis: An update of recent literature. Curr. Opin. Neurol. 2019, 32, 771–776. [Google Scholar] [CrossRef]
- Root, J.; Merino, P.; Nuckols, A.; Johnson, M.; Kukar, T. Lysosome dysfunction as a cause of neurodegenerative diseases: Lessons from frontotemporal dementia and amyotrophic lateral sclerosis. Neurobiol. Dis. 2021, 154, 105360. [Google Scholar] [CrossRef]
- Pawlyk, A.C.; Cassel, J.A.; Reitz, A.B. Current nervous system related drug targets for the treatment of amyotrophic lateral sclerosis. Curr. Pharm. Des. 2010, 16, 2053–2073. [Google Scholar] [CrossRef] [PubMed]
- Pham, J.; Keon, M.; Brennan, S.; Saksena, N. Connecting RNA-modifying similarities of TDP-43, FUS, and SOD1 with microRNA dysregulation amidst a renewed network perspective of amyotrophic lateral sclerosis. Proteinopathy. Int. J. Mol. Sci. 2020, 21, 3464. [Google Scholar] [CrossRef]
- Rowland, L.P.; Shneider, N.A. Amyotrophic lateral sclerosis. N. Engl. J. Med. 2001, 344, 1688–1700. [Google Scholar] [CrossRef]
- Bradley, W.G.; Mash, D.C. Beyond Guam: The cyanobacteria/BMAA hypothesis of the cause of ALS and other neurodegenerative diseases. Amyotroph. Lateral Scler. 2009, 2, 7–20. [Google Scholar] [CrossRef]
- Habib, A.A.; Mitsumoto, H. Emerging drugs for amyotrophic lateral sclerosis. Expert Opin. Emerg. Drugs 2011, 16, 537–558. [Google Scholar] [CrossRef]
- Byrne, S.; Jordan, I.; Elamin, M.; Hardiman, O. Age at onset of amyotrophic lateral sclerosis is proportional to life expectancy. Amyotroph. Lateral Scler. Front. Degener. 2013, 14, 604–607. [Google Scholar] [CrossRef]
- Farace, C.; Fenu, G.; Lintas, S.; Oggiano, R.; Pisano, A.; Sabalic, A.; Solinas, G.; Bocca, B.; Forte, G.; Madeddu, R. Amyotrophic lateral sclerosis and lead: A systematic update. NeuroToxicology 2020, 81, 80–88. [Google Scholar] [CrossRef] [PubMed]
- Okano, H.; Yasuda, D.; Fujimori, K.; Morimoto, S.; Takahashi, S. Ropinirole, a new ALS drug candidate developed using iPSCs. Trends Pharmacol. Sci. 2020, 41, 99–109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Costa, J.; Gomes, C.; de Carvalho, M. Diagnosis, pathogenesis and therapeutic targets in amyotrophic lateral sclerosis. CNS Neurol. Disord. Drug Targets 2010, 9, 764–778. [Google Scholar] [CrossRef] [PubMed]
- Van Harten, A.C.M.; Phatnani, H.; Przedborski, S. Non-cell-autonomous pathogenic mechanisms in amyotrophic lateral sclerosis. Trends Neurosci. 2021, 44, 658–668. [Google Scholar] [CrossRef]
- Nakagawa, Y.; Yamada, S. A novel hypothesis on metal dyshomeostasis and mitochondrial dysfunction in amyotrophic lateral sclerosis: Potential pathogenetic mechanism and therapeutic implications. Eur. J. Pharmacol. 2021, 892, 173737. [Google Scholar] [CrossRef]
- Gurney, M.E.; Pu, H.; Chiu, A.Y.; Dal Canto, M.C.; Polchow, C.Y.; Alexander, D.D.; Caliendo, J.; Hentati, A.; Kwon, Y.W.; Deng, H.X.; et al. Motor neuron degeneration in mice that express a human Cu, Zn superoxide dismutase mutation. Science 1994, 264, 1772–1775. [Google Scholar] [CrossRef]
- Benatar, M. Lost in translation: Treatment trials in the SOD1 mouse and in human ALS. Neurobiol. Dis. 2007, 26, 1–13. [Google Scholar] [CrossRef]
- Gomes, C.; Escrevente, C.; Costa, J. Mutant superoxide dismutase 1 overexpression in NSC-34 cells: Effect of trehalose on aggregation, TDP-43 localization and levels of co-expressed glycoproteins. Neurosci. Lett. 2010, 475, 145–149. [Google Scholar] [CrossRef]
- Chiò, A.; Mazzini, L.; Mora, G. Disease-modifying therapies in amyotrophic lateral sclerosis. Neuropharmacology 2020, 167, 107986. [Google Scholar] [CrossRef] [PubMed]
- Menzies, F.M.; Cookson, M.R.; Taylor, R.W.; Turnbull, D.M.; Chrzanowska-Lightowlers, Z.M.; Dong, L.; Figlewicz, D.A.; Shaw, P.J. Mitochondrial dysfunction in a cell culture model of familial amyotrophic lateral sclerosis. Brain 2002, 125, 1522–1533. [Google Scholar] [CrossRef] [Green Version]
- Goos, M.; Zech, W.D.; Jaiswal, M.K.; Balakrishnan, S.; Ebert, S.; Mitchell, T.; Carrì, M.T.; Keller, B.U.; Nau, R. Expression of a Cu, Zn superoxide dismutase typical for familial amyotrophic lateral sclerosis increases the vulnerability of neuroblastoma cells to infectious injury. BMC Infect. Dis. 2007, 7, 131. [Google Scholar] [CrossRef] [PubMed]
- Hergesheimer, R.; Lanznaster, D.; Vourc’h, P.; Andres, C.; Bakkouche, S.; Beltran, S.; Blasco, H.; Corcia, P.; Couratier, P. Advances in disease-modifying pharmacotherapies for the treatment of amyotrophic lateral sclerosis. Expert Opin. Pharmacother. 2020, 21, 1103–1110. [Google Scholar] [CrossRef]
- Masrori, P.; Van Damme, P. Amyotrophic lateral sclerosis: A clinical review. Eur. J. Neurol. 2020, 27, 1918–1929. [Google Scholar] [CrossRef] [PubMed]
- Behl, T.; Kaur, G.; Sehgal, A.; Bhardwaj, S.; Singh, S.; Buhas, C.; Judea-Pusta, C.; Uivarosan, D.; Munteanu, M.A.; Bungau, S. Multifaceted role of matrix metalloproteinases in neurodegenerative diseases: Pathophysiological and therapeutic perspectives. Int. J. Mol. Sci. 2021, 22, 1413. [Google Scholar] [CrossRef]
- Smart Servier Medical Art. Available online: https://smart.servier.com/ (accessed on 4 November 2021).
- Fang, X. Potential role of gut microbiota and tissue barriers in Parkinson’s disease and amyotrophic lateral sclerosis. Int. J. Neurosci. 2016, 126, 771–776. [Google Scholar] [CrossRef]
- Wu, S.; Yi, J.; Zhang, Y.G.; Zhou, J.; Sun, J. Leaky intestine and impaired microbiome in an amyotrophic lateral sclerosis mouse model. Physiol. Rep. 2015, 3, e12356. [Google Scholar] [CrossRef] [Green Version]
- Zeng, Q.; Shen, J.; Chen, K.; Zhou, J.; Liao, Q.; Lu, K.; Yuan, J.; Bi, F. The alteration of gut microbiome and metabolism in amyotrophic lateral sclerosis patients. Sci. Rep. 2020, 10, 12998. [Google Scholar] [CrossRef]
- Niccolai, E.; Di Pilato, V.; Nannini, G.; Baldi, S.; Russo, E.; Zucchi, E.; Martinelli, I.; Menicatti, M.; Bartolucci, G.; Mandrioli, J.; et al. The gut microbiota-immunity axis in ALS: A role in deciphering disease heterogeneity? Biomedicines 2021, 9, 753. [Google Scholar] [CrossRef] [PubMed]
- Nicholson, K.; Bjornevik, K.; Abu-Ali, G.; Chan, J.; Cortese, M.; Dedi, B.; Jeon, M.; Xavier, R.; Huttenhower, C.; Ascherio, A.; et al. The human gut microbiota in people with amyotrophic lateral sclerosis. Amyotroph. Lateral Scler. Front. Degener. 2021, 22, 186–194. [Google Scholar] [CrossRef]
- Garbuzova-Davis, S.; Haller, E.; Saporta, S.; Kolomey, I.; Nicosia, S.V.; Sanberg, P.R. Ultrastructure of blood-brain barrier and blood-spinal cord barrier in SOD1 mice modeling ALS. Brain Res. 2007, 1157, 126–137. [Google Scholar] [CrossRef]
- Abrahao, A.; Meng, Y.; Llinas, M.; Huang, Y.; Hamani, C.; Mainprize, T.; Aubert, I.; Heyn, C.; Black, S.E.; Hynynen, K.; et al. First-in-human trial of blood-brain barrier opening in amyotrophic lateral sclerosis using MR-guided focused ultrasound. Nat. Commun. 2019, 10, 4373. [Google Scholar] [CrossRef] [Green Version]
- Barp, A.; Gerardi, F.; Lizio, A.; Sansone, V.A.; Lunetta, C. Emerging drugs for the treatment of amyotrophic lateral sclerosis: A focus on recent phase 2 trials. Expert Opin. Emerg. Drugs 2020, 25, 145–164. [Google Scholar] [CrossRef] [PubMed]
- Jaiswal, M.K. Riluzole and edaravone: A tale of two amyotrophic lateral sclerosis drugs. Med. Res. Rev. 2019, 39, 733–748. [Google Scholar] [CrossRef] [PubMed]
- Chang, C.Y.; Ting, H.C.; Liu, C.A.; Su, H.L.; Chiou, T.W.; Lin, S.Z.; Harn, H.J.; Ho, T.J. Induced pluripotent stem cell (iPSC)-based neurodegenerative disease models for phenotype recapitulation and drug screening. Molecules 2020, 25, 2000. [Google Scholar] [CrossRef]
- Vasques, J.F.; Mendez-Otero, R.; Gubert, F. Modeling ALS using iPSCs: Is it possible to reproduce the phenotypic variations observed in patients in vitro? Regen. Med. 2020, 15, 1919–1933. [Google Scholar] [CrossRef]
- Bonaventura, G.; Iemmolo, R.; Attaguile, G.A.; La Cognata, V.; Pistone, B.S.; Raudino, G.; D’Agata, V.; Cantarella, G.; Barcellona, M.L.; Cavallaro, S. iPSCs: A preclinical drug research tool for neurological disorders. Int. J. Mol. Sci. 2021, 22, 4596. [Google Scholar] [CrossRef]
- Ferraiuolo, L.; Maragakis, N.J. Mini-Review: Induced pluripotent stem cells and the search for new cell-specific ALS therapeutic targets. Neurosci. Lett. 2021, 755, 135911. [Google Scholar] [CrossRef]
- Je, G.; Keyhanian, K.; Ghasemi, M. Overview of stem cells therapy in amyotrophic lateral sclerosis. Neurol. Res. 2021, 43, 616–632. [Google Scholar] [CrossRef] [PubMed]
- Wang, L.; Yi, F.; Fu, L.; Yang, J.; Wang, S.; Wang, Z.; Suzuki, K.; Sun, L.; Xu, X.; Yu, Y.; et al. CRISPR/Cas9-mediated targeted gene correction in amyotrophic lateral sclerosis patient iPSCs. Protein Cell 2017, 8, 365–378. [Google Scholar] [CrossRef]
- Osaki, T.; Uzel, S.G.M.; Kamm, R.D. Microphysiological 3D model of amyotrophic lateral sclerosis (ALS) from human iPS-derived muscle cells and optogenetic motor neurons. Sci. Adv. 2018, 4, eaat5847. [Google Scholar] [CrossRef] [Green Version]
- Edmond, E.C.; Stagg, C.J.; Turner, M.R. Therapeutic non-invasive brain stimulation in amyotrophic lateral sclerosis: Rationale, methods and experience. J. Neurol. Neurosurg. Psychiatry 2019, 90, 1131–1138. [Google Scholar] [CrossRef]
- Barber, S.C.; Shaw, P.J. Oxidative stress in ALS: Key role in motor neuron injury and therapeutic target. Free Radic. Biol. Med. 2010, 48, 629–641. [Google Scholar] [CrossRef]
- Obrador, E.; Salvador, R.; López-Blanch, R.; Jihad-Jebbar, A.; Vallés, S.L.; Estrela, J.M. Oxidative stress, neuroinflammation and mitochondria in the pathophysiology of amyotrophic lateral sclerosis. Antioxidants 2020, 9, 901. [Google Scholar] [CrossRef] [PubMed]
- Harley, J.; Clarke, B.E.; Patani, R. The interplay of RNA binding proteins, oxidative stress and mitochondrial dysfunction in ALS. Antioxidants 2021, 10, 552. [Google Scholar] [CrossRef] [PubMed]
- Obrador, E.; Salvador-Palmer, R.; López-Blanch, R.; Jihad-Jebbar, A.; Vallés, S.L.; Estrela, J.M. The link between oxidative stress, redox status, bioenergetics and mitochondria in the pathophysiology of ALS. Int. J. Mol. Sci. 2021, 22, 6352. [Google Scholar] [CrossRef]
- Watanabe, K.; Morinaka, Y.; Iseki, K.; Watanabe, T.; Yuki, S.; Nishi, H. Structure-activity relationship of 3-methyl-1-phenyl-2-pyrazolin-5-one (edaravone). Redox Rep. 2003, 8, 151–155. [Google Scholar] [CrossRef]
- Watanabe, K.; Tanaka, M.; Yuki, S.; Hirai, M.; Yamamoto, Y. How is edaravone effective against acute ischemic stroke and amyotrophic lateral sclerosis? J. Clin. Biochem. Nutr. 2018, 62, 20–38. [Google Scholar] [CrossRef] [Green Version]
- Nakagawa, H.; Ohyama, R.; Kimata, A.; Suzuki, T.; Miyata, N. Hydroxyl radical scavenging by edaravone derivatives: Efficient scavenging by 3-methyl-1-(pyridin-2-yl)-5-pyrazolone with an intramolecular base. Bioorg. Med. Chem. Lett. 2006, 16, 5939–5942. [Google Scholar] [CrossRef]
- Nayak, M.; Batchu, H.; Batra, S. Straightforward copper-catalyzed synthesis of pyrrolopyrazoles from halogenated pyrazolecarbaldehydes. Tetrahedron Lett. 2012, 53, 4206–4208. [Google Scholar] [CrossRef]
- Vijay, K.; Nandi, C.; Samant, S.D. Synthesis of a dihydroquinoline based merocyanine as a ‘naked eye’ and ‘fluorogenic’ sensor for hydrazine hydrate in aqueous medium and hydrazine gas. RSC Adv. 2014, 4, 30712–30717. [Google Scholar] [CrossRef]
- Yoshino, H.; Kimura, A. Investigation of the therapeutic effects of edaravone, a free radical scavenger, on amyotrophic lateral sclerosis (Phase II study). Amyotroph. Lateral Scler. 2006, 7, 241–245. [Google Scholar] [CrossRef]
- Writing Group; Edaravone (MCI-186) ALS 19 Study Group. 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. [Google Scholar] [CrossRef]
- Takei, K.; Watanabe, K.; Yuki, S.; Akimoto, M.; Sakata, T.; Palumbo, J. Edaravone and its clinical development for amyotrophic lateral sclerosis. Amyotroph. Lateral Scler. Front. Degener. 2017, 18, 5–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dash, R.P.; Babu, R.J.; Srinivas, N.R. Two decades-long journey from riluzole to edaravone: Revisiting the clinical pharmacokinetics of the only two amyotrophic lateral sclerosis therapeutics. Clin. Pharmacokinet. 2018, 57, 1385–1398. [Google Scholar] [CrossRef] [PubMed]
- Bailly, C.; Hecquet, P.E.; Kouach, M.; Thuru, X.; Goossens, J.F. Chemical reactivity and uses of 1-phenyl-3-methyl-5-pyrazolone (PMP), also known as edaravone. Bioorg. Med. Chem. 2020, 28, 115463. [Google Scholar] [CrossRef]
- Pattee, G.L.; Post, G.R.; Gerber, R.E.; Bennett, J.P., Jr. Reduction of oxidative stress in amyotrophic lateral sclerosis following pramipexole treatment. Amyotroph. Lateral Scler. Other Mot. Neuron Disord. 2003, 4, 90–95. [Google Scholar] [CrossRef]
- Danzeisen, R.; Schwalenstoecker, B.; Gillardon, F.; Buerger, E.; Krzykalla, V.; Klinder, K.; Schild, L.; Hengerer, B.; Ludolph, A.C.; Dorner-Ciossek, C.; et al. Targeted antioxidative and neuroprotective properties of the dopamine agonist pramipexole and its nondopaminergic enantiomer SND919CL2x [(+)2-amino-4,5,6,7-tetrahydro-6-Lpropylamino-benzathiazole dihydrochloride]. J. Pharmacol. Exp. Ther. 2006, 316, 189–199. [Google Scholar] [CrossRef] [Green Version]
- Cudkowicz, M.E.; van den Berg, L.H.; Shefner, J.M.; Mitsumoto, H.; Mora, J.S.; Ludolph, A.; Hardiman, O.; Bozik, M.E.; Ingersoll, E.W.; Archibald, D.; et al. Dexpramipexole versus placebo for patients with amyotrophic lateral sclerosis (EMPOWER): A randomised, double-blind, phase 3 trial. Lancet Neurol. 2013, 12, 1059–1067. [Google Scholar] [CrossRef]
- Cudkowicz, M.; Bozik, M.E.; Ingersoll, E.W.; Miller, R.; Mitsumoto, H.; Shefner, J.; Moore, D.H.; Schoenfeld, D.; Mather, J.L.; Archibald, D.; et al. The effects of dexpramipexole (KNS-760704) in individuals with amyotrophic lateral sclerosis. Nat. Med. 2011, 17, 1652–1656. [Google Scholar] [CrossRef] [PubMed]
- Tanaka, K.; Kanno, T.; Yanagisawa, Y.; Yasutake, K.; Hadano, S.; Yoshii, F.; Ikeda, J.E. Bromocriptine methylate suppresses glial inflammation and moderates disease progression in a mouse model of amyotrophic lateral sclerosis. Exp. Neurol. 2011, 232, 41–52. [Google Scholar] [CrossRef] [PubMed]
- Nagata, E.; Ogino, M.; Iwamoto, K.; Kitagawa, Y.; Iwasaki, Y.; Yoshii, F.; Ikeda, J.E.; ALS Consortium Investigators. Bromocriptine mesylate attenuates amyotrophic lateral sclerosis: A phase 2a, randomized, double-blind, placebo-controlled research in Japanese patients. PLoS ONE 2016, 11, e0149509. [Google Scholar] [CrossRef] [PubMed]
- Fujimori, K.; Ishikawa, M.; Otomo, A.; Atsuta, N.; Nakamura, R.; Akiyama, T.; Hadano, S.; Aoki, M.; Saya, H.; Sobue, G.; et al. Modeling sporadic ALS in iPSC-derived motor neurons identifies a potential therapeutic agent. Nat. Med. 2018, 24, 1579–1589. [Google Scholar] [CrossRef] [PubMed]
- Morimoto, S.; Takahashi, S.; Fukushima, K.; Saya, H.; Suzuki, N.; Aoki, M.; Okano, H.; Nakahara, J. Ropinirole hydrochloride remedy for amyotrophic lateral sclerosis - Protocol for a randomized, double-blind, placebo-controlled, single-center, and open-label continuation phase I/IIa clinical trial (ROPALS trial). Regen. Ther. 2019, 11, 143–166. [Google Scholar] [CrossRef]
- Bucchia, M.; Ramirez, A.; Parente, V.; Simone, C.; Nizzardo, M.; Magri, F.; Dametti, S.; Corti, S. Therapeutic development in amyotrophic lateral sclerosis. Clin. Ther. 2015, 37, 668–680. [Google Scholar] [CrossRef]
- Waibel, S.; Reuter, A.; Malessa, S.; Blaugrund, E.; Ludolph, A.C. Rasagiline alone and in combination with riluzole prolongs survival in an ALS mouse model. J. Neurol. 2004, 251, 1080–1084. [Google Scholar] [CrossRef]
- Ludolph, A.C.; Schuster, J.; Dorst, J.; Dupuis, L.; Dreyhaupt, J.; Weishaupt, J.H.; Kassubek, J.; Weiland, U.; Petri, S.; Meyer, T.; et al. Safety and efficacy of rasagiline as an add-on therapy to riluzole in patients with amyotrophic lateral sclerosis: A randomised, double-blind, parallel-group, placebo-controlled, phase 2 trial. Lancet Neurol. 2018, 17, 681–688. [Google Scholar] [CrossRef]
- Statland, J.M.; Moore, D.; Wang, Y.; Walsh, M.; Mozaffar, T.; Elman, L.; Nations, S.P.; Mitsumoto, H.; Fernandes, J.A.; Saperstein, D.; et al. Rasagiline for amyotrophic lateral sclerosis: A randomized, controlled trial. Muscle Nerve 2019, 59, 201–207. [Google Scholar] [CrossRef]
- Lange, D.J.; Murphy, P.L.; Diamond, B.; Appel, V.; Lai, E.C.; Younger, D.S.; Appel, S.H. Selegiline is ineffective in a collaborative double-blind, placebo-controlled trial for treatment of amyotrophic lateral sclerosis. Arch. Neurol. 1998, 55, 93–96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Luo, F.; Sandhu, A.F.; Rungratanawanich, W.; Williams, G.E.; Akbar, M.; Zhou, S.; Song, B.J.; Wang, X. Melatonin and autophagy in aging-related neurodegenerative diseases. Int. J. Mol. Sci. 2020, 21, 7174. [Google Scholar] [CrossRef] [PubMed]
- Weishaupt, J.H.; Bartels, C.; Pölking, E.; Dietrich, J.; Rohde, G.; Poeggeler, B.; Mertens, N.; Sperling, S.; Bohn, M.; Hüther, G.; et al. Reduced oxidative damage in ALS by high-dose enteral melatonin treatment. J. Pineal Res. 2006, 41, 313–323. [Google Scholar] [CrossRef]
- Bald, E.M.; Nance, C.S.; Schultz, J.L. Melatonin may slow disease progression in amyotrophic lateral sclerosis: Findings from the pooled resource open-access ALS clinic trials database. Muscle Nerve 2021, 63, 572–576. [Google Scholar] [CrossRef]
- Choi, E.S.; Dokholyan, N.V. SOD1 oligomers in amyotrophic lateral sclerosis. Curr. Opin. Struct. Biol. 2021, 66, 225–230. [Google Scholar] [CrossRef] [PubMed]
- Eleutherio, E.C.A.; Silva Magalhães, R.S.; de Araújo Brasil, A.; Monteiro Neto, J.R.; de Holanda Paranhos, L. SOD1, more than just an antioxidant. Arch. Biochem. Biophys. 2021, 697, 108701. [Google Scholar] [CrossRef]
- Smith, R.A.; Milleri, T.M.; Yamanaka, K.; Monia, B.P.; Condon, T.P.; Hung, G.; Lobsiger, C.S.; Ward, C.M.; McAlonis-Downes, M.; Wei, H.; et al. Antisense oligonucleotide therapy for neurodegenerative disease. J. Clin. Investig. 2006, 116, 2290–2296. [Google Scholar] [CrossRef] [Green Version]
- Miller, T.M.; Pestronk, A.; David, W.; Rothstein, J.; Simpson, E.; Appel, S.H.; Andres, P.L.; Mahoney, K.; Allred, P.; Alexander, K.; et al. An antisense oligonucleotide against SOD1 delivered intrathecally for patients with SOD1 familial amyotrophic lateral sclerosis: A phase 1, randomised, first-in-man study. Lancet Neurol. 2013, 12, 435–442. [Google Scholar] [CrossRef] [Green Version]
- Miller, T.; Cudkowitz, M.; Shaw, P. Safety, PK, PD, and exploratory efficacy in single and multiple dose study of a SOD1 antisense oligonucleotide (BIIB067) administered to participants with ALS. In Proceedings of the American Academy of Neurology 71st Annual Meeting, Philadelphia, PA, USA, 4–10 May 2019. [Google Scholar]
- Kieran, D.; Kalmar, B.; Dick, J.R.; Riddoch-Contreras, J.; Burnstock, G.; Greensmith, L. Treatment with arimoclomol, a coinducer of heat shock proteins, delays disease progression in ALS mice. Nat. Med. 2004, 10, 402–405. [Google Scholar] [CrossRef]
- Kalmar, B.; Lu, C.H.; Greensmith, L. The role of heat shock proteins in Amyotrophic Lateral Sclerosis: The therapeutic potential of Arimoclomol. Pharmacol. Ther. 2014, 141, 40–54. [Google Scholar] [CrossRef]
- Benatar, M.; Wuu, J.; Andersen, P.M.; Atassi, N.; David, W.; Cudkowicz, M.; Schoenfeld, D. Randomized, double-blind, placebo-controlled trial of arimoclomol in rapidly progressive SOD1 ALS. Neurology 2018, 90, e565–e574. [Google Scholar] [CrossRef] [Green Version]
- Liscic, R.M.; Alberici, A.; Cairns, N.J.; Romano, M.; Buratti, E. From basic research to the clinic: Innovative therapies for ALS and FTD in the pipeline. Mol. Neurodegener. 2020, 15, 31. [Google Scholar] [CrossRef] [PubMed]
- Crippa, V.; D’Agostino, V.G.; Cristofani, R.; Rusmini, P.; Cicardi, M.E.; Messi, E.; Loffredo, R.; Pancher, M.; Piccolella, M.; Galbiati, M.; et al. Transcriptional induction of the heat shock protein B8 mediates the clearance of misfolded proteins responsible for motor neuron diseases. Sci. Rep. 2016, 6, 22827. [Google Scholar] [CrossRef] [PubMed]
- Mandrioli, J.; Crippa, V.; Cereda, C.; Bonetto, V.; Zucchi, E.; Gessani, A.; Ceroni, M.; Chio, A.; D’Amico, R.; Monsurrò, M.R.; et al. Proteostasis and ALS: Protocol for a phase II, randomised, double-blind, placebo-controlled, multicentre clinical trial for colchicine in ALS (Co-ALS). BMJ Open. 2019, 9, e028486. [Google Scholar] [CrossRef]
- Nikseresht, S.; Hilton, J.B.W.; Kysenius, K.; Liddell, J.R.; Crouch, P.J. Copper-ATSM as a treatment for ALS: Support from mutant SOD1 models and beyond. Life 2020, 10, 271. [Google Scholar] [CrossRef] [PubMed]
- McAllum, E.J.; Lim, N.K.; Hickey, J.L.; Paterson, B.M.; Donnelly, P.S.; Li, Q.X.; Liddell, J.R.; Barnham, K.J.; White, A.R.; Crouch, P.J. Therapeutic effects of CuII(atsm) in the SOD1-G37R mouse model of amyotrophic lateral sclerosis. Amyotroph. Lateral Scler. Front. Degener. 2013, 14, 586–590. [Google Scholar] [CrossRef]
- Hilton, J.B.W.; Kysenius, K.; Liddell, J.R.; Rautengarten, C.; Mercer, S.W.; Paul, B.; Beckman, J.S.; McLean, C.; White, A.R.; Donnelly, P.S.; et al. Disrupted copper availability in sporadic ALS: Implications for CuII(atsm) as a treatment option. BioRxiv 2020. [Google Scholar] [CrossRef]
- Lange, D.J.; Andersen, P.M.; Remanan, R.; Marklund, S.; Benjamin, D. Pyrimethamine decreases levels of SOD1 in leukocytes and cerebrospinal fluid of ALS patients: A phase I pilot study. Amyotroph. Lateral Scler. Front. Degener. 2013, 14, 199–204. [Google Scholar] [CrossRef]
- Lange, D.J.; Shahbazi, M.; Silani, V.; Ludolph, A.C.; Weishaupt, J.H.; Ajroud-Driss, S.; Fields, K.G.; Remanan, R.; Appel, S.H.; Morelli, C.; et al. Pyrimethamine significantly lowers cerebrospinal fluid Cu/Zn superoxide dismutase in amyotrophic lateral sclerosis patients with SOD1 mutations. Ann. Neurol. 2017, 81, 837–848. [Google Scholar] [CrossRef] [Green Version]
- Capper, M.J.; Wright, G.S.A.; Barbieri, L.; Luchinat, E.; Mercatelli, E.; McAlary, L.; Yerbury, J.J.; O’Neill, P.M.; Antonyuk, S.V.; Banci, L.; et al. The cysteine-reactive small molecule ebselen facilitates effective SOD1 maturation. Nat. Commun. 2018, 9, 1693. [Google Scholar] [CrossRef]
- Jhanji, R.; Behl, T.; Sehgal, A.; Bungau, S. Mitochondrial dysfunction and traffic jams in amyotrophic lateral sclerosis. Mitochondrion 2021, 58, 102–110. [Google Scholar] [CrossRef] [PubMed]
- Bordet, T.; Buisson, B.; Michaud, M.; Drouot, C.; Galéa, P.; Delaage, P.; Akentieva, N.P.; Evers, A.S.; Covey, D.F.; Ostuni, M.A.; et al. Identification and characterization of cholest-4-en-3-one, oxime (TRO19622), a novel drug candidate for amyotrophic lateral sclerosis. J. Pharmacol. Exp. Ther. 2007, 322, 709–720. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lenglet, T.; Lacomblez, L.; Abitbol, J.L.; Ludolph, A.; Mora, J.S.; Robberecht, W.; Shaw, P.J.; Pruss, R.M.; Cuvier, V.; Meininger, V.; et al. A phase II-III trial of olesoxime in subjects with amyotrophic lateral sclerosis. Eur. J. Neurol. 2014, 21, 529–536. [Google Scholar] [CrossRef]
- Klivenyi, P.; Ferrante, R.J.; Matthews, R.T.; Bogdanov, M.B.; Klein, A.M.; Andreassen, O.A.; Mueller, G.; Wermer, M.; Kaddurah-Daouk, R.; Beal, M.F. Neuroprotective effects of creatine in a transgenic animal model of amyotrophic lateral sclerosis. Nat. Med. 1999, 5, 347–350. [Google Scholar] [CrossRef] [PubMed]
- Groeneveld, G.J.; Veldink, J.H.; van der Tweel, I.; Kalmijn, S.; Beijer, C.; de Visser, M.; Wokke, J.H.; Franssen, H.; van den Berg, L.H. A randomized sequential trial of creatine in amyotrophic lateral sclerosis. Ann. Neurol. 2003, 53, 437–445. [Google Scholar] [CrossRef] [PubMed]
- Shefner, J.M.; Cudkowicz, M.E.; Schoenfeld, D.; Conrad, T.; Taft, J.; Chilton, M.; Urbinelli, L.; Qureshi, M.; Zhang, H.; Pestronk, A.; et al. A clinical trial of creatine in ALS. Neurology 2004, 63, 1656–1661. [Google Scholar] [CrossRef]
- Rosenfeld, J.; King, R.M.; Jackson, C.E.; Bedlack, R.S.; Barohn, R.J.; Dick, A.; Phillips, L.H.; Chapin, J.; Gelinas, D.F.; Lou, J.S. Creatine monohydrate in ALS: Effects on strength, fatigue, respiratory status and ALSFRS. Amyotroph. Lateral Scler. 2008, 9, 266–272. [Google Scholar] [CrossRef] [Green Version]
- Matthews, R.T.; Yang, L.; Browne, S.; Baik, M.; Beal, M.F. Coenzyme Q10 administration increases brain mitochondrial concentrations and exerts neuroprotective effects. Proc. Natl. Acad. Sci. USA 1998, 95, 8892–8897. [Google Scholar] [CrossRef] [Green Version]
- Kaufmann, P.; Thompson, J.L.; Levy, G.; Buchsbaum, R.; Shefner, J.; Krivickas, L.S.; Katz, J.; Rollins, Y.; Barohn, R.J.; Jackson, C.E.; et al. Phase II trial of CoQ10 for ALS finds insufficient evidence to justify phase III. Ann. Neurol. 2009, 66, 235–244. [Google Scholar] [CrossRef] [Green Version]
- Ramalho, T.C.; de Castro, A.A.; Tavares, T.S.; Silva, M.C.; Silva, D.R.; Cesar, P.H.; Santos, L.A.; da Cunha, E.F.F.; Nepovimova, E.; Kuca, K. Insights into the pharmaceuticals and mechanisms of neurological orphan diseases: Current status and future expectations. Prog. Neurobiol. 2018, 169, 135–157. [Google Scholar] [CrossRef]
- Ng Kee Kwong, K.C.; Mehta, A.R.; Nedergaard, M.; Chandran, S. Defining novel functions for cerebrospinal fluid in ALS pathophysiology. Acta Neuropathol. Commun. 2020, 8, 140. [Google Scholar] [CrossRef] [PubMed]
- Silva-Hucha, S.; Pastor, A.M.; Morcuende, S. Neuroprotective effect of vascular endothelial growth factor on motoneurons of the oculomotor system. Int. J. Mol. Sci. 2021, 22, 814. [Google Scholar] [CrossRef] [PubMed]
- Cheah, B.C.; Vucic, S.; Krishnan, A.V.; Kiernan, M.C. Riluzole, neuroprotection and amyotrophic lateral sclerosis. Curr. Med. Chem. 2010, 17, 1942–1959. [Google Scholar] [CrossRef]
- Mignani, S.; Majoral, J.P.; Desaphy, J.F.; Lentini, G. From Riluzole to dexpramipexole via substituted-benzothiazole derivatives for amyotrophic lateral sclerosis disease treatment: Case studies. Molecules 2020, 25, 3320. [Google Scholar] [CrossRef]
- Mathis, S.; Couratier, P.; Julian, A.; Vallat, J.M.; Corcia, P.; Le Masson, G. Management and therapeutic perspectives in amyotrophic lateral sclerosis. Expert Rev. Neurother. 2017, 17, 263–276. [Google Scholar] [CrossRef] [PubMed]
- Bensimon, G.; Lacomblez, L.; Meininger, V. ALS/Riluzole Study Group. A controlled trial of riluzole in amyotrophic lateral sclerosis. N. Engl. J. Med. 1994, 330, 585–591. [Google Scholar] [CrossRef]
- Lacomblez, L.; Bensimon, G.; Leigh, P.N.; Guillet, P.; Powe, L.; Durrleman, S.; Delumeau, J.C.; Meininger, V. A confirmatory dose-ranging study of riluzole in ALS. ALS/Riluzole Study Group-II. Neurology 1996, 47, S242–S250. [Google Scholar] [CrossRef]
- Dorst, J.; Ludolph, A.C.; Huebers, A. Disease-modifying and symptomatic treatment of amyotrophic lateral sclerosis. Ther. Adv. Neurol. Disord. 2017, 11, 1756285617734734. [Google Scholar] [CrossRef]
- Kumar, V.; Islam, A.; Hassan, M.I.; Ahmad, F. Therapeutic progress in amyotrophic lateral sclerosis-beginning to learning. Eur. J. Med. Chem. 2016, 121, 903–917. [Google Scholar] [CrossRef]
- Miller, R.G.; Moore, D.H., 2nd; Gelinas, D.F.; Dronsky, V.; Mendoza, M.; Barohn, R.J.; Bryan, W.; Ravits, J.; Yuen, E.; Neville, H.; et al. Phase III randomized trial of gabapentin in patients with amyotrophic lateral sclerosis. Neurology 2001, 56, 843–848. [Google Scholar] [CrossRef]
- Errante, L.D.; Petroff, O.A. Acute effects of gabapentin and pregabalin on rat forebrain cellular GABA, glutamate, and glutamine concentrations. Seizure 2003, 12, 300–306. [Google Scholar] [CrossRef] [Green Version]
- Cudkowicz, M.E.; Shefner, J.M.; Schoenfeld, D.A.; Brown, R.H., Jr.; Johnson, H.; Qureshi, M.; Jacobs, M.; Rothstein, J.D.; Appel, S.H.; Pascuzzi, R.M.; et al. A randomized, placebo-controlled trial of topiramate in amyotrophic lateral sclerosis. Neurology 2003, 61, 456–464. [Google Scholar] [CrossRef]
- Lanka, V.; Cudkowicz, M. Therapy development for ALS: Lessons learned and path forward. Amyotroph. Lateral Scler. 2008, 9, 131–140. [Google Scholar] [CrossRef]
- Akamatsu, M.; Yamashita, T.; Hirose, N.; Teramoto, S.; Kwak, S. The AMPA receptor antagonist perampanel robustly rescues amyotrophic lateral sclerosis (ALS) pathology in sporadic ALS model mice. Sci. Rep. 2016, 6, 28649. [Google Scholar] [CrossRef] [Green Version]
- Hotait, M.; Ismail, H.H.; Saab, G.E.; Salameh, J.S. An open label pilot study of the safety and tolerability of perampanel in amyotrophic lateral sclerosis. Muscle Nerve 2021, 64, 504–508. [Google Scholar] [CrossRef]
- Aizawa, H.; Kato, H.; Oba, K.; Kawahara, T.; Okubo, Y.; Saito, T.; Naito, M.; Urushitani, M.; Tamaoka, A.; Nakamagoe, K.; et al. Randomized phase 2 study of perampanel for sporadic amyotrophic lateral sclerosis. J. Neurol. 2022, 269, 885–896. [Google Scholar] [CrossRef] [PubMed]
- Paizs, M.; Tortarolo, M.; Bendotti, C.; Engelhardt, J.I.; Siklós, L. Talampanel reduces the level of motoneuronal calcium in transgenic mutant SOD1 mice only if applied presymptomatically. Amyotroph. Lateral Scler. 2011, 12, 340–344. [Google Scholar] [CrossRef]
- Pascuzzi, R.M.; Shefner, J.; Chappell, A.S.; Bjerke, J.S.; Tamura, R.; Chaudhry, V.; Clawson, L.; Haas, L.; Rothstein, J.D. A phase II trial of talampanel in subjects with amyotrophic lateral sclerosis. Amyotroph. Lateral Scler. 2010, 11, 266–271. [Google Scholar] [CrossRef] [PubMed]
- Wang, R.; Zhang, D. Memantine prolongs survival in an amyotrophic lateral sclerosis mouse model. Eur. J. Neurosci. 2005, 22, 2376–2380. [Google Scholar] [CrossRef]
- De Carvalho, M.; Pinto, S.; Costa, J.; Evangelista, T.; Ohana, B.; Pinto, A. A randomized, placebo-controlled trial of memantine for functional disability in amyotrophic lateral sclerosis. Amyotroph. Lateral Scler. 2010, 11, 456–460. [Google Scholar] [CrossRef]
- Gredal, O.; Werdelin, L.; Bak, S.; Christensen, P.B.; Boysen, G.; Kristensen, M.O.; Jespersen, J.H.; Regeur, L.; Hinge, H.H.; Jensen, T.S. A clinical trial of dextromethorphan in amyotrophic lateral sclerosis. Acta Neurol. Scand. 1997, 96, 8–13. [Google Scholar] [CrossRef]
- Brimson, J.M.; Brimson, S.; Chomchoei, C.; Tencomnao, T. Using sigma-ligands as part of a multi-receptor approach to target diseases of the brain. Expert Opin. Ther. Targets 2020, 24, 1009–1028. [Google Scholar] [CrossRef] [PubMed]
- Huynh, W.; Ahmed, R.; Mahoney, C.J.; Nguyen, C.; Tu, S.; Caga, J.; Loh, P.; Lin, C.S.; Kiernan, M.C. The impact of cognitive and behavioral impairment in amyotrophic lateral sclerosis. Expert Rev. Neurother. 2020, 20, 281–293. [Google Scholar] [CrossRef] [PubMed]
- Rothstein, J.D.; Patel, S.; Regan, M.R.; Haenggeli, C.; Huang, Y.H.; Bergles, D.E.; Jin, L.; Dykes Hoberg, M.; Vidensky, S.; Chung, D.S.; et al. Beta-lactam antibiotics offer neuroprotection by increasing glutamate transporter expression. Nature 2005, 433, 73–77. [Google Scholar] [CrossRef] [PubMed]
- Cudkowicz, M.E.; Titus, S.; Kearney, M.; Yu, H.; Sherman, A.; Schoenfeld, D.; Hayden, D.; Shui, A.; Brooks, B.; Conwit, R.; et al. Safety and efficacy of ceftriaxone for amyotrophic lateral sclerosis: A multi-stage, randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2014, 13, 1083–1091. [Google Scholar] [CrossRef] [Green Version]
- Yacila, G.; Sari, Y. Potential therapeutic drugs and methods for the treatment of amyotrophic lateral sclerosis. Curr. Med. Chem. 2014, 21, 3583–3593. [Google Scholar] [CrossRef] [Green Version]
- Miller, R.G.; Smith, S.A.; Murphy, J.R.; Brinkmann, J.R.; Graves, J.; Mendoza, M.; Sands, M.L.; Ringel, S.P. A clinical trial of verapamil in amyotrophic lateral sclerosis. Muscle Nerve 1996, 19, 511–515. [Google Scholar] [CrossRef]
- Miller, R.G.; Shepherd, R.; Dao, H.; Khramstov, A.; Mendoza, M.; Graves, J.; Smith, S. Controlled trial of nimodipine in amyotrophic lateral sclerosis. Neuromuscul. Disord. 1996, 6, 101–104. [Google Scholar] [CrossRef]
- Shibuya, K.; Misawa, S.; Kimura, H.; Noto, Y.; Sato, Y.; Sekiguchi, Y.; Iwai, Y.; Mitsuma, S.; Beppu, M.; Watanabe, K.; et al. A single blind randomized controlled clinical trial of mexiletine in amyotrophic lateral sclerosis: Efficacy and safety of sodium channel blocker phase II trial. Amyotroph. Lateral Scler. Front. Degener. 2015, 16, 353–358. [Google Scholar] [CrossRef]
- Weiss, M.D.; Macklin, E.A.; Simmons, Z.; Knox, A.S.; Greenblatt, D.J.; Atassi, N.; Graves, M.; Parziale, N.; Salameh, J.S.; Quinn, C.; et al. A randomized trial of mexiletine in ALS: Safety and effects on muscle cramps and progression. Neurology 2016, 86, 1474–1481. [Google Scholar] [CrossRef] [Green Version]
- Oskarsson, B.; Moore, D.; Mozaffar, T.; Ravits, J.; Wiedau-Pazos, M.; Parziale, N.; Joyce, N.C.; Mandeville, R.; Goyal, N.; Cudkowicz, M.E.; et al. Mexiletine for muscle cramps in amyotrophic lateral sclerosis: A randomized, double-blind crossover trial. Muscle Nerve 2018, 58, 42–48. [Google Scholar] [CrossRef]
- Goldsmith, D.R.; Wagstaff, A.J.; Ibbotson, T.; Perry, C.M. Lamotrigine: A review of its use in bipolar disorder. Drugs 2003, 63, 2029–2050. [Google Scholar] [CrossRef]
- Ryberg, H.; Askmark, H.; Persson, L.I. A double-blind randomized clinical trial in amyotrophic lateral sclerosis using lamotrigine: Effects on CSF glutamate, aspartate, branched-chain amino acid levels and clinical parameters. Acta Neurol. Scand. 2003, 108, 1–8. [Google Scholar] [CrossRef] [PubMed]
- Beattie, K.; Phadke, G.; Novakovic, J. Lamotrigine. In Profiles of Drug Substances, Excipients and Related Methodology; Brittain, H.G., Ed.; Elsevier: Amsterdam, The Netherlands, 2012; Volume 37, pp. 245–285. [Google Scholar]
- Wainger, B.J.; Kiskinis, E.; Mellin, C.; Wiskow, O.; Han, S.S.; Sandoe, J.; Perez, N.P.; Williams, L.A.; Lee, S.; Boulting, G.; et al. Intrinsic membrane hyperexcitability of amyotrophic lateral sclerosis patient-derived motor neurons. Cell Rep. 2014, 7, 1–11. [Google Scholar] [CrossRef] [Green Version]
- Wainger, B.J.; Macklin, E.A.; Vucic, S.; McIlduff, C.E.; Paganoni, S.; Maragakis, N.J.; Bedlack, R.; Goyal, N.A.; Rutkove, S.B.; Lange, D.J.; et al. Effect of ezogabine on cortical and spinal motor neuron excitability in amyotrophic lateral sclerosis: A randomized clinical trial. JAMA Neurol. 2021, 78, 186–196. [Google Scholar] [CrossRef] [PubMed]
- Kaji, R.; Kodama, M.; Imamura, A.; Hashida, T.; Kohara, N.; Ishizu, M.; Inui, K.; Kimura, J. Effect of ultrahigh-dose methylcobalamin on compound muscle action potentials in amyotrophic lateral sclerosis: A double-blind controlled study. Muscle Nerve 1998, 21, 1775–1778. [Google Scholar] [CrossRef]
- Ikeda, K.; Iwasaki, Y.; Kaji, R. Neuroprotective effect of ultra-high dose methylcobalamin in wobbler mouse model of amyotrophic lateral sclerosis. J. Neurol. Sci. 2015, 354, 70–74. [Google Scholar] [CrossRef]
- Kaji, R.; Imai, T.; Iwasaki, Y.; Okamoto, K.; Nakagawa, M.; Ohashi, Y.; Takase, T.; Hanada, T.; Shimizu, H.; Tashiro, K.; et al. Ultra-high-dose methylcobalamin in amyotrophic lateral sclerosis: A long-term phase II/III randomised controlled study. J. Neurol. Neurosurg. Psychiatry 2019, 90, 451–457. [Google Scholar] [CrossRef] [Green Version]
- Levine, B.; Sinha, S.; Kroemer, G. Bcl-2 family members: Dual regulators of apoptosis and autophagy. Autophagy 2008, 4, 600–606. [Google Scholar] [CrossRef] [Green Version]
- Amin, A.; Perera, N.D.; Beart, P.M.; Turner, B.J.; Shabanpoor, F. Amyotrophic lateral sclerosis and autophagy: Dysfunction and therapeutic targeting. Cells 2020, 9, 2413. [Google Scholar] [CrossRef]
- Wang, J. Regulation of cell death by the Abl tyrosine kinase. Oncogene 2000, 19, 5643–5650. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schlatterer, S.D.; Acker, C.M.; Davies, P. c-Abl in neurodegenerative disease. J. Mol. Neurosci. 2011, 45, 445–452. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Karagiannis, P.; Inoue, H. ALS, a cellular whodunit on motor neuron degeneration. Mol. Cell Neurosci. 2020, 107, 103524. [Google Scholar] [CrossRef] [PubMed]
- Kim, B.W.; Jeong, Y.E.; Wong, M.; Martin, L.J. DNA damage accumulates and responses are engaged in human ALS brain and spinal motor neurons and DNA repair is activatable in iPSC-derived motor neurons with SOD1 mutations. Acta Neuropathol. Commun. 2020, 8, 7. [Google Scholar] [CrossRef] [Green Version]
- Rojas, F.; Gonzalez, D.; Cortes, N.; Ampuero, E.; Hernández, D.E.; Fritz, E.; Abarzua, S.; Martinez, A.; Elorza, A.A.; Alvarez, A.; et al. Reactive oxygen species trigger motoneuron death in non-cell-autonomous models of ALS through activation of c-Abl signaling. Front. Cell Neurosci. 2015, 9, 203. [Google Scholar] [CrossRef] [Green Version]
- Katsumata, R.; Ishigaki, S.; Katsuno, M.; Kawai, K.; Sone, J.; Huang, Z.; Adachi, H.; Tanaka, F.; Urano, F.; Sobue, G. c-Abl inhibition delays motor neuron degeneration in the G93A mouse, an animal model of amyotrophic lateral sclerosis. PLoS ONE 2012, 7, e46185. [Google Scholar] [CrossRef]
- Imamura, K.; Izumi, Y.; Watanabe, A.; Tsukita, K.; Woltjen, K.; Yamamoto, T.; Hotta, A.; Kondo, T.; Kitaoka, S.; Ohta, A.; et al. The Src/c-Abl pathway is a potential therapeutic target in amyotrophic lateral sclerosis. Sci. Transl. Med. 2017, 9, eaaf3962. [Google Scholar] [CrossRef] [Green Version]
- Imamura, K.; Izumi, Y.; Banno, H.; Uozumi, R.; Morita, S.; Egawa, N.; Ayaki, T.; Nagai, M.; Nishiyama, K.; Watanabe, Y.; et al. Induced pluripotent stem cell-based Drug Repurposing for Amyotrophic lateral sclerosis Medicine (iDReAM) study: Protocol for a phase I dose escalation study of bosutinib for amyotrophic lateral sclerosis patients. BMJ Open 2019, 9, e033131. [Google Scholar] [CrossRef] [Green Version]
- Amaral, J.D.; Viana, R.J.; Ramalho, R.M.; Steer, C.J.; Rodrigues, C.M. Bile acids: Regulation of apoptosis by ursodeoxycholic acid. J. Lipid Res. 2009, 50, 1721–1734. [Google Scholar] [CrossRef] [Green Version]
- Elia, A.E.; Lalli, S.; Monsurrò, M.R.; Sagnelli, A.; Taiello, A.C.; Reggiori, B.; La Bella, V.; Tedeschi, G.; Albanese, A. Tauroursodeoxycholic acid in the treatment of patients with amyotrophic lateral sclerosis. Eur. J. Neurol. 2016, 23, 45–52. [Google Scholar] [CrossRef]
- Palomo, V.; Nozal, V.; Rojas-Prats, E.; Gil, C.; Martinez, A. Protein kinase inhibitors for amyotrophic lateral sclerosis therapy. Br. J. Pharmacol. 2021, 178, 1316–1335. [Google Scholar] [CrossRef]
- Mandrioli, J.; D’Amico, R.; Zucchi, E.; Gessani, A.; Fini, N.; Fasano, A.; Caponnetto, C.; Chiò, A.; Dalla Bella, E.; Lunetta, C.; et al. Rapamycin treatment for amyotrophic lateral sclerosis: Protocol for a phase II randomized, double-blind, placebo-controlled, multicenter, clinical trial (RAP-ALS trial). Medicine 2018, 97, e11119. [Google Scholar] [CrossRef]
- Fornai, F.; Longone, P.; Cafaro, L.; Kastsiuchenka, O.; Ferrucci, M.; Manca, M.L.; Lazzeri, G.; Spalloni, A.; Bellio, N.; Lenzi, P.; et al. Lithium delays progression of amyotrophic lateral sclerosis. Proc. Natl. Acad. Sci. USA 2008, 105, 2052–2057. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Morrison, K.E.; Dhariwal, S.; Hornabrook, R.; Savage, L.; Burn, D.J.; Khoo, T.K.; Kelly, J.; Murphy, C.L.; Al-Chalabi, A.; et al.; UKMND-LiCALS Study Group Lithium in patients with amyotrophic lateral sclerosis (LiCALS): A phase 3 multicentre, randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2013, 12, 339–345. [Google Scholar]
- Aggarwal, S.P.; Zinman, L.; Simpson, E.; McKinley, J.; Jackson, K.E.; Pinto, H.; Kaufman, P.; Conwit, R.A.; Schoenfeld, D.; Shefner, J.; et al. Safety and efficacy of lithium in combination with riluzole for treatment of amyotrophic lateral sclerosis: A randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2010, 9, 481–488. [Google Scholar] [CrossRef] [Green Version]
- Sugai, F.; Yamamoto, Y.; Miyaguchi, K.; Zhou, Z.; Sumi, H.; Hamasaki, T.; Goto, M.; Sakoda, S. Benefit of valproic acid in suppressing disease progression of ALS model mice. Eur. J. Neurosci. 2004, 20, 3179–3183. [Google Scholar] [CrossRef]
- Piepers, S.; Veldink, J.H.; de Jong, S.W.; van der Tweel, I.; van der Pol, W.L.; Uijtendaal, E.V.; Schelhaas, H.J.; Scheffer, H.; de Visser, M.; de Jong, J.M.; et al. Randomized sequential trial of valproic acid in amyotrophic lateral sclerosis. Ann. Neurol. 2009, 66, 227–234. [Google Scholar] [CrossRef] [PubMed]
- Leng, Y.; Liang, M.H.; Ren, M.; Marinova, Z.; Leeds, P.; Chuang, D.M. Synergistic neuroprotective effects of lithium and valproic acid or other histone deacetylase inhibitors in neurons: Roles of glycogen synthase kinase-3 inhibition. J. Neurosci. 2008, 28, 2576–2588. [Google Scholar] [CrossRef] [Green Version]
- Feng, H.L.; Leng, Y.; Ma, C.H.; Zhang, J.; Ren, M.; Chuang, D.M. Combined lithium and valproate treatment delays disease onset, reduces neurological deficits and prolongs survival in an amyotrophic lateral sclerosis mouse model. Neuroscience 2008, 155, 567–572. [Google Scholar] [CrossRef] [Green Version]
- Boll, M.C.; Bayliss, L.; Vargas-Cañas, S.; Burgos, J.; Montes, S.; Peñaloza-Solano, G.; Rios, C.; Alcaraz-Zubeldia, M. Clinical and biological changes under treatment with lithium carbonate and valproic acid in sporadic amyotrophic lateral sclerosis. J. Neurol. Sci. 2014, 340, 103–108. [Google Scholar] [CrossRef]
- Freeman, L.C.; Ting, J.P. The pathogenic role of the inflammasome in neurodegenerative diseases. J. Neurochem. 2016, 136, 29–38. [Google Scholar] [CrossRef]
- Tang, Y.; Le, W. Differential roles of M1 and M2 microglia in neurodegenerative diseases. Mol. Neurobiol. 2016, 53, 1181–1194. [Google Scholar] [CrossRef] [PubMed]
- Du, L.; Zhang, Y.; Chen, Y.; Zhu, J.; Yang, Y.; Zhang, H.L. Role of microglia in neurological disorders and their potentials as a therapeutic target. Mol. Neurobiol. 2017, 54, 7567–7584. [Google Scholar] [CrossRef] [PubMed]
- Geloso, M.C.; Corvino, V.; Marchese, E.; Serrano, A.; Michetti, F.; D’Ambrosi, N. The dual role of microglia in ALS: Mechanisms and therapeutic approaches. Front. Aging Neurosci. 2017, 9, 242. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kulczyńska-Przybik, A.; Mroczko, P.; Dulewicz, M.; Mroczko, B. The implication of reticulons (RTNs) in neurodegenerative diseases: From molecular mechanisms to potential diagnostic and therapeutic approaches. Int. J. Mol. Sci. 2021, 22, 4630. [Google Scholar] [CrossRef]
- Mizwicki, M.T.; Fiala, M.; Magpantay, L.; Aziz, N.; Sayre, J.; Liu, G.; Siani, A.; Chan, D.; Martinez-Maza, O.; Chattopadhyay, M.; et al. Tocilizumab attenuates inflammation in ALS patients through inhibition of IL6 receptor signaling. Am. J. Neurodegener. Dis. 2012, 1, 305–315. [Google Scholar]
- Fiala, M.; Mizwicki, M.T.; Weitzman, R.; Magpantay, L.; Nishimoto, N. Tocilizumab infusion therapy normalizes inflammation in sporadic ALS patients. Am. J. Neurodegener. Dis. 2013, 2, 129–139. [Google Scholar]
- Milligan, C.; Atassi, N.; Babu, S.; Barohn, R.J.; Caress, J.B.; Cudkowicz, M.E.; Evora, A.; Hawkins, G.A.; Wosiski-Kuhn, M.; Macklin, E.A.; et al. Tocilizumab is safe and tolerable and reduces C-reactive protein concentrations in the plasma and cerebrospinal fluid of ALS patients. Muscle Nerve 2021, 64, 309–320. [Google Scholar] [CrossRef]
- Maier, A.; Deigendesch, N.; Müller, K.; Weishaupt, J.H.; Krannich, A.; Röhle, R.; Meissner, F.; Molawi, K.; Münch, C.; Holm, T.; et al. Interleukin-1 antagonist anakinra in amyotrophic lateral sclerosis-A Pilot Study. PLoS ONE 2015, 10, e0139684. [Google Scholar] [CrossRef]
- Kukharsky, M.S.; Skvortsova, V.I.; Bachurin, S.O.; Buchman, V.L. In a search for efficient treatment for amyotrophic lateral sclerosis: Old drugs for new approaches. Med. Res. Rev. 2021, 41, 2804–2822. [Google Scholar] [CrossRef]
- Vahsen, B.F.; Gray, E.; Thompson, A.G.; Ansorge, O.; Anthony, D.C.; Cowley, S.A.; Talbot, K.; Turner, M.R. Non-neuronal cells in amyotrophic lateral sclerosis-from pathogenesis to biomarkers. Nat. Rev. Neurol. 2021, 17, 333–348. [Google Scholar] [CrossRef]
- Trias, E.; Ibarburu, S.; Barreto-Núñez, R.; Babdor, J.; Maciel, T.T.; Guillo, M.; Gros, L.; Dubreuil, P.; Díaz-Amarilla, P.; Cassina, P.; et al. Post-paralysis tyrosine kinase inhibition with masitinib abrogates neuroinflammation and slows disease progression in inherited amyotrophic lateral sclerosis. J. Neuroinflammation 2016, 13, 177. [Google Scholar] [CrossRef] [PubMed]
- Trias, E.; King, P.H.; Si, Y.; Kwon, Y.; Varela, V.; Ibarburu, S.; Kovacs, M.; Moura, I.C.; Beckman, J.S.; Hermine, O.; et al. Mast cells and neutrophils mediate peripheral motor pathway degeneration in ALS. JCI Insight 2018, 3, e123249. [Google Scholar] [CrossRef] [PubMed]
- Mora, J.S.; Genge, A.; Chio, A.; Estol, C.J.; Chaverri, D.; Hernández, M.; Marín, S.; Mascias, J.; Rodriguez, G.E.; Povedano, M.; et al. Masitinib as an add-on therapy to riluzole in patients with amyotrophic lateral sclerosis: A randomized clinical trial. Amyotroph. Lateral Scler. Front. Degener. 2020, 21, 5–14. [Google Scholar] [CrossRef] [Green Version]
- Chen, Y.; Wang, H.; Ying, Z.; Gao, Q. Ibudilast enhances the clearance of SOD1 and TDP-43 aggregates through TFEB-mediated autophagy and lysosomal biogenesis: The new molecular mechanism of ibudilast and its implication for neuroprotective therapy. Biochem. Biophys. Res. Commun. 2020, 526, 231–238. [Google Scholar] [CrossRef]
- Babu, S.; Hightower, B.G.; Chan, J.; Zürcher, N.R.; Kivisäkk, P.; Tseng, C.J.; Sanders, D.L.; Robichaud, A.; Banno, H.; Evora, A.; et al. Ibudilast (MN-166) in amyotrophic lateral sclerosis- An open label, safety and pharmacodynamic trial. Neuroimage Clin. 2021, 30, 102672. [Google Scholar] [CrossRef]
- Berry, J.D.; Paganoni, S.; Atassi, N.; Macklin, E.A.; Goyal, N.; Rivner, M.; Simpson, E.; Appel, S.; Grasso, D.L.; Mejia, N.I.; et al. Phase IIa trial of fingolimod for amyotrophic lateral sclerosis demonstrates acceptable acute safety and tolerability. Muscle Nerve 2017, 56, 1077–1084. [Google Scholar] [CrossRef] [PubMed]
- Lynch, A.M.; Clevel, M.; Prinjha, R.; Kumar, U.; Stubbs, R.; Wuerthner, J. Non-clinical development of ozanezumab: A humanised antibody targeting the amino terminus of neurite outgrowth inhibitor A (Nogo-A). Toxicol. Res. 2015, 4, 1333. [Google Scholar] [CrossRef]
- Jokic, N.; Gonzalez de Aguilar, J.L.; Dimou, L.; Lin, S.; Fergani, A.; Ruegg, M.A.; Schwab, M.E.; Dupuis, L.; Loeffler, J.P. The neurite outgrowth inhibitor Nogo-A promotes denervation in an amyotrophic lateral sclerosis model. EMBO Rep. 2006, 7, 1162–1167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Scaricamazza, S.; Salvatori, I.; Ferri, A.; Valle, C. Skeletal Muscle in ALS: An unappreciated therapeutic opportunity? Cells 2021, 10, 525. [Google Scholar] [CrossRef]
- Meininger, V.; Pradat, P.F.; Corse, A.; Al-Sarraj, S.; Rix Brooks, B.; Caress, J.B.; Cudkowicz, M.; Kolb, S.J.; Lange, D.; Leigh, P.N.; et al. Safety, pharmacokinetic, and functional effects of the Nogo-A monoclonal antibody in amyotrophic lateral sclerosis: A randomized, first-in-human clinical trial. PLoS ONE 2014, 9, e97803. [Google Scholar] [CrossRef] [Green Version]
- Meininger, V.; Genge, A.; van den Berg, L.H.; Robberecht, W.; Ludolph, A.; Chio, A.; Kim, S.H.; Leigh, P.N.; Kiernan, M.C.; Shefner, J.M.; et al. Safety and efficacy of ozanezumab in patients with amyotrophic lateral sclerosis: A randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Neurol. 2017, 16, 208–216. [Google Scholar] [CrossRef] [Green Version]
- Koch, J.C.; Kuttler, J.; Maass, F.; Lengenfeld, T.; Zielke, E.; Bähr, M.; Lingor, P. Compassionate use of the ROCK inhibitor fasudil in three patients with amyotrophic lateral sclerosis. Front. Neurol. 2020, 11, 173. [Google Scholar] [CrossRef]
- Takata, M.; Tanaka, H.; Kimura, M.; Nagahara, Y.; Tanaka, K.; Kawasaki, K.; Seto, M.; Tsuruma, K.; Shimazawa, M.; Hara, H. Fasudil, a rho kinase inhibitor, limits motor neuron loss in experimental models of amyotrophic lateral sclerosis. Br. J. Pharmacol. 2013, 170, 341–351. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lingor, P.; Weber, M.; Camu, W.; Friede, T.; Hilgers, R.; Leha, A.; Neuwirth, C.; Günther, R.; Benatar, M.; Kuzma-Kozakiewicz, M.; et al. ROCK-ALS: Protocol for a randomized, placebo-controlled, double-blind phase IIa trial of safety, tolerability and efficacy of the rho kinase (ROCK) inhibitor fasudil in amyotrophic lateral sclerosis. Front. Neurol. 2019, 10, 293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wobst, H.J.; Mack, K.L.; Brown, D.G.; Brandon, N.J.; Shorter, J. The clinical trial landscape in amyotrophic lateral sclerosis—Past, present, and future. Med. Res. Rev. 2020, 40, 1352–1384. [Google Scholar] [CrossRef]
- Hwee, D.T.; Kennedy, A.; Ryans, J.; Russell, A.J.; Jia, Z.; Hinken, A.C.; Morgans, D.J.; Malik, F.I.; Jasper, J.R. Fast skeletal muscle troponin activator tirasemtiv increases muscle function and performance in the B6SJL-SOD1G93A ALS mouse model. PLoS ONE 2014, 9, e96921. [Google Scholar] [CrossRef] [PubMed]
- Shefner, J.M.; Wolff, A.A.; Meng, L.; Bian, A.; Lee, J.; Barragan, D.; Andrews, J.A. BENEFIT-ALS Study Group. A randomized, placebo-controlled, double-blind phase IIb trial evaluating the safety and efficacy of tirasemtiv in patients with amyotrophic lateral sclerosis. Amyotroph. Lateral Scler. Front. Degener. 2016, 17, 426–435. [Google Scholar] [CrossRef]
- Andrews, J.A.; Cudkowicz, M.E.; Hardiman, O.; Meng, L.; Bian, A.; Lee, J.; Wolff, A.A.; Malik, F.I.; Shefner, J.M. VITALITY-ALS, a phase III trial of tirasemtiv, a selective fast skeletal muscle troponin activator, as a potential treatment for patients with amyotrophic lateral sclerosis: Study design and baseline characteristics. Amyotroph. Lateral Scler. Front. Degener. 2018, 19, 259–266. [Google Scholar] [CrossRef] [Green Version]
- Shefner, J.M.; Andrews, J.A.; Genge, A.; Jackson, C.; Lechtzin, N.; Miller, T.M.; Cockroft, B.M.; Meng, L.; Wei, J.; Wolff, A.A.; et al. A phase 2, double-blind, randomized, dose-ranging trial of reldesemtiv in patients with ALS. Amyotroph. Lateral Scler. Front. Degener. 2021, 22, 287–299. [Google Scholar] [CrossRef]
- MacGowan, D.J.; Scelsa, S.N.; Imperato, T.E.; Liu, K.N.; Baron, P.; Polsky, B. A controlled study of reverse transcriptase in serum and CSF of HIV-negative patients with ALS. Neurology 2007, 68, 1944–1946. [Google Scholar] [CrossRef] [PubMed]
- McCormick, A.L.; Brown, R.H., Jr.; Cudkowicz, M.E.; Al-Chalabi, A.; Garson, J.A. Quantification of reverse transcriptase in ALS and elimination of a novel retroviral candidate. Neurology 2008, 70, 278–283. [Google Scholar] [CrossRef] [PubMed]
- Douville, R.; Liu, J.; Rothstein, J.; Nath, A. Identification of active loci of a human endogenous retrovirus in neurons of patients with amyotrophic lateral sclerosis. Ann. Neurol. 2011, 69, 141–151. [Google Scholar] [CrossRef]
- Castanedo-Vazquez, D.; Bosque-Varela, P.; Sainz-Pelayo, A.; Riancho, J. Infectious agents and amyotrophic lateral sclerosis: Another piece of the puzzle of motor neuron degeneration. J. Neurol. 2019, 266, 27–36. [Google Scholar] [CrossRef] [PubMed]
- Gold, J.; Rowe, D.B.; Kiernan, M.C.; Vucic, S.; Mathers, S.; van Eijk, R.P.A.; Nath, A.; Garcia Montojo, M.; Norato, G.; Santamaria, U.A.; et al. Safety and tolerability of Triumeq in amyotrophic lateral sclerosis: The lighthouse trial. Amyotroph. Lateral Scler. Front. Degener. 2019, 20, 595–604. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nagtegaal, I.D.; Lakke, E.A.; Marani, E. Trophic and tropic factors in the development of the central nervous system. Arch. Physiol. Biochem. 1998, 106, 161–202. [Google Scholar] [CrossRef] [PubMed]
- Zinman, L.; Cudkowicz, M. Emerging targets and treatments in amyotrophic lateral sclerosis. Lancet Neurol. 2011, 10, 481–490. [Google Scholar] [CrossRef]
- Nagel, G.; Peter, R.S.; Rosenbohm, A.; Koenig, W.; Dupuis, L.; Rothenbacher, D.; Ludolph, A.C. Association of Insulin-like Growth Factor 1 Concentrations with Risk for and Prognosis of Amyotrophic Lateral Sclerosis—Results from the ALS Registry Swabia. Sci. Rep. 2020, 10, 736. [Google Scholar] [CrossRef]
- Azzouz, M.; Ralph, G.S.; Storkebaum, E.; Walmsley, L.E.; Mitrophanous, K.A.; Kingsman, S.M.; Carmeliet, P.; Mazarakis, N.D. VEGF delivery with retrogradely transported lentivector prolongs survival in a mouse ALS model. Nature 2004, 429, 413–417. [Google Scholar] [CrossRef]
- Sun, W.; Funakoshi, H.; Nakamura, T. Overexpression of HGF retards disease progression and prolongs life span in a transgenic mouse model of ALS. J. Neurosci. 2002, 22, 6537–6548. [Google Scholar] [CrossRef]
- Kitamura, K.; Nagoshi, N.; Tsuji, O.; Matsumoto, M.; Okano, H.; Nakamura, M. 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. [Google Scholar] [CrossRef] [Green Version]
- Le Pichon, C.E.; Dominguez, S.L.; Solanoy, H.; Ngu, H.; Lewin-Koh, N.; Chen, M.; Eastham-Anderson, J.; Watts, R.; Scearce-Levie, K. EGFR inhibitor erlotinib delays disease progression but does not extend survival in the SOD1 mouse model of ALS. PLoS ONE 2013, 8, e62342. [Google Scholar] [CrossRef]
- Chen, T.; Benmohamed, R.; Arvanites, A.C.; Ralay Ranaivo, H.; Morimoto, R.I.; Ferrante, R.J.; Watterson, D.M.; Kirsch, D.R.; Silverman, R.B. Arylsulfanyl pyrazolones block mutant SOD1-G93A aggregation. Potential application for the treatment of amyotrophic lateral sclerosis. Bioorg. Med. Chem. 2011, 19, 613–622. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Neymotin, A.; Calingasan, N.Y.; Wille, E.; Naseri, N.; Petri, S.; Damiano, M.; Liby, K.T.; Risingsong, R.; Sporn, M.; Beal, M.F.; et al. Neuroprotective effect of Nrf2/ARE activators, CDDO ethylamide and CDDO trifluoroethylamide, in a mouse model of amyotrophic lateral sclerosis. Free Radic. Biol. Med. 2011, 51, 88–96. [Google Scholar] [CrossRef] [Green Version]
- Zhang, W.; Benmohamed, R.; Arvanites, A.C.; Morimoto, R.I.; Ferrante, R.J.; Kirsch, D.R.; Silverman, R.B. Cyclohexane 1,3-diones and their inhibition of mutant SOD1-dependent protein aggregation and toxicity in PC12 cells. Bioorg. Med. Chem. 2012, 20, 1029–1045. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ahn, S.W.; Jeon, G.S.; Kim, M.J.; Shon, J.H.; Kim, J.E.; Shin, J.Y.; Kim, S.M.; Kim, S.H.; Ye, I.H.; Lee, K.W.; et al. Neuroprotective effects of JGK-263 in transgenic SOD1-G93A mice of amyotrophic lateral sclerosis. J. Neurol. Sci. 2014, 340, 112–116. [Google Scholar] [CrossRef] [PubMed]
- Tanaka, K.; Kanno, T.; Yanagisawa, Y.; Yasutake, K.; Inoue, S.; Hirayama, N.; Ikeda, J.E. A novel acylaminoimidazole derivative, WN1316, alleviates disease progression via suppression of glial inflammation in ALS mouse model. PLoS ONE 2014, 9, e87728. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Getter, T.; Zaks, I.; Barhum, Y.; Ben-Zur, T.; Böselt, S.; Gregoire, S.; Viskind, O.; Shani, T.; Gottlieb, H.; Green, O.; et al. A chemical chaperone-based drug candidate is effective in a mouse model of amyotrophic lateral sclerosis (ALS). ChemMedChem 2015, 10, 850–861. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Sever, B.; Ciftci, H.; DeMirci, H.; Sever, H.; Ocak, F.; Yulug, B.; Tateishi, H.; Tateishi, T.; Otsuka, M.; Fujita, M.; et al. Comprehensive Research on Past and Future Therapeutic Strategies Devoted to Treatment of Amyotrophic Lateral Sclerosis. Int. J. Mol. Sci. 2022, 23, 2400. https://doi.org/10.3390/ijms23052400
Sever B, Ciftci H, DeMirci H, Sever H, Ocak F, Yulug B, Tateishi H, Tateishi T, Otsuka M, Fujita M, et al. Comprehensive Research on Past and Future Therapeutic Strategies Devoted to Treatment of Amyotrophic Lateral Sclerosis. International Journal of Molecular Sciences. 2022; 23(5):2400. https://doi.org/10.3390/ijms23052400
Chicago/Turabian StyleSever, Belgin, Halilibrahim Ciftci, Hasan DeMirci, Hilal Sever, Firdevs Ocak, Burak Yulug, Hiroshi Tateishi, Takahisa Tateishi, Masami Otsuka, Mikako Fujita, and et al. 2022. "Comprehensive Research on Past and Future Therapeutic Strategies Devoted to Treatment of Amyotrophic Lateral Sclerosis" International Journal of Molecular Sciences 23, no. 5: 2400. https://doi.org/10.3390/ijms23052400
APA StyleSever, B., Ciftci, H., DeMirci, H., Sever, H., Ocak, F., Yulug, B., Tateishi, H., Tateishi, T., Otsuka, M., Fujita, M., & Başak, A. N. (2022). Comprehensive Research on Past and Future Therapeutic Strategies Devoted to Treatment of Amyotrophic Lateral Sclerosis. International Journal of Molecular Sciences, 23(5), 2400. https://doi.org/10.3390/ijms23052400