Skip to main content

Advertisement

Log in

Approaches to Remyelination Therapies in Multiple Sclerosis

  • Multiple Sclerosis and Related Disorders (J Graves, Section Editor)
  • Published:
Current Treatment Options in Neurology Aims and scope Submit manuscript

Abstract

Purpose of review

While there are a growing number of therapies targeting relapse prevention in multiple sclerosis (MS), there are no approved therapies promoting remyelination. Understanding endogenous myelin formation, remyelination strategies, pre-clinical models, and clinical outcomes is essential to the interpretation of current and future clinical trials of remyelinating agents.

Recent findings

Several recent clinical trials of remyelination therapies, including opicinumab, clemastine, and GSK239512, showed negative or modest results. These results could highlight challenges translating pre-clinical studies into subjects with MS and current strategies to measure remyelination.

Summary

Current approaches to remyelination include (1) blocking inhibitors of remyelination, (2) improving the clearance of myelin debris, (3) increasing the number of oligodendrocyte precursor cells (OPCs), and (4) stimulating OPC differentiation. To date, no therapies have led to robust remyelination. Future efforts to promote remyelination will likely require a combination of these mechanistic strategies.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

References and Recommended Reading

Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance

  1. Wang H, et al. miR-219 cooperates with miR-338 in myelination and promotes myelin repair in the CNS. Dev Cell. 2017;40(6):566–582.e5.

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Simons M, Nave K-A. Oligodendrocytes: myelination and axonal support. Cold Spring Harbor Perspectives in Biology. 2016;8(1):a020479.

    PubMed Central  Google Scholar 

  3. Hughes EG, et al. Oligodendrocyte progenitors balance growth with self-repulsion to achieve homeostasis in the adult brain. Nature Neuroscience. 2013;16(6):668–676-676.

    CAS  PubMed  PubMed Central  Google Scholar 

  4. • Tsai H-H, et al. Oligodendrocyte precursors migrate along vasculature in the developing nervous system. Science. 2016;351(6271):379–384-384 This paper shows that oligodendrocyte precursor cells in both mouse brain and human cortex rely upon physical interaction with the vasculature to migrate within the brain. This highlights the complex interactions between cerebral vasculature and oligodendrocytes in creating a permissive environment for myelination.

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Lee S, et al. A rapid and reproducible assay for modeling myelination by oligodendrocytes using engineered nanofibers. Nature Protocols. 2013;8(4):771.

    PubMed  PubMed Central  Google Scholar 

  6. Rosenberg SS, et al. The geometric and spatial constraints of the microenvironment induce oligodendrocyte differentiation. Proceedings of the National Academy of Sciences. 2008;105(38):14662–14667-14667.

    CAS  Google Scholar 

  7. Brinkmann BG, et al. Neuregulin-1/ErbB signaling serves distinct functions in myelination of the peripheral and central nervous system. Neuron. 2008;59(4):581–95.

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Schoenemann TP, Sheehan MJ, Glotzer DL. Prefrontal white matter volume is disproportionately larger in humans than in other primates. Nature Neuroscience. 2005;8(2):nn1394.

    Google Scholar 

  9. Stassart RM, et al. The axon-myelin unit in development and degenerative disease. Frontiers in Neuroscience. 2018;12:467.

    PubMed  PubMed Central  Google Scholar 

  10. Roth AD, Ivanova A, Colman DR. New observations on the compact myelin proteome. Neuron Glia Biology. 2006;2(1):15–21.

    PubMed  Google Scholar 

  11. Lee Y, et al. Oligodendroglia metabolically support axons and contribute to neurodegeneration. Nature. 2012;487(7408):443.

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Nijland PG, et al. Cellular distribution of glucose and monocarboxylate transporters in human brain white matter and multiple sclerosis lesions. Glia. 2014;62(7):1125–1141-1141.

    PubMed  Google Scholar 

  13. Wu GF, Alvarez E. The immunopathophysiology of multiple sclerosis. Neurologic Clinics. 2011;29(2):257–278-278.

    PubMed  PubMed Central  Google Scholar 

  14. Waxman SG, Craner MJ, Black JA. Na+ channel expression along axons in multiple sclerosis and its models. Trends in Pharmacological Sciences. 2004;25(11):584–591-591.

    CAS  PubMed  Google Scholar 

  15. Trapp BD, et al. Axonal transection in the lesions of multiple sclerosis. N Engl J Med. 1998;338(5):278–85.

    CAS  PubMed  Google Scholar 

  16. Kornek B, et al. Multiple sclerosis and chronic autoimmune encephalomyelitis: a comparative quantitative study of axonal injury in active, inactive, and remyelinated lesions. The American Journal of Pathology. 2000;157(1):267–276-276.

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Bakshi R, et al. MRI in multiple sclerosis: current status and future prospects. The Lancet Neurology. 2008;7(7):615–625-625.

    PubMed  PubMed Central  Google Scholar 

  18. Fancy S, et al. Myelin regeneration: a recapitulation of development? Annual Review of Neuroscience. 2011;34(1):21 –43-43.

    CAS  PubMed  Google Scholar 

  19. Lucchinetti C, et al. A quantitative analysis of oligodendrocytes in multiple sclerosis lesionsA study of 113 cases. Brain. 1999;122(12):2279–2295-2295.

    PubMed  Google Scholar 

  20. Popescu BF, et al. Pathology of multiple sclerosis: where do we stand?. Continuum (Minneapolis, Minn.) Lifelong Learning in Neurology. 2013. 19(4, Multiple Sclerosis):901–921. https://doi.org/10.1212/01.CON.0000433291.23091.65

    PubMed  Google Scholar 

  21. Bramow S, et al. Demyelination versus remyelination in progressive multiple sclerosis. Brain. 2010;133(10):2983–2998-2998.

    PubMed  Google Scholar 

  22. Stangel M, et al. Achievements and obstacles of remyelinating therapies in multiple sclerosis. Nature Reviews Neurology. 2017;13(12):742.

    PubMed  Google Scholar 

  23. Lundgaard I, et al. Neuregulin and BDNF induce a switch to NMDA receptor-dependent myelination by oligodendrocytes. PLoS Biology. 2013;11(12):e1001743.

    PubMed  PubMed Central  Google Scholar 

  24. Lee S, et al. A culture system to study oligodendrocyte myelination processes using engineered nanofibers. Nature Methods. 2012;9(9):917.

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Chong SY, Chan JR. Tapping into the glial reservoir: cells committed to remaining uncommitted. The Journal of cell biology. 2010;188(3):305–12-12.

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Gibson EM, et al. Neuronal activity promotes oligodendrogenesis and adaptive myelination in the mammalian brain. Science. 2014;344(6183):1252304.

    PubMed  PubMed Central  Google Scholar 

  27. Kotter MR, et al. Myelin impairs CNS remyelination by inhibiting oligodendrocyte precursor cell differentiation. The Journal of Neuroscience. 2006;26(1):328–332-332.

    CAS  PubMed  Google Scholar 

  28. Jensen SK, et al. Multimodal enhancement of remyelination by exercise with a pivotal role for oligodendroglial PGC1alpha. Cell Rep. 2018;24(12):3167–79.

    CAS  PubMed  Google Scholar 

  29. Setzu A, et al. Inflammation stimulates myelination by transplanted oligodendrocyte precursor cells. Glia. 2006;54(4):297–303-303.

    PubMed  Google Scholar 

  30. Vallée A, et al. Interactions between the canonical WNT/beta-catenin pathway and PPAR gamma on neuroinflammation, demyelination, and remyelination in multiple sclerosis. Cellular and Molecular Neurobiology. 2018;38(4):783–795-795.

    PubMed  Google Scholar 

  31. Fancy SP, et al. Dysregulation of the Wnt pathway inhibits timely myelination and remyelination in the mammalian CNS. Genes Dev. 2009;23(13):1571–85.

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Mi S, et al. LINGO-1 negatively regulates myelination by oligodendrocytes. Nature Neuroscience. 2005;8(6):745–751-751.

    CAS  PubMed  Google Scholar 

  33. Kuhlmann T, et al. Differentiation block of oligodendroglial progenitor cells as a cause for remyelination failure in chronic multiple sclerosis. Brain. 2008;131(7):1749–1758-1758.

    CAS  PubMed  Google Scholar 

  34. Chang A, et al. Premyelinating oligodendrocytes in chronic lesions of multiple sclerosis. N Engl J Med. 2002;346(3):165–73.

    PubMed  Google Scholar 

  35. Ciric B, et al. Human monoclonal IgM antibody promotes CNS myelin repair independent of Fc function. Brain Pathology. 2003;13(4):608–616-616.

    CAS  PubMed  Google Scholar 

  36. Huang JK, et al. Retinoid X receptor gamma signaling accelerates CNS remyelination. Nature Neuroscience. 2011;14(1):45.

    CAS  PubMed  Google Scholar 

  37. Buckley CE, et al. Drug reprofiling using zebrafish identifies novel compounds with potential pro-myelination effects. Neuropharmacology. 2010;59(3):149–59.

    CAS  PubMed  Google Scholar 

  38. Mei F, et al. Micropillar arrays as a high-throughput screening platform for therapeutics in multiple sclerosis. Nat Med. 2014;20(8):954–60.

    CAS  PubMed  PubMed Central  Google Scholar 

  39. •• Green AJ, et al. Clemastine fumarate as a remyelinating therapy for multiple sclerosis (ReBUILD): a randomised, controlled, double-blind, crossover trial. Clemastine fumarate as a remyelinating therapy for multiple sclerosis (ReBUILD): a randomised, controlled, double-blind, crossover trial, The Lancet 390.10111. 2017; 2481–2489. This randomized clinical trial of clemastine demonstrated an improvement in VEP after 3–5 months of therapy. This trial demonstrated the potential for remyelination in chronically demyelinated optic nerves.

  40. •• Schwartzbach CJ, et al. Lesion remyelinating activity of GSK239512 versus placebo in patients with relapsing-remitting multiple sclerosis: a randomised, single-blind, phase II study. J Neurol. 2017;264(2):304–15 This randomized clinical trial of GSK239512 did not obtain positive results, but did show feasibility for a multicenter trial using MTR as a primary endpoint.

    CAS  PubMed  Google Scholar 

  41. Hall SM. The effect of injections of lysophosphatidyl choline into white matter of the adult mouse spinal cord. J Cell Sci. 1972;10(2):535–46.

    CAS  PubMed  Google Scholar 

  42. Blakemore WF, Franklin RJ. Remyelination in experimental models of toxin-induced demyelination. Curr Top Microbiol Immunol. 2008;318:193–212.

    CAS  PubMed  Google Scholar 

  43. Woodruff RH, Franklin RJ. Demyelination and remyelination of the caudal cerebellar peduncle of adult rats following stereotaxic injections of lysolecithin, ethidium bromide, and complement/anti-galactocerebroside: a comparative study. Glia. 1999;25(3):216–28.

    CAS  PubMed  Google Scholar 

  44. Plemel JR, Liu WQ, Yong VW. Remyelination therapies: a new direction and challenge in multiple sclerosis. Nat Rev Drug Discov. 2017;16(9):617–34.

    CAS  PubMed  Google Scholar 

  45. Carlton WW. Studies on the induction of hydrocephalus and spongy degeneration by cuprizone feeding and attempts to antidote the toxicity. Life Sci. 1967;6(1):11–9.

    CAS  PubMed  Google Scholar 

  46. Mason JL, et al. Oligodendrocytes and progenitors become progressively depleted within chronically demyelinated lesions. Am J Pathol. 2004;164(5):1673–82.

    PubMed  PubMed Central  Google Scholar 

  47. Doan V, et al. Abbreviated exposure to cuprizone is sufficient to induce demyelination and oligodendrocyte loss. J Neurosci Res. 2013;91(3):363–73.

    CAS  PubMed  Google Scholar 

  48. Miljkovic D, Spasojevic I. Multiple sclerosis: molecular mechanisms and therapeutic opportunities. Antioxid Redox Signal. 2013;19(18):2286–334.

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Matsushima GK, Morell P. The neurotoxicant, cuprizone, as a model to study demyelination and remyelination in the central nervous system. Brain Pathol. 2001;11(1):107–16.

    CAS  PubMed  Google Scholar 

  50. Baxi EG, et al. Lineage tracing reveals dynamic changes in oligodendrocyte precursor cells following cuprizone-induced demyelination. Glia. 2017;65(12):2087–98.

    PubMed  PubMed Central  Google Scholar 

  51. Kipp M, et al. Multiple sclerosis animal models: a clinical and histopathological perspective. Brain Pathol. 2017;27(2):123–37.

    PubMed  Google Scholar 

  52. Gudi V, et al. Glial response during cuprizone-induced de- and remyelination in the CNS: lessons learned. Front Cell Neurosci. 2014;8:73.

    PubMed  PubMed Central  Google Scholar 

  53. Baker D, Amor S. Experimental autoimmune encephalomyelitis is a good model of multiple sclerosis if used wisely. Mult Scler Relat Disord. 2014;3(5):555–64.

    PubMed  Google Scholar 

  54. Munoz JJ, Bernard CC, Mackay IR. Elicitation of experimental allergic encephalomyelitis (EAE) in mice with the aid of pertussigen. Cell Immunol. 1984;83(1):92–100.

    CAS  PubMed  Google Scholar 

  55. Bjelobaba I, et al. Animal models of multiple sclerosis: focus on experimental autoimmune encephalomyelitis. J Neurosci Res. 2018;96(6):1021–42.

    CAS  PubMed  Google Scholar 

  56. van der Star BJ, et al. In vitro and in vivo models of multiple sclerosis. CNS Neurol Disord Drug Targets. 2012;11(5):570–88.

    PubMed  Google Scholar 

  57. Gumpel M, et al. Myelination and remyelination in the central nervous system by transplanted oligodendrocytes using the shiverer model. Discussion on the remyelinating cell population in adult mammals. Dev Neurosci. 1989;11(2):132–9.

    CAS  PubMed  Google Scholar 

  58. Frischer JM, et al. Clinical and pathological insights into the dynamic nature of the white matter multiple sclerosis plaque. Ann Neurol. 2015;78(5):710–21.

    PubMed  PubMed Central  Google Scholar 

  59. Shen S, et al. Age-dependent epigenetic control of differentiation inhibitors is critical for remyelination efficiency. Nat Neurosci. 2008;11(9):1024–34.

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Zhao C, Li WW, Franklin RJ. Differences in the early inflammatory responses to toxin-induced demyelination are associated with the age-related decline in CNS remyelination. Neurobiol Aging. 2006;27(9):1298–307.

    CAS  PubMed  Google Scholar 

  61. Patrikios P, et al. Remyelination is extensive in a subset of multiple sclerosis patients. Brain. 2006;129(Pt 12):3165–72.

    PubMed  Google Scholar 

  62. Patani R, et al. Remyelination can be extensive in multiple sclerosis despite a long disease course. Neuropathol Appl Neurobiol. 2007;33(3):277–87.

    CAS  PubMed  Google Scholar 

  63. Wolswijk G. Oligodendrocyte precursor cells in the demyelinated multiple sclerosis spinal cord. Brain. 2002;125(Pt 2):338–49.

    PubMed  Google Scholar 

  64. Fern R, Moller T. Rapid ischemic cell death in immature oligodendrocytes: a fatal glutamate release feedback loop. J Neurosci. 2000;20(1):34–42.

    CAS  PubMed  Google Scholar 

  65. Beck RW, Cleary PA. Optic neuritis treatment trial. One-year follow-up results. Arch Ophthalmol. 1993;111(6):773–5.

    CAS  PubMed  Google Scholar 

  66. Toosy AT, Mason DF, Miller DH. Optic neuritis. Lancet Neurol. 2014;13(1):83–99.

    CAS  PubMed  Google Scholar 

  67. Matsunaga Y, et al. Visual functional and histopathological correlation in experimental autoimmune optic neuritis. Invest Ophthalmol Vis Sci. 2012;53(11):6964–71.

    PubMed  Google Scholar 

  68. Frohman EM, Racke MK, Raine CS. Multiple sclerosis--the plaque and its pathogenesis. N Engl J Med. 2006;354(9):942–55.

    CAS  PubMed  Google Scholar 

  69. Silbermann E, Wooliscroft L, Bourdette D. Using the anterior visual system to assess neuroprotection and remyelination in multiple sclerosis trials. Curr Neurol Neurosci Rep. 2018;18(8):49.

    PubMed  Google Scholar 

  70. Klistorner A, Graham SL. Objective perimetry in glaucoma. Ophthalmology. 2000;107(12):2283–99.

    CAS  PubMed  Google Scholar 

  71. Klistorner A, et al. Correlation between full-field and multifocal VEPs in optic neuritis. Doc Ophthalmol. 2008;116(1):19–27.

    PubMed  Google Scholar 

  72. Brusa A, Jones SJ, Plant GT. Long-term remyelination after optic neuritis: A 2-year visual evoked potential and psychophysical serial study. Brain. 2001;124(Pt 3):468–79.

    CAS  PubMed  Google Scholar 

  73. Weinstock-Guttman B, et al. Pattern reversal visual evoked potentials as a measure of visual pathway pathology in multiple sclerosis. Mult Scler. 2003;9(5):529–34.

    CAS  PubMed  Google Scholar 

  74. Yeung HN, Aisen AM. Magnetization transfer contrast with periodic pulsed saturation. Radiology. 1992;183(1):209–14.

    CAS  PubMed  Google Scholar 

  75. Dousset V, et al. Experimental allergic encephalomyelitis and multiple sclerosis: lesion characterization with magnetization transfer imaging. Radiology. 1992;182(2):483–91.

    CAS  PubMed  Google Scholar 

  76. Schmierer K, et al. Magnetization transfer ratio and myelin in postmortem multiple sclerosis brain. Ann Neurol. 2004;56(3):407–15.

    PubMed  Google Scholar 

  77. Vavasour IM, et al. Is the magnetization transfer ratio a marker for myelin in multiple sclerosis? J Magn Reson Imaging. 2011;33(3):713–8.

    PubMed  Google Scholar 

  78. MacKay A, et al. In vivo visualization of myelin water in brain by magnetic resonance. Magn Reson Med. 1994;31(6):673–7.

    CAS  PubMed  Google Scholar 

  79. Laule C, et al. Myelin water imaging of multiple sclerosis at 7 T: correlations with histopathology. Neuroimage. 2008;40(4):1575–80.

    PubMed  Google Scholar 

  80. Gareau PJ, et al. Magnetization transfer and multicomponent T2 relaxation measurements with histopathologic correlation in an experimental model of MS. J Magn Reson Imaging. 2000;11(6):586–95.

    CAS  PubMed  Google Scholar 

  81. Le Bihan D, et al. Diffusion tensor imaging: concepts and applications. J Magn Reson Imaging. 2001;13(4):534–46.

    PubMed  Google Scholar 

  82. Song SK, et al. Demyelination increases radial diffusivity in corpus callosum of mouse brain. Neuroimage. 2005;26(1):132–40.

    PubMed  Google Scholar 

  83. Cross AH, Song SK. A new imaging modality to non-invasively assess multiple sclerosis pathology. J Neuroimmunol. 2017;304:81–5.

    CAS  PubMed  Google Scholar 

  84. Tran JQ, et al. Randomized phase I trials of the safety/tolerability of anti-LINGO-1 monoclonal antibody BIIB033. Neurol Neuroimmunol Neuroinflamm. 2014;1(2):e18.

    PubMed  PubMed Central  Google Scholar 

  85. Mi S, et al. LINGO-1 is a component of the Nogo-66 receptor/p75 signaling complex. Nat Neurosci. 2004;7(3):221–8.

    CAS  PubMed  Google Scholar 

  86. Mi S, Pepinsky RB, Cadavid D. Blocking LINGO-1 as a therapy to promote CNS repair: from concept to the clinic. CNS Drugs. 2013;27(7):493–503.

    CAS  PubMed  Google Scholar 

  87. Ranger A, et al. Anti-LINGO-1 has no detectable immunomodulatory effects in preclinical and phase 1 studies. Neurol Neuroimmunol Neuroinflamm. 2018;5(1):e417.

    PubMed  Google Scholar 

  88. Cadavid D, et al. Safety and efficacy of opicinumab in acute optic neuritis (RENEW): a randomised, placebo-controlled, phase 2 trial. Lancet Neurol. 2017;16(3):189–99.

    CAS  PubMed  Google Scholar 

  89. •• Klistorner A, et al. Assessment of opicinumab in acute optic neuritis using multifocal visual evoked potential. CNS Drugs. 2018;32(12):1159–71 This randomized clinical trial of opicinumab (an anti-LINGO antibody) had negative results but demonstrated feasibility of a multisite clinical trial using VEP as a primary outcome.

    PubMed  PubMed Central  Google Scholar 

  90. Bove RM, Green AJ. Remyelinating pharmacotherapies in multiple sclerosis. Neurotherapeutics. 2017;14(4):894–904.

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Deshmukh VA, et al. A regenerative approach to the treatment of multiple sclerosis. Nature. 2013;502(7471):327–32.

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Li Z, et al. Clemastine rescues behavioral changes and enhances remyelination in the cuprizone mouse model of demyelination. Neurosci Bull. 2015;31(5):617–25.

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Liu J, et al. Clemastine enhances myelination in the prefrontal cortex and rescues behavioral changes in socially isolated mice. J Neurosci. 2016;36(3):957–62.

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Green AJ, et al. Clemastine fumarate as a remyelinating therapy for multiple sclerosis (ReBUILD): a randomised, controlled, double-blind, crossover trial. Lancet. 2017;390(10111):2481–9.

    CAS  PubMed  Google Scholar 

  95. Ashworth S, et al. Unexpectedly high affinity of a novel histamine H(3) receptor antagonist, GSK239512, in vivo in human brain, determined using PET. Br J Pharmacol. 2014;171(5):1241–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Wilson DM, et al. Identification of clinical candidates from the benzazepine class of histamine H3 receptor antagonists. Bioorg Med Chem Lett. 2013;23(24):6890–6.

    CAS  PubMed  Google Scholar 

  97. Grove RA, et al. A randomized, double-blind, placebo-controlled, 16-week study of the H3 receptor antagonist, GSK239512 as a monotherapy in subjects with mild-to-moderate Alzheimer’s disease. Curr Alzheimer Res. 2014;11(1):47–58.

    CAS  PubMed  Google Scholar 

  98. Jarskog LF, et al. A phase II study of a histamine H(3) receptor antagonist GSK239512 for cognitive impairment in stable schizophrenia subjects on antipsychotic therapy. Schizophr Res. 2015;164(1–3):136–42.

    PubMed  Google Scholar 

  99. Chen Y, et al. Histamine receptor 3 negatively regulates oligodendrocyte differentiation and remyelination. PLoS One. 2017;12(12):e0189380.

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgments

Dr. Wooliscroft would like to thank the Veterans Administration MS Center of Excellence-West for their support in her fellowship. Dr. Silbermann would like to thank the National MS Society for their support of her fellowship through a Sylvia Lawry Award.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Lindsey Wooliscroft MD.

Ethics declarations

Conflict of Interest

Lindsey Wooliscroft and Elizabeth Silbermann each declare no potential conflicts of interest. Michelle Cameron reports consulting fees from Adamas Pharmaceuticals and Greenwich Biosciences outside the submitted work. Dennis Bourdette reports consultancy for reviewing patient medical records and providing opinion on treatments for Magellan Health Care and Best Doctors Inc. He also served as an expert witness on MS for the US Department of Justice, reports a bench research grant and collaborative center award from the National MS Society, and reports a founder’s stock valued at $1000 from Llama Therapeutics.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

This article is part of the Topical Collection on Multiple Sclerosis and Related Disorders

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wooliscroft, L., Silbermann, E., Cameron, M. et al. Approaches to Remyelination Therapies in Multiple Sclerosis. Curr Treat Options Neurol 21, 34 (2019). https://doi.org/10.1007/s11940-019-0574-1

Download citation

  • Published:

  • DOI: https://doi.org/10.1007/s11940-019-0574-1

Keywords

Navigation