You are on page 1of 18

REviEwS

Progressive multiple sclerosis:


from pathophysiology to therapeutic
strategies
Simon Faissner   1,2*, Jason R. Plemel   3, Ralf Gold1 and V. Wee Yong   2*
Abstract | Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system
that involves demyelination and axonal degeneration. Although substantial progress has been
made in drug development for relapsing–remitting MS, treatment of the progressive forms of
the disease, which are characterized clinically by the accumulation of disability in the absence
of relapses, remains unsatisfactory. This unmet clinical need is related to the complexity of the
pathophysiological mechanisms involved in MS progression. Chronic inflammation, which occurs
behind a closed blood–brain barrier with activation of microglia and continued involvement
of T cells and B cells, is a hallmark pathophysiological feature. Inflammation can enhance
mitochondrial damage in neurons, which, consequently, develop an energy deficit, further
reducing axonal health. The growth-inhibitory and inflammatory environment of lesions also
impairs remyelination, a repair process that might protect axons from degeneration. Moreover,
neurodegeneration is accelerated by the altered expression of ion channels on denuded axons.
In this Review, we discuss the current understanding of these disease mechanisms and
highlight emerging therapeutic strategies based on these insights, including those targeting the
neuroinflammatory and degenerative aspects as well as remyelination-promoting approaches.

Multiple sclerosis (MS) is a chronic inflammatory condi­ PPMS6 led to approval of the anti-CD20 antibody ocre­
tion of the central nervous system (CNS) that is charac­ lizumab as the first therapy in this setting. Moreover,
terized by demyelination with concomitant axonal and in March 2019, the US Food and Drug Administration
neuronal degeneration1. Approximately 85% of patients (FDA) approved the S1PR modulator siponimod for the
with MS present with a relapsing–remitting course of the treatment of SPMS in patients with active disease on
disease (RRMS), and the majority of these, as shown in the basis of findings from the phase III EXPAND study7.
natural history studies, advance to a progressive disease The reason why treatment of progressive MS remains
course — termed secondary progressive MS (SPMS) — elusive is multifaceted. A major explanation is that degen­
after 15–20 years of disease manifestation2. The remain­ erative mechanisms that characterize progressive MS
ing ~10–15% of patients have a slow and continuous are distinct from the largely inflammatory mechanisms
neurological deterioration without definable relapses, a that give rise to relapses in RRMS, and that the neuro­
1
Department of Neurology, type known as primary progressive MS (PPMS). Despite degenerative processes are not sufficiently targeted by the
St. Josef-Hospital,
tremendous successes in the development of therapies approved immunomodulatory compounds (for example,
Ruhr-University Bochum,
Bochum, Germany.
for RRMS3 and disease-modifying therapies that delay siponimod and ocrelizumab). In addition, uncertainty
2
Hotchkiss Brain Institute
the conversion to SPMS4, progressive MS has limited regarding the pathophysiological mechanisms involved
and Department of Clinical treatment options. Indeed, medications that are highly in progressive MS, an understanding that has been lim­
Neurosciences, University effective in RRMS have failed in the treatment of progres­ ited by a paucity of relevant preclinical animal models,
of Calgary, Calgary, Alberta, sive MS, as exemplified by the sphingosine-1-phosphate challenges the clinical translation of new treatments
Canada.
receptor (S1PR) modulator fingolimod, which did not (Box 1). Our current understanding is that progressive
3
Department of Medicine, reduce the risk of disability progression in patients MS is characterized by chronic inflammation behind a
University of Alberta,
Edmonton, Alberta, Canada.
with PPMS in the phase III INFORMS trial5. However, closed blood–brain barrier with activation of microglia
modest advances have been made in progressive MS and continued involvement of T cells and B cells. Release
*e-mail: simon.faissner@
rub.de; vyong@ucalgary.ca with two positive phase III clinical trials showing a of reactive oxygen species (ROS) and nitrogen species
https://doi.org/10.1038/ reduc­tion in confirmed disability progression of approxi­ (RNS) contributes to mitochondrial and axonal damage,
s41573-019-0035-2 mately 21–24%. Indeed, the phase III ORATORIO trial in which, ultimately, leads to neurodegeneration.

Nature Reviews | Drug Discovery


Reviews

Box 1 | controversies in progressive Ms pathophysiology within the CNS parenchyma, as few breaches seem to
exist in the blood–brain barrier 11,12. Infiltration of
Developing new therapeutic agents with precise modes of action requires an monocytes — which differentiate into macrophages
understanding of pathological mechanisms. However, many controversies exist — and local activation of microglia is seen in all types
regarding the pathophysiology of progressive multiple sclerosis (Ms). For example, of MS, but the macrophage/microglia representation
progressive Ms is characterized by neurodegeneration, yet whether early
is more pronounced in progressive MS, and these cells
neurodegeneration drives autoimmune injury, or whether ongoing inflammation
reaches a threshold to trigger neurodegeneration, is still unclear179. another unclear seem to drive the expansion of established lesions13.
point is whether neurodegeneration is independent of or dependent on chronic The normal-appearing white matter (NAWM) contains
inflammation. still, the notion that neurodegenerative and inflammatory injury diffuse microglial inflammation with few hypercellular
mechanisms interact to drive disability progression is widely agreed upon. without lesions and inflammatory perivascular cuffs14. In addition,
a clear understanding of the pathological mechanisms that drive progressive Ms, oxidative injury (including to mitochondria), which is
the in vivo modelling of the disease in animals is challenging. prominently contributed to by macrophages/microglia,
animal models that have been useful in the development of therapies for relapsing– is prevalent in progressive MS lesions12.
remitting Ms (rrMs), such as experimental autoimmune encephalomyelitis (eae), Given that progressive MS is characterized by a
have been largely unsuccessful in the identification of therapies that slow Ms mostly intact blood–brain barrier, medications for pro­
progression. Models that have been championed for progressive Ms, such as the
gressive MS that target pathophysiological features of
non-obese diabetic (NOD) mouse model or the Biozzi antibody high (aBH) mouse
model of eae (a proposed model of progression), have autoimmune injury and progressive MS should cross the blood–brain barrier,
secondary progression of disability, but these models assume that inflammation drives have the capacity to inhibit the activity of microglia,
progression, which is debatable. Non-autoimmune models, such as the mouse model reduce oxidative stress and retain the capacity to alter
of cuprizone-induced demyelination, have white matter degeneration, but the extent the activity of T lymphocytes and B lymphocytes (Fig. 1).
to which this approach models progressive Ms is unclear. a 2019 study showed that
intravenous injection of β-synuclein-reactive T cells induces grey matter demyelination Targeting T lymphocytes
and neurodegeneration in Lewis rats, which could be a new model to study cortical The goal of therapeutic modulation of T cell composi­
degeneration180. Nevertheless, preclinical models that mimic pathological aspects of tion and differentiation in progressive MS is generally to
progression as well as valid and reliable paraclinical markers are lacking. normalize inflammation and minimize any involvement
the value of testing and clinically translating new medications for progressive
of T cell infiltrates. T cells can be found in the spinal
MS developed in these preclinical models is unclear, as has been illustrated by
several negative trials, including those investigating fingolimod (INFORMS)5, cord parenchyma of patients with PPMS, but, unlike
natalizumab (asCeND)19 and rituximab (OLYMPus)45. the poor understanding of the those of patients with RRMS, these T cells are found
pathological mechanisms in progressive Ms makes it challenging to tailor targeted at lower levels and are more diffuse in the NAWM and
therapeutic agents. normal-appearing grey matter (NAGM)15. Surprisingly,
despite the low level of T cell-mediated inflammation
in progressive MS lesions, levels of inflammatory mark­
In this Review, we discuss the current understanding ers of T cell activation in cerebrospinal fluid (CSF) are
of the pathophysiology of progressive MS and highlight comparable between patients with progressive MS and
promising treatment approaches as well as auspicious those with RRMS16. In contrast to these low levels of
preclinical and clinical studies. Owing to the controv­ T cells in the parenchyma of patients with progressive
ersies regarding the pathophysiology of progressive MS, increased numbers of T cells are located within the
MS (Box 1), we address both aspects of inflammation meninges in patients with progressive MS. The extent
and degeneration in mediating progression. In addi­ of meningeal T cell inflammation is correlated with
tion to medications that target the inflammatory and axonal loss in the NAWM15, suggesting that the detri­
degenerative aspects, we also discuss medications that mental role of T cells in progressive MS is potentially
promote remyelination8, given that re-formed myelin mediated by long-range soluble factors. In progressive
might restore trophic support to denuded axons to slow MS forms, T cells slowly accumulate in spaces con­
Neuromyelitis optica axonal degeneration. Although the focus of this Review necting the lepto­meninges to the parenchyma, such as
spectrum disorder is on progressive MS, we also refer to important relevant Virchow–Robin spaces and meninges17. This finding
Inflammatory disorder of
data from other inflammatory and neurodegenerative might indicate that progressive MS is not characterized
the central nervous system
mediated by disease-specific conditions, such as neuromyelitis optica spectrum disorder, by a lack of T cell activation, but rather by exclusion of
antibodies against owing to similarities in their pathological mechanisms. T cells from the brain and spinal cord parenchyma. In
aquaporin-4, leading to As the study populations investigated in different trials addition, tissue-resident memory T cells, such as the few
severe immune-mediated differed considerably regarding their inclusion criteria that are found in the CNS parenchyma of patients with
demyelination and axonal
damage.
(for example, SPMS with or without relapse activity, or progressive MS, tend to have downregulated expression
PPMS) and changed over time, comparison of the results of S1PRs compared with central memory or effector
Normal-appearing white of different studies is difficult. memory T cells18, which is prob­ably why these cells are
matter retained in tissues and not targeted by S1PR modulators
(NAWM). An area in the white
Targeting neuroinflammation such as fingolimod or siponimod12.
matter without obvious lesions
or significant abnormalities, RRMS is characterized by immune cell-driven, Targeting T cells to slow disease progression remains
such as axonal damage, plaque-like demyelination with consequent neuro­ difficult, as demonstrated by the negative findings of
astrogliosis and microgliosis. degeneration9, whereas the invasion of T cells into the the phase III ASCEND trial, in which natalizumab, an
CNS parenchyma is markedly lower — albeit still present antibody directed against the α4 integrin subunit, failed
Normal-appearing grey
matter
— in progressive MS10. Moreover, in progressive MS, to reduce confirmed disability progression compared
(NAGM). An area in the grey inflammation is deemed to be compartmentalized at with placebo in patients with SPMS19. However, several
matter without obvious lesions. both the leptomeningeal and the blood vessel levels interesting approaches could effectively target T cells,

www.nature.com/nrd
Reviews

Mitochondrion-protective strategies

• Minocycline
Fe2+ • Chelating drugs
• Antioxidants

Brain

Anti-inflammatory strategies

• Riluzole
• Memantine
• Amantadine Siponimod
Astrocyte
Strategies targeting B and T lymphocytes
Glutamate
S1PR

Lactosylceramide

Inhibitor
ROS/
RNS Microglia

CD20 • Dimethyl fumarate


• Ocrelizumab • Minocycline
• Rituximab • Dextromethorphan
• Hydroxychloroquine
Meningeal
B cells

CD52
Remyelination strategies
GZ402668
Denuded axon

Lymphocyte
Amiloride Opicinumab LINGO1

Statins
ASIC1

MIF
Ibudilast

Macrophage Na+ channels

Lamotrigine • MD1003
HSC or MSC transplantation • Domperidone
• Clemastine

HSC MSC
Fig. 1 | Treatment approaches for progressive Ms. Mitochondrion- antihistamine clemastine (which targets muscarinic receptors). MD1003
protective strategies include the use of minocycline, iron (Fe2+)-chelating (biotin) is postulated to promote remyelination through its function as a
compounds and antioxidants that reduce oxidative stress. Anti- co-enzyme for carboxylases that might promote the synthesis of myelin
inflammatory strategies targeting microglia and astrocytes include lipids. Strategies targeting lymphocytes include the CD20-targeting
the S1PR modulator siponimod, which is licensed by the US Food and antibodies ocrelizumab (which is approved for primary progressive
Drug Administration as therapy for secondary progressive multiple multiple sclerosis (PPMS)) and rituximab. Other lymphocyte-targeting
sclerosis (SPMS), and inhibitors of microglial activity, including dimethyl approaches include the use of the anti-CD52 antibody GZ402668 and the
fumarate, minocycline, dextromethorphan and hydroxychloroquine. phospho­diesterase inhibitor ibudilast, which targets migration inhibitory
Axonoprotective approaches targeting ion channels include amiloride and factor (MIF). Stem cell transplantation, including haematopoietic stem cell
lamotrigine. Remyelination therapies are being tested with the leucine-rich (HSC) or mesenchymal stem cell (MSC) transplantation, is another method
repeat neuronal protein 1 (LINGO1)-targeted antibody opicinumab, of normalizing T cell populations. RNS, reactive nitrogen species; ROS,
domperidone (which elevates prolactin levels) and the first-generation reactive oxygen species.

Nature Reviews | Drug Discovery


Reviews

Table 1 | ongoing and completed clinical trials in progressive Ms


Treatment Trial details and status Patients Primary end points secondary end points
Neuroprotection and remyelination
Laquinimod Phase II (NCT02284568); completed PPMS PBVC • Time to CDP
October 2017 • Change from baseline in T25FW
• Number of new T2 brain lesions
Amiloride, fluoxetine, Phase IIb (MS-SMART; NCT01910259); SPMS MRI-derived PBVC • T2 lesions
riluzole active, not recruiting; final results • Pseudoatrophy
pending • Clinical measures of
neuroprotection
Oxcarbazepine Phase II (PROXIMUS; NCT02104661); Definite MS NFL levels in CSF • Safety
completed January 2018 • Clinical outcome (neurological
examination)
• Cognition
Domperidone Phase II (NCT02308137); recruiting; SPMS T25FW • 9HPT
planned study end date January 2020 • EDSS
• MFIS
• MS quality of life
Biotin (MD1003) Phase III (NCT02936037); active, not PPMS and • EDSS • 12-weeks confirmed EDSS
recruiting; planned study end date SPMS • T25FW progression
June 2023 • CGI-I score
Caprylic triglyceride Phase II (NCT01848327); completed RRMS, SPMS • Changes in learning • EDSS
December 2017 and PPMS • SDMT • BDI-II
• MSQOL-54
• MFIS
Liothyronine Phase Ib (NCT02506751); completed MS Adverse events –
September 2017
Idebenone Phase I/II (IPPoMS; NCT00950248); PPMS Inhibition of brain atrophy Inhibition of individualized rates
completed August 2018 (SIENA) of development of brain atrophy
Phase I/II (NCT01854359); active, not PPMS Inhibition of brain atrophy Inhibition of individualized rates
recruiting; planned study end date (SIENA) of development of brain atrophy
August 2019
Lipoic acid Phase II (NCT03161028); recruiting; Progressive T25FW • 2-minute timed walk
planned study end date April 2021 MS • Fall count
• Brain atrophy
B cell-targeted or T cell-targeted agents
Rituximab (intravenous Phase II (EFFRITE; NCT02545959); PPMS and Change in osteopontin level • TNF, neurofilament and IgG levels
and intrathecal), status unknown; planned study end SPMS in CSF in CSF
methylprednisolone date September 2019 • Brain volume atrophy (SIENA)
(intravenous)
GZ402668 Phase I (NCT02977533, Progressive Adverse events • Cmax
NCT02313285); completed, MS • AUC
open-label follow-up; planned study • Lymphocyte depletion
end date April 2022 • Anti-drug antibodies
• Injection site reactions
Simvastatin Phase II (MS-OPT; NCT03896217); not SPMS Effect on cerebral • MRI: glutamate levels and brain
yet recruiting; planned study end date blood flow atrophy
January 2021 • EDSS and functional scores
Stem cell therapy
Biological: human Phase I/II (NCT01364246); status PPMS and EDSS • VEP
umbilical cord unknown; planned study end date SPMS • SEP
mesenchymal stem cells December 2014 • BAEP
• Changes in MRI
HSC therapy Phase III (NCT00273364); active, not MS (not EDSS progression • Number of relapses
recruiting; planned study end date further • MSFC, PASAT and others
September 2024 specified)
Autologous Phase I/II (EMMES; NCT02495766); RRMS or Adverse events and safety • Cumulative number of
mesenchymal active, not recruiting; planned study SPMS profiles gadolinium-enhancing lesions
stromal cells end date June 2020 • MS outbreaks
(XCEL-MC-ALPHA) • EDSS
• Cumulative T2 lesions
Bone marrow Phase II (ACTiMuS; NCT01815632); PPMS and GEP • Safety
transplantation recruiting; planned study end date SPMS • EDSS
October 2019 • MSFC
• MRI brain and spinal cord
(lesion load and atrophy)
• OCT

www.nature.com/nrd
Reviews

Table 1 (cont.) | ongoing and completed clinical trials in progressive Ms


Treatment Trial details and status Patients Primary end points secondary end points
Stem cell therapy (cont.)
Autologous Phase II (NCT02166021); status RPMS and • Brain MRI • EDSS
mesenchymal bone unknown; planned study end date SPMS • Immunological response • Neurological function
marrow stem cells March 2018 • Electrophysiology
Tolerogenic dendritic Phase I (TOLERVIT-MS; RRMS and • Safety • ARR
cells NCT02903537); recruiting; planned SPMS with • EDSS score changes • EDSS
study end date September 2019 relapse • Number of new T2 lesions • 9HPT
activity and gadolinium-enhancing • T25FW
lesions
Interventions targeting inflammation
Masitinib Phase IIb/III (NCT01433497); active, PPMS and MSFC • EDSS
not recruiting; planned study end SPMS • MSQOL-54
date September 2020 • T25FW
• 9HPT
Methotrexate Early phase I (ITMTXPMS; – • Change in disability • Changes in brain MRI
(intrathecal) NCT02644044); status unknown; • Safety and tolerability measurements
planned study end date January 2018 • Laboratory measurements
Dimethyl fumarate Phase IV (SURV-SEP; NCT02901106); RRMS, SPMS • Relapse –
terminated October 2017 and PPMS • EDSS
Phase II (FUMAPMS; NCT02959658); PPMS NFL • EDSS
active, not recruiting; planned study • T25FW
end date December 2019 • 9HPT
• SDMT
Hydroxychloroquine Phase II (NCT02913157); PPMS T25FW • 9HPT
recruiting; planned study end date • EDSS
December 2020 • SDMT
• MFIS
Andrographolide Phase I/II (NCT02273635); status SPMS and Brain atrophy • EDSS
unknown; planned study end date PPMS • PASAT
April 2017 • MSIS29
• New T2 lesions
Ibudilast Phase II (NCT01982942); completed PPMS and • Mean rate of change in brain Diffusion tensor imaging,
December 2017 SPMS atrophy over 96 weeks magnetization transfer ratio,
• Safety measures retinal nerve fibre layer (OCT),
cortical atrophy
MIS416 Phase IIb (NCT02228213); completed SPMS Change from baseline of SAEs
June 2017 neuromuscular function at 12
months (MSFC and other tests)
Miscellaneous interventions
Oestrogen Phase II (NCT01466114); unknown RRMS, SPMS Changes in cognition (PASAT) • EDSS
status; planned study end date and PPMS • T25FW
December 2018
ACTH Phase II (NCT01950234); recruiting; SPMS, PPMS Proportion of patients • Safety and tolerability
planned study end date March 2020 and RPMS exhibiting a 20% worsening in • Slowed progression of cognitive
T25FW at 36 months disability
• Retinal nerve fibre layer thickness
NeuroVax (TCR Phase II/III (NCT02057159); unknown SPMS Cumulative number of new • Relapses
peptide vaccine) status; planned study end date gadolinium-enhancing lesions • EDSS
February 2019 on brain MRI • Immunological evaluation
• Safety
ABT-555 (antibody Phase I (NCT02601885); completed RRMS and • Adverse events • T25FW
against RGMa) April 2018 SPMS • Pharmacokinetics and • 9HPT
pharmacodynamics • Quality of life
• T1 gadolinium-enhancing lesions
Methylprednisolone Phase I (MSECP; NCT02296346); SPMS Accumulation of disability Changes in immunological
via extracorporeal recruitment suspended; planned (composite of components of parameters
photopheresis study end date October 2017 MSFC score)
Insulin (intranasal) Phase I/II (NCT02988401); recruiting; RRMS, SPMS Change in SDMT • Safety
planned study end date June 2020 and PPMS • Cognition
9HPT, 9-Hole Peg Test; ACTH, adrenocorticotropic hormone; ARR, annualized relapse rate; AUC, area under the curve; BAEP, brainstem auditory evoked potential; BDI-II,
Beck depression inventory 2nd edition; CDP, confirmed disability progression; CGI, Clinical Global Impression rating scales; CSF, cerebrospinal fluid; EDSS, Expanded
Disability Status Scale; GEP, global evoked potential; HSC, haematopoietic stem cell; MFIS, Modified Fatigue Impact Scale; MSFC, Multiple Sclerosis Functional
Composite; MSIS29, Multiple Sclerosis Impact Scale; MSQOL-54, Multiple Sclerosis Quality Of Life-54; NFL, neurofilament light chain; OCT, optical coherence tomography ;
PASAT, Paced Auditory Serial Addition Test; PBVC, percentage brain volume change; PPMS, primary progressive multiple sclerosis; RGMa, repulsive guidance molecule a;
RRMS, relapsing–remitting multiple sclerosis; SAE, serious adverse event; SDMT, Symbol Digit Modalities Test; SEP, somatosensory evoked potential; SIENA, structural
image evaluation using normalization, of atrophy ; SPMS, secondary progressive multiple sclerosis; T25FW, Timed 25-Foot Walk; VEP, visual evoked potential.

Nature Reviews | Drug Discovery


Reviews

a number of which are under investigation in ongoing or of mesenchymal stem cells (MSCs) in EAE models
completed trials (Fig. 1; Table 1). improved clinical scores, and the improvement was cor­
related with the generation of classical regulatory T (Treg)
Statins. One therapeutic option to modulate T cell activ­ cells in an IL-17A-dependent manner27.
ity in progressive MS is the use of statins. Simvastatin has Regarding autologous bone marrow transplanta­
anti-inflammatory effects and can induce a shift from tion, one study in patients with PPMS showed that this
a pro-inflammatory T helper type 1 (TH1) phenotype treatment is relatively safe and a small phase I trial in six
to a less-damaging, anti-inflammatory TH2 phenotype20. patients with progressive–relapsing MS (an old classifica­
Simvastatin reduces the number of T H1 cells and tion to describe patients who are relapsing but also pro­
TH17 cells (the latter of which are particularly pro- gressing) found that treatment normalized or improved
inflammatory), improves clinical scores in the inflam­ EDSS scores over 1 year28. In a phase IIa proof-of-
matory experimental autoimmune encephalitis (EAE) concept study, autologous bone marrow transplantation
model of MS21, and inhibits the in vitro TH17 differen­ improved visual acuity and visual evoked potentials
tiation of CD4+ T cells isolated from patients with MS22. and increased optic nerve area in patients with SPMS
In addition, statins induce remyelination in the spinal who had visual impairment29. A phase III randomized
cord of EAE animals23. controlled trial evaluating bone marrow-derived cellular
In one Cochrane review that included four trials therapy in patients with progressive MS (ACTiMuS) has
including 458 patients, the combination of simvastatin been underway since March 2014 (ref.30) (Table 1).
or atorvastatin with interferon-β (IFNβ) did not reduce In addition, delivery of autologous haematopoietic
relapses, disease progression or the development of stem cells (HSCs) reduced T2 lesions in patients with
gadolinium-enhancing lesions 24. In the randomized, SPMS and RRMS, but did not slow progression of disa­
double-blind, placebo-controlled phase II MS-STAT bility31. This approach is currently being investigated in a
trial, simvastatin (80 mg daily) reduced the number of phase III clinical trial (NCT00273364; Table 1). In 2019,
new and enlarging T2 lesions (2.19 versus 1.50 events Burt and colleagues32 reported the efficacy of nonmyelo­
per person-year; P = 0.176) and the rate of brain atrophy ablative HSC transplantation (HSCT) compared with
(0.288% versus 0.584% per year; P = 0.003) compared continued disease-modifying therapy (DMT) in patients
with placebo in patients with SPMS25. In a sub-analysis with RRMS. While patients who received nonmyelo­
of MS-STAT trial data, patients on treatment experi­ ablative HSCT had a decrease in mean EDSS score after
enced improvements in frontal lobe function, as meas­ 1 year (from 3.38 to 2.36) and only three patients had
ured using the Frontal Assessment Battery score and progressed after 1 year, DMT-treated patients had a
the mean physical component score, a subscale of the higher mean EDSS score after 1 year (from 3.31 to 3.98)
self-reported 36-item Short Form Survey (SF-36)26. and 34 patients had disability progression (median time
The contradictory results of the MS-STAT trial (positive) to progression 24 months). Muraro et al.33 retrospec­
and the Cochrane meta-analysis (negative) could be tively evaluated long-term outcomes in 281 patients from
because of the use of different outcome parameters: 25 centres after autologous HSCT following treatment
the MS-STAT study used brain atrophy, which is pre­ with a low-intensity regimen (cyclophosphamide alone or
sumably more valid in patients with progressive MS. in combination with anti-thymocyte globulin or fludara­
Thus, statin therapy might both induce neuroprotection bine) or a high-intensity regimen (busulfan or total-body
and target T cell-mediated inflammation. irradiation). Of the 281 patients, 78% had progressive
To further address potential effects on progression, a forms of MS, and, interestingly, older patients and patients
group at University College London announced the phase with progressive disease were more likely to progress rel­
II MS-OPT trial in patients with SPMS (NCT03896217), ative to those with RRMS (HR 1.03, 95% CI 1.00–1.05,
which started recruitment in March 2019 (Table  1). and HR 2.33, 95% CI 1.27–4.28, respectively)33, rais­
This double-blind, randomized, placebo-controlled ing the concern that this therapeutic approach is only
single-site study will evaluate the effects of simvasta­ applicable to younger patients with early stage disease
tin (80 mg daily) on cerebral blood flow (the primary and presumably has limited efficacy in patients with
Experimental autoimmune
encephalitis end point), but also on brain atrophy and Expanded progression. Another approach under investigation in
(EAE). An inflammatory animal Disability Status Scale (EDSS) and functional scores ongoing trials is the use of MSCs derived either from the
model of multiple sclerosis, (secondary end points). However, given that the utility umbilical cord (phase I and II; NCT01364246) or from
mediated by inoculation of of cerebral blood flow as a good marker of neuroprotec­ adipose tissue (phase I; NCT02326935; Table 1).
myelin components with
adjuvants.
tion is highly speculative, the results of the MS-OPT trial Thus, increasing evidence suggests that SCT may be
could be challenging to interpret. able to reduce MS progression, but long-term data are
Gadolinium-enhancing needed to better evaluate whether such reprogramming
lesions Stem cell transplantation. Another means to normalize of the immune system is a good therapeutic approach
T1 lesions showing
the T cell populations in patients with progressive MS is for progressive MS. Moreover, differing protocols and
contrast agent (gadolinium)
enhancement on magnetic via stem cell transplantation (SCT). A number of studies methods exist (for example, nonmyeloablative versus
resonance imaging. evaluating stem cell therapy for progressive MS are ongo­ myeloablative SCT, and transplantation of different
ing or have been completed (Table 1). Of note, the stud­ cell types), the relative efficacy of which remains to be
T2 lesions ies discussed herein were performed using different SCT established. Future investigations of the effects of SCT on
Hyperintense magnetic
resonance imaging sequences,
protocols, which vary in their mechanisms of action, compartmentalized meningeal inflammation associated
indicating multiple sclerosis efficacy and adverse effect profiles. Preclinical studies with neurodegeneration, which has not been done to the
lesion load. support this approach; for instance, transplantation best of our knowledge, might also be interesting.

www.nature.com/nrd
Reviews

Targeting B lymphocytes long-lived plasma cells are not targeted by rituximab


B cells have multiple roles and contribute to progres­ because these cells downregulate CD20. Whether the
sion in MS34 via antibody production, antigen presenta­ beneficial depletion of peripheral B cells or B cell clones
tion to T cells35 and the secretion of pro-inflammatory with rituximab in the stated OLYMPUS subgroups
cytokines36. Studies of brains from individuals who died occurred in the meninges or the brain is unclear, because
with progressive MS have demonstrated increased cellu­ only a low amount of rituximab (0.1%) passes the
lar inflammatory aggregates, particularly in the menin­ blood–brain barrier after intravenous administration47.
ges, some of which are enriched with B cells and have Intrathecal administration of rituximab might
been referred to as ectopic follicles37. The importance of improve the targeting of B cells in the brain and menin­
this meningeal B cell accumulation is demonstrated by ges. Indeed, after intrathecal injection of even very low
reports of subpial grey matter demyelination and micro­ doses of rituximab (two injections of 3.5 mg), exten­
glial activity in cortical structures38–40. Meningeal B cell sive peripheral B cell depletion occurred in patients
accumulation seems to be clinically relevant as patients with progressive MS48. Of note, combined peripheral
with B cell follicular structures have a faster progression and intrathecal administration of rituximab is cur­
of disability, an earlier disease onset and a younger age at rently under investigation (NCT02545959; Table 1).
death than those without such structures38. In progres­ In the RIVITaLISe study, combined intravenous and
sive MS, the presence of a subset of B cells (DC-SIGN+ intrathecal administration of rituximab in patients with
B cells) and T helper cells (ICOS+ TFH– cells) correlates SPMS, led to a robust depletion of B cells in the CSF49.
with disease progression, suggesting the presence of However, levels of neurofilament light chain and other
ongoing peripheral inflammation41. Furthermore, clo­ biomarkers were unchanged, and therefore the study was
nally expanded plasma cells from patients with MS pro­ stopped. The authors hypothesized that the study might
duce antibodies directed against neurons and astrocytes, have been underpowered to detect treatment effects,
but rarely against myelin; however, these antibodies lead especially regarding the biomarkers investigated.
to demyelination in murine spinal cord slice cultures, In a retrospective propensity-matched cohort
stressing the pathological role of clonally expanding study in 44 matched pairs of patients with SPMS,
B cells in progressive MS42. rituximab-treated patients had a significantly lower
On the contrary, new evidence shows that the num­ mean EDSS score (mean difference –0.52, 95% CI –0.79
ber of IgA+ plasma cells is reduced in the gut during EAE, to –0.26; P < 0.001) and a delay in time to confirmed
and that over-abundance of IgA+ plasmablasts or plasma disability progression (HR 0.49, 95% CI 0.26–0.93;
cells leads to resistance against EAE development43. P = 0.03) than patients never treated with rituximab50.
These observations are in line with the findings of a Positive findings of the placebo-controlled ORATORIO
clinical study in patients with RRMS, in which weekly trial provide evidence that B cell depletion using the
treatment with atacicept — a soluble fusion protein CD20-targeting antibody ocrelizumab might indeed be
containing a fragment of transmembrane activator and efficacious in patients with PPMS6. The study included
CAML interactor (TACI; a TNF receptor superfamily 732 patients with PPMS and showed a 24% reduced
member) that binds to B lymphocyte stimulator (BLYS; risk of disability progression 12 weeks after treatment
also known as BAFF) and a proliferation-inducing with 600 mg ocrelizumab and a 17.5% reduced rate of
ligand (APRIL) — led to deterioration with a higher brain atrophy. These findings were accompanied by less
annualized relapse rate in the atacicept groups (25 mg, worsening of the Timed 25-Foot Walk (T25FW) after
0.86, 95% CI 0.43–1.74; 75 mg, 0.79, 95% CI 0.40–1.58; 120 weeks in the ocrelizumab group than in the placebo
and 150 mg, 0.98, 95% CI 0.52–1.81) than in the placebo group (38.9% versus 55.1%; P = 0.04). In March 2017,
group (0.38, 95% CI 0.17–0.87)44. These data show that ocrelizumab received market authorization from the
the role and type of plasma cells or different lineage pre­ FDA and European Medicines Agency (EMA) and is,
cursor stages of B cells in different organs can be either therefore, the first medication approved for PPMS — a
beneficial or deleterious, speaking to the complexities milestone in MS therapy.
of potential therapeutic approaches. Thus, B cell deple­
tion has been considered a promising approach to the Anti-CD52 antibodies. Another approach is to simultane­
treatment of progressive MS and is under current clinical ously target T and B lymphocytes using next-generation
investigation (Fig. 1; Table 1). antibodies directed against CD52 (which is expressed on
both T and B cells), with the goal of reducing immuno­
Anti-CD20 antibodies. Although in the OLYMPUS genicity and infusion-related events compared with
study intravenous infusion of the CD20-specific anti­ the currently used CD52-targeting antibody alemtu­
body rituximab failed to improve the primary end point zumab (which is approved for the treatment of highly
of confirmed disability progression compared with pla­ active RRMS). The next-generation anti-CD52 anti­
cebo in patients with progressive MS, subgroup analyses body GZ402668 binds to different CD52 epitopes than
showed partial efficacy in patients who were younger those targeted by alemtuzumab, and it reduces the
than 51 years and/or had gadolinium-enhancing levels of tumour necrosis factor (TNF) and IFNγ, and
lesions45. However, B cell depletion with rituximab does dose-dependently decreases lymphocyte numbers in vivo
not change oligoclonal bands46, implying that meningeal in CD52-humanized mice51. Currently, the antibody is
B cell clones, for example, are not completely depleted, or under investigation in a phase I trial in patients with pro­
that the disappearance of oligoclonal bands has limited gressive MS and in an open-label, long-term follow-up
diagnostic value in predicting progression. Moreover, study (NCT02977533 and NCT02313285; Table 1).

Nature Reviews | Drug Discovery


Reviews

Targeting myeloid cells under investigation in a phase II trial in patients with


Microglia have a crucial role in the homeostasis of the PPMS (NCT02913157; Table 1).
brain but also have an important role in the progression The compound MIS416, which targets myeloid cells
of MS52. Activated microglia and monocyte-derived via activation of the cytosolic receptors nucleotide-
macrophages are often indistinguishable from one binding oligomerization domain-containing protein 2
another in pathology studies, and are often collec­ (NOD2) and Toll-like receptor 9 (TLR9), reduced dis­
tively termed ‘macrophages/microglia’. Microglia and ease severity in a mouse EAE model of MS61 and is
macrophages are generally assumed to differ in their currently being evaluated in a phase IIb trial in SPMS
phenotype and function, but, because of the histori­ (NCT02228213).
cal challenges in differentiating these cells, their rela­ Another approach uses the phosphodiesterase inhibi­
tive contribution to MS is unclear. Nonetheless, these tor ibudilast, which targets macrophage migration
cells are prominent producers of ROS and RNS and inhibitory factor (MIF)62 and might, therefore, affect
pro-inflammatory cytokines and chemokines. trafficking of macrophages into the CNS. Ibudilast has
In patients with MS, activation of macrophages/ been investigated in a phase II trial, which included a
microglia occurs in the NAWM and NAGM, corre­ total of 255 patients with PPMS or SPMS63,64; the primary
lating with axonal injury14. In mouse models, release end point was the rate of change in the brain parenchy­
of ROS or RNS from macrophages can initiate focal mal fraction. Over 96 weeks, patients in the ibudilast arm
axonal degeneration, even if myelin sheaths are intact53. (n = 129) experienced ~2.5 ml less brain tissue loss than
In EAE, NADPH oxidase activity in microglia and those in the placebo arm (n = 126)64. However, whether
astrocytes has been shown to remain high after peak this effect is clinically relevant for disability or cognitive
disease into the chronic setting, suggesting the ongo­ function remains to be investigated in future trials.
ing production of ROS54; elevated NADPH oxidase
activity is associated with increased neuronal calcium Antioxidants. Another therapeutic approach for pro­
(Ca2+) levels, which might indicate impaired neuronal gressive MS is to target the release of ROS and RNS from
function, and is also associated with ROS production myeloid cells. Inhibiting the ROS-generating enzyme
by macrophages/microglia. Moreover, ageing-related NADPH oxidase 2 (NOX2) with low-dose dextromethor­
phenotypic changes have been reported in macrophages/ phan treatment led to an improved disease course in the
microglia. Indeed, microglia from aged mice are EAE mouse model65. Importantly, NOX2 is expressed
more prone to generation of neurotoxins than those at high concentrations in microglia and macrophages,
from neonatal mice55 and, in post mortem analyses of and its inhibition with dextromethorphan can increase
patients with MS, microglia displayed ageing-related the proliferation of oligodendroglial precursor cells
morphological changes with less ramified morpho­ fourfold66 and attenuate microglial activity, leading to
logy56. This finding links the observations that most decreased production of nitric oxide (NO), superoxide
patients with RRMS develop a progressive phenotype free radicals and TNF67.
only 15–20 years after disease onset and that progres­ An agent with auspicious preclinical and clinical
sion of disability occurs at a similar age in both SPMS data is lipoic acid, an endogenously produced antioxi­
and PPMS. dant with numerous physiological roles. Lipoic acid was
In addition to ROS, macrophages/microglia are major investigated in a small study in patients with SPMS68.
sources of pro-inflammatory cytokines and chemokines. After 2 years, the annualized percentage change in brain
These cells can adopt many polarized states, including volume was significantly lower in the lipoic acid arm
pro-inflammatory or regulatory, anti-inflammatory (n = 27; 1,200 mg lipoic acid daily) than in the placebo
phenotypes; however, the categorization into M1 and arm (n = 24; –0.21 versus –0.65; P = 0.002). Currently, this
M2 phenotypes is oversimplified57. In the Theiler’s murine approach is being investigated in a phase II multi-centre
encephalomyelitis virus infection model of MS, the early clinical trial (NCT03161028; Table 1).
phase is dominated by pro-inflammatory macrophages/ A medication with pleiotropic mechanisms of action,
microglia, whereas the chronic–progressive phase is including antioxidant activity, is dimethyl fumarate
characterized by regulatory and/or anti-inflammatory (DMF). Indeed, DMF has cytoprotective effects in
polarization with sustained pro-inflammatory forms58. immune cells, glia and neurons (for reviews see refs69,70).
Thus, normalization of macrophage/microglia func­ DMF modifies the Kelch-like ECH-associated protein 1
tion is a promising therapeutic approach, given that a (KEAP1)–nuclear factor erythroid 2-related factor 2
switch towards a regulatory, anti-inflammatory pheno­ (NFE2L2) complex, thereby reducing oxidative stress
type promoted oligodendrocyte differentiation and led and promoting cytoprotection71. Owing to these effects,
to enhanced remyelination in the lysolecithin mouse the use of DMF for the treatment of PPMS has been
model59. Indeed, a number of therapeutic approaches investigated. A small observational study in 26 patients
have been investigated (Fig. 1; Table 1). treated with either fumaderm (a mixture of fumaric acid
esters; n = 18) or DMF (n = 8) showed that 75% of the
Inhibitors of macrophage/microglia activity. Hydroxy­ patients either remained stable or had a slightly reduced
chloroquine, an antimalarial medication that is also EDSS score (that is, improvement in disability), whereas
used in rheumatoid arthritis, has been shown to no differences were found between the two pharma­
inhibit the lipopolysaccharide-induced production of ceutical forms72. Since 2016, the phase II FUMAPMS
pro-inflammatory cytokines by microglia concomitant study has investigated use of DMF in the treatment of
with attenuation of EAE in mice60. This agent is currently PPMS (Table 1).

www.nature.com/nrd
Reviews

Targeting cytokines. In addition to the production of microglial cytokine production84 — support the rationale
ROS, pro-inflammatory cytokine secretion remains for further evaluation of minocycline in progressive MS.
prevalent during progressive MS. Infiltrating macro­
phages contribute to progressive MS through sustained Targeting astrocytes
production of TNF, as shown in a mouse model of pro­ Evidence from chronic EAE studies supports the role
gressive EAE, in which treatment with an anti-TNF of astrocytes in promoting chronic inflammation.
agent reduced myelin loss and leukocyte infiltration73. Astrocytes are the most abundant cell type in the CNS.
However, demyelinating neurological disease was They have a crucial role in neural development and
reported after treatment with an anti-TNF agent in function, and contribute to MS pathophysiology due to
patients with rheumatological disease74, and a study scar formation and release of inflammatory mediators90.
in 168 patients, most with RRMS, showed increased Activated ‘A1’ astrocytes can be induced by microglial
and earlier exacerbations of MS in patients receiving secretion of IL-1α, TNF and complement C1q. These A1
the TNF-blocking agent lenercept compared with those astrocytes lose the ability to promote neuronal survival,
receiving placebo75. However, the pathology underlying outgrowth and synaptogenesis, and, instead, induce
these observations remains elusive. TNF production the death of neurons and oligodendrocytes91. Another
by microglia is enhanced by galectin-9, which is pro­ study found that microglia-derived vascular endothe­
duced by astrocytes76. Galectin-9 is elevated in the CSF lial growth factor (VEGF) triggers pro-inflammatory
of patients with SPMS and correlates with T1 lesion load, signalling in astrocytes and worsens EAE, whereas
but not with gadolinium-enhancing lesions, IgG index microglia-derived transforming growth factor-α
or CSF cell count, supporting the concept of sustained (TGF-α) suppresses the pathophysiological activity of
compartmentalized inflammation by resident innate astrocytes92. In an EAE model of MS, the genetic dele­
immune cells in progressive MS77. tion of astrocytes in the acute phase of disease worsened
Another pro-inflammatory cytokine involved in pro­ disease severity, but their depletion during the chronic
gressive MS is IL-6, the production of which is sustained progressive phase ameliorated disease activity93. Thus,
during the chronic phase of EAE in astrocytes and the astrocytes with pro-inflammatory and neurotoxic
blockade of which results in decreased disability, myelin activities are thought to contribute to pathology in the
loss and axonal damage, as well as reduced activation progressive phase of MS.
of microglia in a progressive EAE model78. Despite the In astrocytes, the glycosphingolipid lactosylceramide
evidence of successful therapy using the IL-6-blocking — which is synthesized by the enzyme β-1,4-galacto­
antibody tocilizumab in neuromyelitis optica79,80, there is syltransferase 6 (B4GALT6) — functions in an auto­
only limited experience in MS81. Alarmingly, one report crine manner to activate the nuclear factor-κB (NF-κB)
described the development of MS in a patient with rheu­ and interferon regulatory factor 1 (IRF1) pathways,
matoid arthritis receiving tocilizumab therapy, although leading to the production and release of C-C motif
this patient had prior treatment with TNF-blocking chemokine 2 (CCL2) and granulocyte–macrophage
therapy, which is known to induce demyelination82. colony-stimulating factor (GMCSF), which subsequently
Thus, further studies are warranted to investigate activate microglia and monocytes93. Inhibition of lacto­
whether IL-6-targeted therapy is effective in reducing sylceramide synthesis markedly improved behavioural
progression in MS. outcomes in a chronic progressive EAE mouse model,
Another medication that has potential for use in the and was associated with reduced neurodegeneration93.
treatment of progressive MS is minocycline, a widely Another way to block astrocyte activity is through
used tetracycline with a good safety profile. This agent therapeutic modulation of the S1PRs, which induce
inhibited the activity of matrix metalloproteinases astrocyte proliferation upon activation by sphingosine-1-
(MMPs), decreased MMP-9 production and led to an phosphate (S1P)94. Indeed, down-modulation of S1PR1
improved disease course in a mouse model of EAE83. In (also known as S1P1) leads to reduced astrogliosis95, and
addition, minocycline has been shown to reduce micro­ treatment of astrocytes with S1P inhibits gap junctions
glial TNF production during T cell–microglia interaction between astrocytes via activation of both Gi and Rho
by downregulation of CD40 ligand (CD40L) expression GTPases96. Treatment of EAE mice with fingolimod, a
on T cells84. Minocycline treatment exerted positive modulator of S1PR1–5 (also known as S1P1–5), during
effects in both acute and chronic EAE animal models85,86. the chronic phase reduced astrogliosis, demyelination
However, the findings of studies in humans have partially and axonal loss, and improved EAE97. The first phase III
contradicted the preclinical findings. While minocycline trial evaluating fingolimod in patients with progressive
has been reported to have positive effects in RRMS87 and MS failed to meet its primary end point5. However, the
to reduce the risk of developing definite MS in patients follow-up substance siponimod, another S1PR modu­
with clinically isolated syndrome (CIS)88, this decreased lator that has greater specificity for S1PR1 and S1PR5,
Clinically isolated syndrome
risk of conversion to MS in CIS is lost after 24 months88. showed efficacy in the phase III EXPAND study. In
(CIS). The first clinical episode
of neurological symptoms Furthermore, in the RECYCLINE study, the addition of patients with SPMS, siponimod reduced 3-month con­
lasting at least 24 h, with minocycline to IFNβ1a in patients with RRMS did not firmed disability progression by 21% relative to pla­
features that are indicative show any additional effect regarding time to first qual­ cebo7. Moreover, siponimod reduced brain atrophy by
of multiple sclerosis. ifying relapse beyond that of IFNβ1a plus placebo89. 15% (adjusted mean over months 12 and 24), suggest­
Astrogliosis
Although the minocycline CIS trial (at 24 months) and ing its neuroprotective effects7. In October 2018, both
An increase in the number of the RECYCLINE trial were underpowered, the proposed the FDA and EMA accepted the New Drug Application
astrocytes due to damage. mechanisms of action — which include the inhibition of and Marketing Authorization Application for the use

Nature Reviews | Drug Discovery


Reviews

Periplaque white matter


of siponimod in the treatment of SPMS. In addition, in lesion edges, which suggests iron export, and are down­
The area around lesions in the March 2019, the FDA approved the use of siponimod regulated in the NAWM. Similarly, chronic MS lesions
white matter. in the treatment of SPMS in patients with active disease have reduced iron levels101. In active MS lesions, iron is
on the basis of these findings. released from dying oligodendrocytes and then taken
Iron chelation
Binding of iron by a chelating
up by macrophages and microglia101, but whether this
agent. Promoting neuroprotection removal of iron by these phagocytes is responsible for the
Conferring neuroprotection is a key goal in reducing reduction in iron content in the chronic lesions and/or
axonal degeneration and neuronal loss in MS. This the NAWM of patients with chronic MS, or whether it
approach involves interfering with mediators of injury is the result of a decreased number of iron-containing
in the progressive phase of MS, which includes the accu­ oligodendrocytes, is unclear98. The complexities of iron
mulation of iron, the altered expression of ion channels in MS are discussed in Box 2.
and mitochondrial damage (Fig. 1). Decreased iron content could potentially impair the
function of iron-requiring enzymes such as glucose-6-
Targeting iron phosphate dehydrogenase, mitochondrial respiratory
Iron accumulates in an age-dependent manner in the chain enzymes or enzymes involved in the production
CNS and has been proposed as an important contrib­ of lipids, which might decrease mitochondrial func­
utor to the pathogenesis of progressive MS98. Indeed, tion and reduce remyelination98. By contrast, increased
iron deposition in the deep NAGM correlates with uptake of iron by macrophages/microglia is associated
MS progression 99. This deposition might amplify with impaired function of these cells and increased toxi­
ongoing inflammation, exacerbate mitochondrial dys­ city102. One reason for the increased uptake of iron by
function through oxidative stress, potentiate axonal macrophages/microglia could be an imbalance between
damage and impair CNS homeostasis via the release the expression of iron-importing and iron-exporting
of pro-inflammatory cytokines100. Under normal phys­ enzymes. Evidence from EAE shows that the efflux
iological conditions within the CNS, iron is stored transporter ferroportin-1 (also known as SLC40A1)
mostly in oligodendrocytes. Iron accumulates in the is internalized in macrophages/microglia, leading to
CNS in patients with MS for different reasons, such increased intracellular levels of iron103. In addition,
as the breakdown of iron-containing oligodendrocytes, a study using 7-T MRI in patients with MS showed that
the infiltration of immune cells and the dysregulation of slowly expanding lesions and some inactive lesions
iron-transport proteins (reviewed by Stephenson et al.98). have iron rims and that iron is mainly found within
However, the role of iron in MS progression is contro­ pro-inflammatory macrophages/microglia 13. More
versial. Although iron accumulation can contribute to importantly, these iron-containing lesions expanded
neuro­toxicity and propagation of inflammation, evidence to a greater degree over 3.5 years than lesions with­
also suggests that iron deficiency has deleterious effects. out iron rims13. Together, these data indicate that tar­
Indeed, compared with healthy individuals, patients with geting the maldistribution and processing of iron is a
chronic MS have decreased iron levels in the NAWM, promising approach to the treatment of progressive
which corresponds with an increased disease dura­ MS (Fig. 1).
tion101. Concurrently, iron-exporting ferroxidases are One such approach is the combination of mino­
upregulated in the periplaque white matter close to active cycline with hydroxychloroquine, as both reduce the
neurotoxicity of iron in vitro and both also target other
features relevant to progression, such as mitochondrial
Box 2 | competing roles of iron in progressive Ms damage and T cell and/or B cell activation. This com­
bination has been shown to improve disease pheno­
In progressive multiple sclerosis (MS), competing roles of iron in mediating
types in the chronic EAE mouse model and the Biozzi
neurotoxicity and neuronal restoration have been reported.
antibody high (ABH) progressive MS mouse model104.
Toxic effects Importantly, other tetracyclines do not protect neurons
Iron can elicit neurotoxic effects through the generation of reactive oxygen species (ROS) from iron-mediated neurotoxicity for reasons that are
via the Fenton reaction, leading to mitochondrial damage and neurotoxicity98. although currently unknown104.
iron is normally stored within oligodendrocytes, it is released upon demyelination.
Iron chelation strategies have also been proposed as
In regions where iron accumulates, such as the MS deep grey matter, a global reduction in
neuronal density occurs with the presence of acutely injured axons and the accumulation a therapeutic avenue105. However, thus far, only pilot
of oxidized phospholipids and DNa in neurons, oligodendrocytes and axons99. studies have been performed to evaluate tolerability,
and only one patient experienced an improvement
restorative effects
in EDSS score using the chelator deferoxamine106,107.
restorative roles of iron include its role as a cofactor for enzymes involved in
remyelination and the repopulation of oligodendrocyte lineage cells98. For instance, iron
However, the adverse effect profile which includes local
is a cofactor for enzymes involved in cholesterol and lipid production; one example is skin irritation, anaemia, neutropenia, agranulocytosis
hydroxy-3-methylglutaryl coenzyme a reductase, which is enriched in oligodendrocytes and thrombocytopenia, as well as the unpleasant route
and catalyses the major regulatory step in cholesterol synthesis181. Other iron-containing of administration (subcutaneous infusion over 6–8 h for
enzymes important for myelin synthesis and turnover include squalene epoxidase and deferoxamine), might have hampered further investiga­
lipid dehydrogenases, respectively182,183. In rat pups fed an iron-deficient milk diet from tion of this treatment avenue in larger studies. Newer
birth to 3 weeks of age, the development of myelin is delayed184. Following lysolecithin- agents such as deferasirox and deferiprone are available
induced demyelination in adult mice, ferroportin-induced iron efflux from astrocytes is in oral formulation, but are still at preclinical stages of
required for remyelination, which occurs in part by a direct effect of iron on regeneration development and, therefore, the long-term tolerability
of oligodendrocyte lineage cells185.
of iron chelation strategies remains unclear.

www.nature.com/nrd
Reviews

After screening a library of 1,040 generic medica­ patients with PPMS to mitochondrial malfunctioning.
tions, our group found that 35 medications reduced Therefore, identifying and targeting mitochondrial
iron-mediated neurotoxicity in culture108. In addition, one genes associated with the risk of developing PPMS could
of these medications, the tricyclic antidepressant clomi­ be a potential approach for gene therapy. However, all
pramine, also reduced T cell and B cell proliferation and gene therapy approaches are still at a preclinical stage
had anti-oxidative properties. Clomipramine improved of development and, therefore, whether this treatment
the chronic course of EAE, including in the Biozzi ABH approach could have a clinically meaningful effect
mouse model of progression, and might, therefore, be a remains unclear.
promising candidate for progressive MS treatment108. The mtDNA deletions and mitochondrial energy
imbalances observed in progressive MS are probably
Targeting mitochondria caused by mitochondrial oxidative damage and impaired
Oxidative damage to cellular structures such as mito­ function of their respiratory chains, to which chronic
chondria is present in pathological specimens from neuroinflammation, in part, contributes116. The result is
patients with progressive MS109, and several studies a hypoxia-like state that produces injury, similar to that
have shown an increase in the number of mitochon­ observed in stroke109. To date, the reason why the num­
dria in both active and inactive lesions110. Additionally, ber of mitochondria increases in both active and inactive
demyelination leads to an increase in size of station­ lesions with a contrary global reduction in mitochon­
ary mitochondria and to an increase in the speed of drial density in NAWM remains elusive. Lucchinetti
axonal mitochondrial transport 111. These findings and colleagues117 hypothesized that mitochondrial
collectively infer an increased energy demand during defects are involved in the development of hypoxia.
demyelination. To investigate this hypothesis, the group analysed
However, after remyelination, the number of mito­ pattern III MS lesions, which are found in patients with
chondria does not necessarily return to the number seen fulminant MS and are defined by early loss of specific
under normal physiological conditions, as demonstrated myelin proteins and apoptosis of oligodendrocytes.
both in humans and in an animal model of demyelina­ In these lesions, investigation of cytochrome c oxidase
tion112. In addition to an increased density of mitochon­ — a major mitochondrial respiratory chain protein —
dria, an accumulation of mitochondrial DNA (mtDNA) revealed a reduction in the immunoreactivity of one
deletions also occurs in progressive MS, which is thought subunit, complex IV109. The catalytic component of
to account for some of the susceptibility to neurodegen­ complex IV, cytochrome c oxidase subunit 1 (COX1), as
eration52. Evidence from post-mortem studies in patients shown by immunohistochemistry, is reduced in oligo­
with SPMS shows that respiratory chain-deficient neu­ dendrocytes, astrocytes and axons, but not microglia.
rons are predominantly located in layer VI and the The authors postulated that the observed mitochon­
subcortical white matter113. Irrespective of the loca­ drial alterations could explain the damage to oligoden­
tion of known lesions, these neurons in post-mortem drocytes and axons that occurs during progression of
studies contained a high number of clonally expanded MS. Although the overall number of mitochondria is
mtDNA deletions, indicating that mitochondrial dam­ increased, this theory might explain why energy imbal­
age can self-replicate in a feed-forward manner; this ance occurs in progressive MS. Indeed, damage to the
expansion of impaired mitochondria could further respiratory chain could be indirectly linked to neuronal
exacerbate mitochondrial dysfunction and enhance damage. In post-mortem studies in patients with pro­
neurodegeneration. gressive MS, the reduction in complex IV activity was
Impairment of mitochondrial function is also found associated with axonal degeneration, as indicated by
in other cell types in patients with MS. Compared with the presence of non-phosphorylated neurofilament118.
patients with Alzheimer disease and Parkinson dis­ Furthermore, the inhibition of complex IV leads to
ease, patients with MS exhibit a fourfold increase in increased glutamate-mediated axonal injury in vitro118.
the numbers of respiratory enzyme-deficient choroid These findings stress the importance of mitochon­
plexus epithelial cells with clonally expanded mtDNA drial damage for the degenerative aspect of progressive
deletions114. Enhanced choroid plexus inflammation MS pathophysiology.
with consecutive mtDNA deletions could, therefore, Mitochondrial damage is partly explained by the
inform about delayed energy failure in progressive aforementioned oxidative stress via immune cells, but
MS. Gene therapy targeting clonally expanded mtDNA mitochondria themselves are also major producers
deletions might, therefore, be an interesting approach of ROS under homeostatic conditions. In addition to
to the treatment of progressive MS, but is far from mtDNA alterations, oxidative damage to DNA has been
clinical application. found in RRMS and SPMS lesions, although in the lat­
In addition to mtDNA deletions, other mitochon­ ter damaged DNA is associated with a reduced level of
drial defects have been associated with PPMS. Tranah inflammation119. Oxidative stress can be evaluated by
and colleagues 115 found an association between a measuring the expression of heat shock proteins such as
mitochondrial genetic variant and PPMS using mitochondrial 70-kDa heat shock protein (mtHSP70),
a genome-wide approach. Specifically, haplotypes J and a chaperone that is upregulated in cells during oxida­
T were associated with an increased risk of MS. When tive stress120 and is involved in the folding and assembly
stratified for disease course, haplotype J was associated of mitochondrial proteins121. In MS lesions, mtHSP70
with an increased risk of PPMS. This finding could, is upregulated in astrocytes and axons110. In EAE, over­
in part, explain the reportedly higher susceptibility of expression of mtHSP70 using gene therapy led to a

Nature Reviews | Drug Discovery


Reviews

major reduction in axonal damage, which was associated beneficially alter clinical signs in a model of acute
with improved mitochondrial respirational activities (as EAE. The authors argue that anchoring of mitochon­
assessed by the activity of NADH cytochrome oxido­ dria might have distinct effects during the acute phase,
reductase)121. The benefits of therapeutic restoration of in which anchored mitochondria produce increased
mtHSP70 under inflammatory conditions along with levels of ATP. During progression, the number of
the preponderance of oxidative stress in progressive damaged mitochondria increases, which, according to
MS suggest that targeting mitochondrial function via the authors, might result in a maladaptive state with
gene therapy could be a promising therapeutic avenue the production of ROS and a perpetuation of damage.
in progressive MS. Thus, inhibiting anchoring to improve mitophagy and
the transport of new mitochondria to the axon might
Axonal damage and potential treatments be a potential therapeutic approach. However, such a
Mitochondrial damage is one of several factors linked treatment approach would be challenging and require
to axonal damage, which begins with focal swelling and optimal timing.
can later progress to transection and fragmentation
of the distal axon. In MS lesions, axonal transection is Targeting ion channels
detected and is a pathological correlate of irreversible Progressive MS is associated with disturbances in ion
neurological impairment122. Axonal damage is an impor­ channels. To compensate for demyelination and per­
tant contributor to brain atrophy and is associated with mit axonal conduction, denuded axons express an
increasing disability in patients with progressive MS123. increased number of sodium ion (Na+) channels along
Interestingly, a study of clinically silent MS lesions showed the length of the axon129, which presumably increases
a huge variability in axonal pathology. Some patients Na+ influx130. Myelinated axons from an optic nerve
had nearly complete axonal loss, whereas others had isolated from a rat also produced a decrease in Na+/K+-
nearly normal axonal density. However, the study ATPase activity under anoxic conditions, resulting in a
had the drawback that the lesion distribution was hetero­ collapse of the ionic gradient and Na+ influx via incom­
geneous124. Damage to axons is found in acute demye­ pletely inactivated Na+ channels131. At a certain thresh­
linating plaques and also in inactive or remyelinating old, this elevated intra-axonal Na+ influx could reverse
lesions125. Axonal injury is associated with inflammation, the activity of the Na+/Ca2+ exchanger, leading to toxic
as shown by CD8+ T cell and macro­phage infiltrates levels of Ca2+ influx. New studies using sodium MRI
within lesions125. The presence of these inflammatory support these mechanisms by demonstrating that Na+
cells suggests that axonal damage is mediated by inflam­ levels are higher in the cortical grey matter, NAWM and
mation. Indeed, in animal models, axons can be injured deep grey matter, as well as in T1 lesions, from patients
by application of ROS and RNS, which can be derived with SPMS than in those from patients with RRMS132.
from activated macrophages and microglia, independent These heightened axonal Na+ levels might be a corre­
of demyelination53. In contrast to the situation in RRMS, late of axonal damage, as well as a mechanism driving
a small subset of slowly expanding lesions in progressive axonal injury in progressive MS. Thus, targeting Na+
MS show low-grade myelin and axonal destruction at channel alterations is considered a promising therapeu­
the white matter lesion margins with a substantially more tic intervention, although data from clinical trials are
diffuse inflammatory reaction, comprising perivascular controversial (Fig. 1).
cuffs of mononuclear cells, a diffuse infiltration of the Phenytoin is a therapeutic agent that targets voltage-
tissue by T cells and microglial activation14. gated Na+ channels. This agent was investigated in a
The disturbance of axonal transport of organelles placebo-controlled phase II trial in 86 patients with
contributes to both axonal and mitochondrial damage. optic neuritis, in which measurement of retinal nerve
Transport deficits even precede structural alterations fibre layer (RNFL) thickness was the primary end
of axons, as shown in vivo with two-photon imaging in point133. In the trial, a 30% reduction in the loss of
MS models126. Importantly, the use of anti-inflammatory RNFL thickness was found in the phenytoin-treated
interventions or redox-scavenging agents was able to group compared with the placebo group after 6 months,
reverse these transport deficits. To our knowledge, this supporting the neuroprotective effects of this approach.
approach has not been translated into clinical inves­ Another Na+ channel blocker tested in progressive MS
tigations. Taken together, ROS might damage axons is the anti-epileptic drug lamotrigine. Unfortunately, in
directly, but can also slow axonal transport, which results a randomized phase II trial that included 120 patients
in decreased mitochondrial densities in axons over a with SPMS, lamotrigine (at a target dose of 400 mg
long period of time, culminating in potential energetic daily) failed to attenuate the loss of cerebral volume
run-down. relative to placebo over 24 months134. Interestingly,
Axons remove damaged mitochondria through patients treated with lamotrigine had an early loss in
transport to the soma, where they are targeted for partial (central) cerebral volume, white matter volume
mitophagy127. Mitochondrial transport along axons is and whole-brain volume compared with the placebo
antagonized by anchoring proteins, such as syntaphilin group during the first 12 months, which was hypothe­
(SNPH), a neuron-specific mitochondrial protein. In sized to be related to its anti-inflammatory properties.
an animal model of progressive MS lacking compact However, this theory is still speculative. Importantly, no
myelin, deletion of Snph led to a decrease in axonal decrease in grey matter volume occurred with lamo­
injury, reduced oxidative stress and improved mito­ trigine treatment, and volume loss partially reversed
chondrial health128. However, Snph deletion did not after stopping treatment. Of interest, lamotrigine

www.nature.com/nrd
Reviews

treatment significantly reduced the deterioration of (an amino acid precursor for glutamate synthesis)145. The
the T25FW (P = 0.02) without an effect on other sec­ authors argued that increased glutamate release during
ondary outcome measures. As activated microglia and early stages of MS could be followed by synapse dysfunc­
macrophages exhibit increased expression of the Na+ tion or loss, leading to reduced glutamate levels in pro­
channel Nav1.6 and as blockade of Na+ channels leads gressive MS. Reducing early glutamatergic excitotoxicity
to improved EAE outcomes135, lamotrigine treatment is, therefore, a promising approach to reduce disability
in this trial might have dampened the inflammatory progression (Fig. 1).
response, which, paradoxically, might have decreased Preclinical evidence supporting this approach
brain volume. The potential benefits of lamotrigine comes from blockade of the glutamatergic α-amino-3-
are, however, still probably due to its assumed mech­ hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)
anism of action. Indeed, early volume loss has been and kai­nate (KA) receptors in EAE using the antago­
reported with the use of immunomodulatory drugs in nist NBQX, which improved clinical scores146. Another
patients with RRMS136 and can in part be explained by strategy to modulate glutamate neurotransmission is
fluid shifts137. These data speak to the complexities of by treatment with the amyotrophic lateral sclerosis
using morphometrics as a primary outcome measure drug riluzole, an inhibitor of glutamate release147 and a
in a clinical trial, as it remains challenging to separate modulator of AMPA and KA receptors that increases
pseudoatrophy related to fluid shifts from real atrophy. glutamate uptake 148. Riluzole reduces demyelina­
Although lamotrigine failed in this trial, blocking Na+ tion and axonal degeneration in EAE and leads to an
channels might, nevertheless, be a promising approach. amelio­rated clinical disease course149. In small studies
Another approach to the prevention of Na+ and in patients with PPMS, treatment with riluzole reduced
Ca influx is by blocking the activity of acid-sensing
2+
the rate of cervical cord atrophy and the development
ion channel 1 (ASIC1), which is permeable to Na+ of T1 hypointense lesions, but not the development of
and Ca2+. This approach is supported by preclinical new T2 lesions150,151. This finding might suggest a neuro­
data, whereby Asic1-knockout mice have reduced protective effect of riluzole through the inhibition of glu­
axonal degeneration following EAE138. Similarly, in tamatergic excitotoxicity. Currently, riluzole, amilo­ride
mice exhibiting chronic–relapsing EAE, treatment and the selective serotonin reuptake inhibitor (SSRI)
with the ASIC1 antagonist amiloride was neuroprotec­ fluoxetine are being tested in patients with SPMS in the
tive139. In samples from patients with progressive MS, phase IIb MS-SMART trial (NCT01910259; Table 1).
ASIC1 expression was associated with axonal damage Unfortunately, preliminary results presented at the 2018
in chronic lesions140, suggesting that targeting this ion Congress of the European Committee for Treatment
channel could have neuroprotective effects. Indeed, and Research in Multiple Sclerosis (ECTRIMS) showed
amiloride treatment markedly decreased brain atrophy no effect on percentage brain volume change in any
in the thalamus and also decreased corticospinal tract treatment arm152; final results are pending.
atrophy in a small cohort of 14 patients with PPMS140. Treatment with the N-methyl-d-aspartate receptor
Thus, targeting ASIC1 could be a promising approach (NMDAR) antagonist amantadine also improved func­
to the treatment of progressive MS. Currently, amiloride tional outcomes in EAE models but had no effect on
is being investigated in the randomized, double-blind, the mRNA expression of the key glutamate transport­
placebo-controlled ACTION trial in optic neuritis ers GLT1 and GLAST, which were increased by ~200%
(NCT01802489) and in the phase IIb MS-SMART in EAE153. Another NMDAR antagonist, memantine
trial in patients with SPMS (NCT01910259; Table 1). (used for the treatment of dementia), decreased glu­
Pregabalin, a medication targeting voltage-dependent tamate release and uptake, which was associated with
Ca2+ channels, ameliorated clinical symptoms in EAE behavioural improvements in an EAE model153. Despite
models (when administered in both a prophylactic and the benefits of these NMDAR antagonists in preclini­
a therapeutic treatment paradigm) and reversed the cal models, delayed administration resulted in synaptic
increase in neuronal intracellular Ca2+ levels in EAE degeneration and mitochondrial abnormalities, indi­
lesions141. Moreover, pregabalin improved myelin repair cating that treatment should be administered early to
in a model of lysolecithin-induced focal demyelination reduce potential glutamate-mediated damage to nerve
concomitantly with improvement in visual evoked endings153. Memantine and amantadine also decreased
potentials142. However, to our knowledge, no trials are the production of the pro-inflammatory cytokines
evaluating pregabalin in MS. IL-6, IL-1β and TNF in the brain154, and memantine
preserved axons in an acute optic neuritis model155,
Modulating glutamate neurotransmission demonstrating the general neuroprotective properties
Excess release of a specific neurotransmitter, glutamate, of these medications. Memantine attenuated B cell
is also observed in patients with progressive MS. At high receptor (BCR) and Toll-like receptor 4 (TLR4) signal­
concentrations, glutamate increases intracellular calcium ling through blockade of the Kv1.3 and KCa3.1 potas­
levels by directly opening postsynaptic ion channels143 sium channels, leading to enhanced production of the
and, therefore, excess glutamate levels are thought to anti-inflammatory cytokine IL-10 (ref.156). Thus, these
be a source of neuronal injury in PPMS. Glutamate medications might also diminish the detrimental effects
concentrations are increased in the NAWM in patients of lymphocytes. Indeed, Kv1.3 channels have been pos­
with early MS144, but decrease over time. Indeed, in an tulated to be an interesting target for T cell activation,
MRI-based study, patients with SPMS had an annual as blockade of Kv1.3 channels in T cells has been shown
decrease of ~5% in the levels of glutamate and glutamine to reduce clinical signs in EAE157.

Nature Reviews | Drug Discovery


Reviews

Promoting remyelination Neuro-Ophthalmic Supplement and high-contrast


Demyelination is a key feature of MS that is present in visual acuity) in participants in the RENEW trial con­
both grey and white matter structures. Although remye­ cluded that there was no difference in mean change
lination occurs, it is highly variable in patients with MS from baseline at 24 weeks between the placebo and
and generally declines with age (for a review see ref.8). opicinumab groups168.
As myelin is responsible for fast axonal conduction, its Another phase II trial (SYNERGY) evaluating the use
loss leads to slowed or absent communication along of opicinumab (3, 10, 30 or 100 mg kg−1 intravenously
axons. Another key attribute of myelin, and the oligo­ every 4 weeks up to week 72) versus placebo in the treat­
dendrocytes that produce it, is that it provides a con­ ment of relapsing MS was stopped by the sponsor as its
duit for axonal support by glia158. One method by which primary end point (proportion of participants confirmed
axons receive lactate, which can be used as a source of as positive responders for the primary multicomponent
energy, is through oligodendrocyte monocarboxylate end point) was missed; data have not yet been published.
transporter 1 (MCT1). Disruption of MCT1 in oligo­ However, as demonstrated in a platform presentation,
dendrocytes results in axonal degeneration, emphasiz­ patients in the SYNERGY trial who received intermedi­
ing the importance of oligodendrocyte health for axonal ate doses of opicinumab and had shorter disease dura­
degeneration independent of myelination159. A persistent tions might have benefited from the medication169. This
lack of myelin is thought to make axons more prone to possibility could form the basis of future targeted clinical
axonal degeneration. Indeed, when remyelination after trials investigating the effects of opicinumab treatment
cuprizone-induced demyelination is prevented by irra­ on remyelination in patients with MS.
diation, an increase in axonal loss occurs160, suggesting
that prolonged lack of myelin in MS accounts for axonal Elevating prolactin levels
drop-out. As remyelination might limit axonal degen­ Another possible approach to improving remyelina­
eration in MS, promoting remyelination is a potential tion is by elevating levels of prolactin, a hormone that
therapeutic strategy for the treatment of progressive induces remyelination170 and reduces disease severity
MS8 (Fig. 1). in EAE171. Pharmacologically, domperidone can be
One of the biggest challenges for remyelination is used to induce prolactin production, and is currently
that the microenvironment in lesions contains a large under investigation in a phase II clinical trial in SPMS
number of molecules that impair remyelination, such as (NCT02308137; Table 1).
chondroitin sulfate proteoglycans (CSPGs)161, fibronec­
tin162 and netrin-1 (ref.163). Thus, as discussed else­ Biotin
where8, the simultaneous targeting of several inhibitory High-dose biotin (MD1003) was investigated in a phase
molecules may be necessary for remyelination. III trial including 154 patients with progressive MS172.
After 12 months of treatment with either MD1003 or
LINGO1-targeted antibodies placebo, all patients received MD1003 for 12 months.
Largely as a means to spare axons, remyelination- The primary end point of improvement in EDSS scores
promoting therapies are deemed to be the next fron­ was achieved in 12.6% of MD1003-treated patients but
tier in the treatment of MS. One exciting approach in none of the placebo-treated patients. Since biotin is
is targeting leucine-rich repeat neuronal protein 1 a cofactor for four carboxylases involved in fatty acid
(LINGO1), a negative regulator of oligodendrocyte dif­ synthesis, the authors proposed that the mechanism
ferentiation164. Indeed, antagonizing LINGO1 improved involved in the beneficial effect of biotin was through
remyelination and clinical signs in EAE models 165. the promotion of remyelination172. Although the effect
A phase I clinical trial showed that the LINGO1- size was small, high-dose biotin might be a promising
targeted antibody opicinumab (BIIB033) is safe and treatment approach. However, the long-term benefit of
well tolerated in healthy volunteers and patients with MD1003 remains to be investigated. MD1003 is being
MS166. In the phase II RENEW trial, which evaluated investigated in a phase III follow-up study in patients
the use of opicinumab (100 mg kg−1 intravenously every with SPMS and PPMS, with EDSS and T25FW as the
4 weeks for six doses) in the treatment of optic neuritis, primary end points (NCT02936037; Table 1).
the primary end point of improving the latency of visual
evoked potentials of the affected eye compared with Emerging remyelinating agents
the non-affected eye167 was missed (mean difference Other drugs that promote remyelination are under active
in the intention-to-treat population –3.5 ms; P = 0.33). In investigation8, with several promising drugs for clinical
addition, optical coherence tomography did not show translation in progressive MS.
any improvement in retinal nerve thickness follow­ A screening approach investigating more than 1,000
ing treatment with opicinumab. This finding might compounds regarding their potential to improve remye­
have been partially due to the short treatment period, lination showed that antimuscarinic medications have
since a trend towards improvement was observed after strong remyelinating effects173. The most effective can­
32 weeks (mean difference in the intention-to-treat popu­ didate in vitro was the first-generation antihistamine
lation –6.1 ms; P = 0.071). The trial is being extended clemastine. In addition, clemastine improved remyelina­
(RENEWED; NCT02657915) to evaluate whether there tion in the mouse model of cuprizone-induced demye­
are positive effects after 2 years. A 2019 analysis of lination173. These findings led to the initiation of two
vision-related functional scores (25-item National Eye clinical trials evaluating clemastine in MS: ReBUILD
Institute Visual Functioning Questionnaire, 10-item (NCT02040298) and ReCOVER (NCT02521311).

www.nature.com/nrd
Reviews

ReBUILD was a phase II randomized, double-blinded, Additionally, available models incompletely mirror
placebo-controlled crossover trial in 50 patients with the human situation and, therefore, preclinical test­
MS and chronic demyelinating optic neuropathy that ing of treatment strategies remains challenging (Box 1).
assessed improvement in visual evoked potentials174. Importantly, the complexity of pathophysiological
Clemastine therapy significantly improved laten­ processes involved in progressive MS suggests that
cies by 1.7 ms (95% CI 0.5–2.9; P = 0.0048). However, targeting different processes simultaneously would
clemastine-treated patients reported more fatigue than be a promising therapeutic approach. Thus, the ideal
those in the placebo arm. To date, clemastine has not future of progressive MS therapy would involve precise,
been investigated in progressive MS. multi-dimensional therapeutic approaches, and would
Another remyelinating agent, the selective oestrogen probably involve combination therapies. As discussed in
receptor modulator bazedoxifene, has been found to this Review, a plethora of potential treatment approaches
enhance the differentiation of oligodendrocyte precursor are at different stages of development. However, prioriti­
cells into oligodendrocytes and to promote remyelination zation of these therapeutic agents is currently difficult as
in an in vivo model of focal demyelination, presum­ably the most prominent pathophysio­logical processes driving
functioning via 3β-hydroxysteroid-Δ8,Δ7-isomerase, progression, which are the most crucial to target, remain
a key enzyme involved in the cholesterol biosynthesis uncertain. Nonetheless, our opinion is that neuroinflam­
pathway175. Another screen using primary rat optic mation that is driven not only by microglia, astrocytes and
nerve-derived progenitor cells identified benztropine, CNS-infiltrated macrophages, but also by B and T cells,
an anticholinergic, which enhanced remyelination both at least needs to be attenuated. In parallel, we believe that
in vitro and in vivo176. neuro­protective and remyelination-promoting strategies
need to be introduced. Comorbid factors associated with
Conclusions progression, including diabetes mellitus, hypertension
The pathological hallmarks of progressive MS dif­ and smoking (which have been reviewed elsewhere177),
fer from those of RRMS in their extent. In contrast to also need to be addressed therapeutically to reduce pro­
RRMS, progressive MS is characterized by a greater gression; however, their relative importance is probably
degree of atrophy resulting from chronic axonal loss, less than that of neuroinflammation-driven injury or the
impaired homeostasis of glia with production of ROS need for strategies that promote neuroprotection and
and RNS, and mitochondrial damage with clonal expan­ remyelination.
sion of mtDNA deletions. Demyelination increases the In addition to selecting the best compounds for
energy demand of axons. Impaired axonal transport future trials, trial design must be optimized (discussed
of organelles, especially mitochondria, contributes elsewhere178) as outcome parameters are heterogeneous
to axonal dysfunction with altered metabolism and and often do not show treatment effects on disease pro­
disturbances of ion channels such as sodium chan­ gression. In conclusion, with the growing understanding
nels. In addition, different immune cell types, such as of the pathophysiology of progressive MS, the number of
B and T lymphocytes, are involved in the perpetuation treatment approaches targeting specific pathological
of progression, for example, through the expansion of mechanisms has increased, raising hope for a bright
B cell aggregates. future for progressive MS therapy.
Targeting these pathophysiological processes is dif­
Published online xx xx xxxx
ficult owing to the diversity of mechanisms involved.

1. Reich, D. S., Lucchinetti, C. F. & Calabresi, P. A. 8. Plemel, J. R., Liu, W. Q. & Yong, V. W. Remyelination 16. Komori, M. et al. Cerebrospinal fluid markers reveal
Multiple sclerosis. N. Engl. J. Med. 378, 169–180 therapies: a new direction and challenge in multiple intrathecal inflammation in progressive multiple
(2018). sclerosis. Nat. Rev. Drug Discov. 16, 617–634 sclerosis. Ann. Neurol. 78, 3–20 (2015).
2. Weinshenker, B. G. et al. The natural history of multiple (2017). 17. Lassmann, H. Pathogenic mechanisms associated
sclerosis: a geographically based study. I. clinical course A comprehensive summary of potential with different clinical courses of multiple sclerosis.
and disability. Brain 112 (Pt 1), 133–146 (1989). remyelinating medications. Front. Immunol. 9, 3116 (2018).
3. Torkildsen, O., Myhr, K. M. & Bo, L. Disease-modifying 9. Kutzelnigg, A. & Lassmann, H. Pathology of 18. Schenkel, J. M. & Masopust, D. Tissue-resident
treatments for multiple sclerosis – a review of approved multiple sclerosis and related inflammatory memory T cells. Immunity 41, 886–897 (2014).
medications. Eur. J. Neurol. 23 (Suppl. 1), 18–27 demyelinating diseases. Handb. Clin. Neurol. 122, 19. Kapoor, R. et al. Effect of natalizumab on disease
(2016). 15–58 (2014). progression in secondary progressive multiple
4. Brown, J. W. L. et al. Association of initial disease- 10. Frischer, J. M. et al. The relation between sclerosis (ASCEND): a phase 3, randomised,
modifying therapy with later conversion to secondary inflammation and neurodegeneration in multiple double-blind, placebo-controlled trial with an
progressive multiple sclerosis. JAMA 321, 175–187 sclerosis brains. Brain 132, 1175–1189 (2009). open-label extension. Lancet Neurol. 17, 405–415
(2019). 11. Hochmeister, S. et al. Dysferlin is a new marker (2018).
5. Lublin, F. et al. Oral fingolimod in primary progressive for leaky brain blood vessels in multiple sclerosis. 20. Greenwood, J., Steinman, L. & Zamvil, S. S. Statin
multiple sclerosis (INFORMS): a phase 3, randomised, J. Neuropathol. Exp. Neurol. 65, 855–865 (2006). therapy and autoimmune disease: from protein
double-blind, placebo-controlled trial. Lancet 387, 12. Lassmann, H. Targets of therapy in progressive MS. prenylation to immunomodulation. Nat. Rev. Immunol.
1075–1084 (2016). Mult. Scler. 23, 1593–1599 (2017). 6, 358–370 (2006).
6. Montalban, X. et al. Ocrelizumab versus placebo in 13. Dal-Bianco, A. et al. Slow expansion of multiple 21. de Oliveira, D. M. et al. Simvastatin ameliorates
primary progressive multiple sclerosis. N. Engl. J. Med. sclerosis iron rim lesions: pathology and 7T magnetic experimental autoimmune encephalomyelitis by
376, 209–220 (2017). resonance imaging. Acta Neuropathol. 133, 25–42 inhibiting Th1/Th17 response and cellular infiltration.
This pivotal trial shows the efficacy of ocrelizumab (2017). Inflammopharmacology 23, 343–354 (2015).
in PPMS and forms the basis of its FDA and EMA 14. Kutzelnigg, A. et al. Cortical demyelination and diffuse 22. Zhang, X., Tao, Y., Troiani, L. & Markovic-Plese, S.
approval. white matter injury in multiple sclerosis. Brain 128, Simvastatin inhibits IFN regulatory factor 4 expression
7. Kappos, L. et al. Siponimod versus placebo in secondary 2705–2712 (2005). and Th17 cell differentiation in CD4+T cells derived
progressive multiple sclerosis (EXPAND): a double-blind, A key neuropathology study, showing the association from patients with multiple sclerosis. J. Immunol. 187,
randomised, phase 3 study. Lancet 391, 1263–1273 between cortical demyelination and white matter 3431–3437 (2011).
(2018). injury. 23. Paintlia, A. S. et al. HMG-CoA reductase inhibitor
This study reports that siponimod treatment 15. Androdias, G. et al. Meningeal T cells associate with augments survival and differentiation of oligodendrocyte
led to a 21% reduced risk of disability progression diffuse axonal loss in multiple sclerosis spinal cords. progenitors in animal model of multiple sclerosis.
in SPMS. Ann Neurol 68, 465–476 (2010). FASEB J. 19, 1407–1421 (2005).

Nature Reviews | Drug Discovery


Reviews

24. Wang, J., Xiao, Y., Luo, M. & Luo, H. Statins for multiple 47. Fereidan-Esfahani, M., Bruck, W. & Weber, M. S. 70. Faissner, S. & Gold, R. Oral therapies for multiple
sclerosis. Cochrane Database Syst. Rev. 12, CD008386 Targeting central nervous system B cells in progression sclerosis. Cold Spring Harb. Perspect. Med. 9,
(2011). of multiple sclerosis: is intrathecal anti-CD20 a a032011 (2019).
25. Chataway, J. et al. Effect of high-dose simvastatin on therapeutic option? JAMA Neurol. 72, 1407–1408 71. Linker, R. A. et al. Fumaric acid esters exert
brain atrophy and disability in secondary progressive (2015). neuroprotective effects in neuroinflammation via
multiple sclerosis (MS-STAT): a randomised, placebo- 48. Svenningsson, A. et al. Rapid depletion of B lymphocytes activation of the Nrf2 antioxidant pathway. Brain 134,
controlled, phase 2 trial. Lancet 383, 2213–2221 by ultra-low-dose rituximab delivered intrathecally. 678–692 (2011).
(2014). Neurol. Neuroimmunol. Neuroinflamm. 2, e79 (2015). 72. Strassburger-Krogias, K. et al. Fumarate treatment in
26. Chan, D. et al. Effect of high-dose simvastatin on 49. Komori, M. et al. Insufficient disease inhibition by progressive forms of multiple sclerosis: first results of
cognitive, neuropsychiatric, and health-related intrathecal rituximab in progressive multiple sclerosis. a single-center observational study. Ther. Adv. Neurol.
quality-of-life measures in secondary progressive Ann. Clin. Transl. Neurol. 3, 166–179 (2016). Disord. 7, 232–238 (2014).
multiple sclerosis: secondary analyses from the 50. Naegelin, Y. et al. Association of rituximab treatment 73. Valentin-Torres, A. et al. Sustained TNF production
MS-STAT randomised, placebo-controlled trial. with disability progression among patients with by central nervous system infiltrating macrophages
Lancet Neurol. 16, 591–600 (2017). secondary progressive multiple sclerosis. JAMA promotes progressive autoimmune encephalomyelitis.
27. Kurte, M. et al. IL17/IL17RA as a novel signaling axis Neurol. 76, 274-281, (2019). J. Neuroinflammation 13, 46 (2016).
driving mesenchymal stem cell therapeutic function 51. Siders, W. et al. GZ402668, a next-generation 74. Theibich, A., Dreyer, L., Magyari, M. & Locht, H.
in experimental autoimmune encephalomyelitis. anti-CD52 antibody, displays decreased Demyelinizing neurological disease after treatment
Front. Immunol. 9, 802 (2018). proinflammatory cytokine release in vitro. Neurology with tumor necrosis factor alpha-inhibiting agents
28. Rice, C. M. et al. Safety and feasibility of autologous 86 (Suppl. 16), P3.068 (2016). in a rheumatological outpatient clinic: description
bone marrow cellular therapy in relapsing-progressive 52. Lassmann, H., van Horssen, J. & Mahad, D. of six cases. Clin. Rheumatol. 33, 719–723
multiple sclerosis. Clin. Pharmacol. Ther. 87, 679–685 Progressive multiple sclerosis: pathology and (2014).
(2010). pathogenesis. Nat. Rev. Neurol. 8, 647–656 (2012). 75. The Lenercept Multiple Sclerosis Study Group and
29. Connick, P. et al. Autologous mesenchymal stem cells A key review on the neuropathology of the University of British Columbia MS/MRI Analysis
for the treatment of secondary progressive multiple progressive MS. Group. TNF neutralization in MS: results of a
sclerosis: an open-label phase 2a proof-of-concept 53. Nikic, I. et al. A reversible form of axon damage in randomized, placebo-controlled multicenter study.
study. Lancet Neurol. 11, 150–156 (2012). experimental autoimmune encephalomyelitis and Neurology 53, 457–465 (1999).
30. Rice, C. M. et al. Assessment of bone marrow-derived multiple sclerosis. Nat. Med. 17, 495–499 (2011). 76. Steelman, A. J. & Li, J. Astrocyte galectin-9 potentiates
cellular therapy in progressive multiple sclerosis 54. Radbruch, H. et al. Ongoing oxidative stress causes microglial TNF secretion. J. Neuroinflammation 11,
(ACTiMuS): study protocol for a randomised controlled subclinical neuronal dysfunction in the recovery phase 144 (2014).
trial. Trials 16, 463 (2015). of EAE. Front. Immunol. 7, 92 (2016). 77. Burman, J. & Svenningsson, A. Cerebrospinal fluid
31. Mancardi, G. L. et al. Autologous hematopoietic stem 55. Sawada, H. et al. Activated microglia affect the nigro- concentration of galectin-9 is increased in secondary
cell transplantation in multiple sclerosis: a phase II striatal dopamine neurons differently in neonatal and progressive multiple sclerosis. J. Neuroimmunol. 292,
trial. Neurology 84, 981–988 (2015). aged mice treated with 1-methyl-4-phenyl-1,2,3,6- 40–44 (2016).
32. Burt, R. K. et al. Effect of nonmyeloablative tetrahydropyridine. J. Neurosci. Res. 85, 1752–1761 78. Savarin, C. et al. Astrocyte response to IFN-gamma
hematopoietic stem cell transplantation vs continued (2007). limits IL-6-mediated microglia activation and
disease-modifying therapy on disease progression in 56. Streit, W. J. & Xue, Q. S. Human CNS immune progressive autoimmune encephalomyelitis.
patients with relapsing-remitting multiple sclerosis: senescence and neurodegeneration. Curr. Opin. J. Neuroinflammation 12, 79 (2015).
a randomized clinical trial. JAMA 321, 165–174 Immunol. 29, 93–96 (2014). 79. Araki, M. et al. Clinical improvement in a patient
(2019). 57. Ransohoff, R. M. A polarizing question: do M1 and M2 with neuromyelitis optica following therapy with the
33. Muraro, P. A. et al. Long-term outcomes after microglia exist? Nat. Neurosci. 19, 987–991 (2016). anti-IL-6 receptor monoclonal antibody tocilizumab.
autologous hematopoietic stem cell transplantation for 58. Herder, V. et al. Dynamic changes of microglia/ Mod. Rheumatol. 23, 827–831 (2013).
multiple sclerosis. JAMA Neurol. 74, 459–469 (2017). macrophage M1 and M2 polarization in Theiler’s 80. Araki, M. et al. Efficacy of the anti-IL-6 receptor
A meta-analysis of studies investigating stem cell murine encephalomyelitis. Brain Pathol. 25, 712–723 antibody tocilizumab in neuromyelitis optica: a pilot
transplantation in MS. (2015). study. Neurology 82, 1302–1306 (2014).
34. Michel, L. et al. B cells in the multiple sclerosis 59. Miron, V. E. et al. M2 microglia and macrophages 81. Sato, H. et al. Tocilizumab treatment safety in
central nervous system: trafficking and contribution drive oligodendrocyte differentiation during CNS rheumatoid arthritis in a patient with multiple sclerosis:
to CNS-compartmentalized inflammation. Front. remyelination. Nat. Neurosci. 16, 1211–1218 a case report. BMC Res. Notes 7, 641 (2014).
Immunol. 6, 636 (2015). (2013). 82. Beauchemin, P. & Carruthers, R. MS arising during
35. Pollinger, B. et al. Spontaneous relapsing-remitting This study shows the role of M2 myeloid cells in tocilizumab therapy for rheumatoid arthritis.
EAE in the SJL/J mouse: MOG-reactive transgenic supporting oligodendrocyte differentiation. Mult. Scler. 22, 254–256 (2016).
T cells recruit endogenous MOG-specific B cells. 60. Koch, M. W. et al. Hydroxychloroquine reduces 83. Brundula, V., Rewcastle, N. B., Metz, L. M.,
J. Exp. Med. 206, 1303–1316 (2009). microglial activity and attenuates experimental Bernard, C. C. & Yong, V. W. Targeting leukocyte
36. Li, R. et al. Proinflammatory GM-CSF-producing B cells autoimmune encephalomyelitis. J. Neurol. Sci. 358, MMPs and transmigration: minocycline as a
in multiple sclerosis and B cell depletion therapy. 131–137 (2015). potential therapy for multiple sclerosis. Brain 125,
Sci. Transl Med. 7, 310ra166 (2015). 61. White, M., Webster, G., O’Sullivan, D., Stone, S. 1297–1308 (2002).
37. Magliozzi, R. et al. B-cell enrichment and Epstein-Barr & La Flamme, A. C. Targeting innate receptors with 84. Giuliani, F., Hader, W. & Yong, V. W. Minocycline
virus infection in inflammatory cortical lesions in MIS416 reshapes Th responses and suppresses attenuates T cell and microglia activity to impair
secondary progressive multiple sclerosis. J. Neuropathol. CNS disease in a mouse model of multiple sclerosis. cytokine production in T cell-microglia interaction.
Exp. Neurol. 72, 29–41 (2013). PLOS ONE 9, e87712 (2014). J. Leuk. Biol. 78, 135–143 (2005).
38. Howell, O. W. et al. Meningeal inflammation is 62. Cho, Y. et al. Allosteric inhibition of macrophage 85. Giuliani, F., Fu, S. A., Metz, L. M. & Yong, V. W. Effective
widespread and linked to cortical pathology in migration inhibitory factor revealed by ibudilast. combination of minocycline and interferon-beta in a
multiple sclerosis. Brain 134, 2755–2771 (2011). Proc. Natl Acad. Sci. USA 107, 11313–11318 model of multiple sclerosis. J. Neuroimmunol. 165,
A key neuropathological study, demonstrating the (2010). 83–91 (2005).
link between meningeal inflammation and cortical 63. Fox, R. J. et al. Design, rationale, and baseline 86. Giuliani, F. et al. Additive effect of the combination
neurodegeneration. characteristics of the randomized double-blind of glatiramer acetate and minocycline in a model
39. Choi, S. R. et al. Meningeal inflammation plays a phase II clinical trial of ibudilast in progressive of MS. J. Neuroimmunol. 158, 213–221 (2005).
role in the pathology of primary progressive multiple multiple sclerosis. Contemp. Clin. Trials 50, 166–177 87. Metz, L. M. et al. Glatiramer acetate in combination
sclerosis. Brain 135, 2925–2937 (2012). (2016). with minocycline in patients with relapsing–remitting
40. Magliozzi, R. et al. A gradient of neuronal loss 64. Fox, R. J. et al. Phase 2 trial of ibudilast in progressive multiple sclerosis: results of a Canadian, multicenter,
and meningeal inflammation in multiple sclerosis. multiple sclerosis. N. Engl. J. Med. 379, 846–855 double-blind, placebo-controlled trial. Mult. Scler. 15,
Ann. Neurol. 68, 477–493 (2010). (2018). 1183–1194 (2009).
41. Romme Christensen, J. et al. Systemic inflammation This trial showed reduced brain atrophy in 88. Metz, L. M. et al. Trial of minocycline in a clinically
in progressive multiple sclerosis involves follicular progressive MS upon ibudilast treatment. isolated syndrome of multiple sclerosis. N. Engl. J. Med.
T-helper, Th17- and activated B-cells and correlates 65. Chechneva, O. V. et al. Low dose dextromethorphan 376, 2122–2133 (2017).
with progression. PLOS ONE 8, e57820 (2013). attenuates moderate experimental autoimmune 89. Sorensen, P. S. et al. Minocycline added to
42. Blauth, K. et al. Antibodies produced by clonally encephalomyelitis by inhibiting NOX2 and reducing subcutaneous interferon beta-1a in multiple sclerosis:
expanded plasma cells in multiple sclerosis peripheral immune cells infiltration in the spinal cord. randomized RECYCLINE study. Eur. J. Neurol. 23,
cerebrospinal fluid cause demyelination of spinal cord Neurobiol. Dis. 44, 63–72 (2011). 861–870 (2016).
explants. Acta Neuropathol. 130, 765–781 (2015). 66. Lisak, R. P., Nedelkoska, L. & Benjamins, J. A. 90. Yi, W., Schluter, D. & Wang, X. Astrocytes in
43. Rojas, O. L. et al. Recirculating intestinal IgA-producing Effects of dextromethorphan on glial cell function: multiple sclerosis and experimental autoimmune
cells regulate neuroinflammation via IL-10. Cell 176, proliferation, maturation, and protection from encephalomyelitis: Star-shaped cells illuminating the
610–624.e618 (2019). cytotoxic molecules. Glia 62, 751–762 (2014). darkness of CNS autoimmunity. Brain Behav. Immun.
44. Kappos, L. et al. Atacicept in multiple sclerosis (ATAMS): 67. Liu, Y. et al. Dextromethorphan protects dopaminergic https://doi.org/10.1016/j.bbi.2019.05.029 (2019).
a randomised, placebo-controlled, double-blind, phase 2 neurons against inflammation-mediated degeneration 91. Liddelow, S. A. et al. Neurotoxic reactive astrocytes
trial. Lancet Neurol. 13, 353–363 (2014). through inhibition of microglial activation. are induced by activated microglia. Nature 541,
45. Hawker, K. et al. Rituximab in patients with primary J. Pharmacol. Exp. Ther. 305, 212–218 (2003). 481–487 (2017).
progressive multiple sclerosis: results of a randomized 68. Spain, R. et al. Lipoic acid in secondary progressive 92. Rothhammer, V. et al. Microglial control of astrocytes
double-blind placebo-controlled multicenter trial. MS: a randomized controlled pilot trial. Neurol. in response to microbial metabolites. Nature 557,
Ann. Neurol. 66, 460–471 (2009). Neuroimmunol. Neuroinflamm. 4, e374 (2017). 724–728 (2018).
46. Piccio, L. et al. Changes in B- and T-lymphocyte and 69. Faissner, S. & Gold, R. Efficacy and safety of the newer 93. Mayo, L. et al. Regulation of astrocyte activation
chemokine levels with rituximab treatment in multiple multiple sclerosis drugs approved since 2010. CNS by glycolipids drives chronic CNS inflammation.
sclerosis. Arch. Neurol. 67, 707–714 (2010). Drugs 32, 269–287 (2018). Nat. Med. 20, 1147–1156 (2014).

www.nature.com/nrd
Reviews

94. Pebay, A. et al. Sphingosine-1-phosphate induces 118. Mahad, D. J. et al. Mitochondrial changes within 142. Daneshdoust, D. et al. Pregabalin enhances myelin
proliferation of astrocytes: regulation by intracellular axons in multiple sclerosis. Brain 132, 1161–1174 repair and attenuates glial activation in lysolecithin-
signalling cascades. Eur. J. Neurosci. 13, 2067–2076 (2009). induced demyelination model of rat optic chiasm.
(2001). 119. Haider, L. et al. Oxidative damage in multiple sclerosis Neuroscience 344, 148–156 (2017).
95. Bigaud, M., Guerini, D., Billich, A., Bassilana, F. & lesions. Brain 134, 1914–1924 (2011). 143. Mehta, A., Prabhakar, M., Kumar, P., Deshmukh, R. &
Brinkmann, V. Second generation S1P pathway 120. Mitsumoto, A., Takeuchi, A., Okawa, K. & Sharma, P. L. Excitotoxicity: bridge to various triggers
modulators: research strategies and clinical Nakagawa, Y. A subset of newly synthesized in neurodegenerative disorders. Eur. J. Pharmacol.
developments. Biochim. Biophys. Acta 1841, polypeptides in mitochondria from human endothelial 698, 6–18 (2013).
745–758 (2014). cells exposed to hydroperoxide stress. Free Radic. 144. Tisell, A. et al. Increased concentrations of glutamate
96. Rouach, N. et al. S1P inhibits gap junctions in Biol. Med. 32, 22–37 (2002). and glutamine in normal-appearing white matter
astrocytes: involvement of G and Rho GTPase/ROCK. 121. Talla, V. et al. Gene therapy with mitochondrial of patients with multiple sclerosis and normal MR
Eur. J. Neurosci. 23, 1453–1464 (2006). heat shock protein 70 suppresses visual loss imaging brain scans. PLOS ONE 8, e61817 (2013).
97. Choi, J. W. et al. FTY720 (fingolimod) efficacy in an and optic atrophy in experimental autoimmune 145. MacMillan, E. L. et al. Progressive multiple sclerosis
animal model of multiple sclerosis requires astrocyte encephalomyelitis. Invest. Ophthalmol. Vis. Sci. 55, exhibits decreasing glutamate and glutamine over
sphingosine 1-phosphate receptor 1 (S1P1) 5214–5226 (2014). two years. Mult. Scler. 22, 112–116 (2016).
modulation. Proc. Natl Acad. Sci. USA 108, 122. Trapp, B. D. et al. Axonal transection in the lesions 146. Pitt, D., Werner, P. & Raine, C. S. Glutamate
751–756 (2011). of multiple sclerosis. N. Engl. J. Med. 338, 278–285 excitotoxicity in a model of multiple sclerosis.
98. Stephenson, E., Nathoo, N., Mahjoub, Y., Dunn, J. F. (1998). Nat. Med. 6, 67–70 (2000).
& Yong, V. W. Iron in multiple sclerosis: roles in This is the first report of the profound destruction 147. Wang, S. J., Wang, K. Y. & Wang, W. C. Mechanisms
neurodegeneration and repair. Nat. Rev. Neurol. 10, of axons within MS lesions. underlying the riluzole inhibition of glutamate release
459–468 (2014). 123. Mahad, D. H., Trapp, B. D. & Lassmann, H. from rat cerebral cortex nerve terminals
99. Haider, L. et al. Multiple sclerosis deep grey matter: Pathological mechanisms in progressive multiple (synaptosomes). Neuroscience 125, 191–201 (2004).
the relation between demyelination, neurodegeneration, sclerosis. Lancet Neurol 14, 183–193 (2015). 148. Azbill, R. D., Mu, X. & Springer, J. E. Riluzole increases
inflammation and iron. J. Neurol. Neurosurg. Psychiatry 124. Mews, I., Bergmann, M., Bunkowski, S., Gullotta, F. high-affinity glutamate uptake in rat spinal cord
85, 1386–1395 (2014). & Bruck, W. Oligodendrocyte and axon pathology in synaptosomes. Brain Res. 871, 175–180 (2000).
100. Friese, M. A., Schattling, B. & Fugger, L. Mechanisms of clinically silent multiple sclerosis lesions. Mult. Scler. 149. Gilgun-Sherki, Y., Panet, H., Melamed, E. & Offen, D.
neurodegeneration and axonal dysfunction in multiple 4, 55–62 (1998). Riluzole suppresses experimental autoimmune
sclerosis. Nat. Rev. Neurol. 10, 225–238 (2014). 125. Bitsch, A., Schuchardt, J., Bunkowski, S., Kuhlmann, T. encephalomyelitis: implications for the treatment of
101. Hametner, S. et al. Iron and neurodegeneration in the & Bruck, W. Acute axonal injury in multiple sclerosis. multiple sclerosis. Brain Res. 989, 196–204 (2003).
multiple sclerosis brain. Ann. Neurol. 74, 848–861 Correlation with demyelination and inflammation. 150. Kalkers, N. F., Barkhof, F., Bergers, E., van Schijndel, R.
(2013). Brain 123 (Pt 6), 1174–1183 (2000). & Polman, C. H. The effect of the neuroprotective agent
102. Kroner, A. et al. TNF and increased intracellular iron 126. Sorbara, C. D. et al. Pervasive axonal transport riluzole on MRI parameters in primary progressive
alter macrophage polarization to a detrimental M1 deficits in multiple sclerosis models. Neuron 84, multiple sclerosis: a pilot study. Mult. Scler. 8, 532–533
phenotype in the injured spinal cord. Neuron 83, 1183–1190 (2014). (2002).
1098–1116 (2014). 127. Zheng, Y. R., Zhang, X. N. & Chen, Z. Mitochondrial 151. Killestein, J., Kalkers, N. F. & Polman, C. H. Glutamate
103. Zarruk, J. G. et al. Expression of iron homeostasis transport serves as a mitochondrial quality control inhibition in MS: the neuroprotective properties of
proteins in the spinal cord in experimental autoimmune strategy in axons: implications for central nervous riluzole. J. Neurol. Sci. 233, 113–115 (2005).
encephalomyelitis and their implications for iron system disorders. CNS Neurosci. Ther. 25, 876–886 152. Chataway, J. MS-SMART trial: a multi-arm phase 2b
accumulation. Neurobiol. Dis. 81, 93–107 (2015). (2019). randomised, double blind, parallel group, placebo-
104. Faissner, S. et al. Unexpected additive effects of 128. Joshi, D. C. et al. Deletion of mitochondrial anchoring controlled clinical trial comparing the efficacy of three
minocycline and hydroxychloroquine in models protects dysmyelinating shiverer: implications for neuroprotective drugs in secondary progressive
of multiple sclerosis: prospective combination progressive MS. J. Neurosci. 35, 5293–5306 (2015). multiple sclerosis [NCT01910259]. ECTRIMS Online
treatment for progressive disease? Mult. Scler. 24, 129. Craner, M. J. et al. Molecular changes in neurons Library 232077, 324 (2018).
1543–1556 (2017). in multiple sclerosis: altered axonal expression of 153. Sulkowski, G., Dabrowska-Bouta, B., Salinska, E. &
105. Weigel, K. J., Lynch, S. G. & LeVine, S. M. Iron Nav1.2 and Nav1.6 sodium channels and Na+/Ca2+ Struzynska, L. Modulation of glutamate transport and
chelation and multiple sclerosis. ASN Neuro. 6, exchanger. Proc. Natl Acad. Sci. USA 101, 8168–8173 receptor binding by glutamate receptor antagonists
e00136 (2014). (2004). in EAE rat brain. PLOS ONE 9, e113954 (2014).
106. Lynch, S. G., Peters, K. & LeVine, S. M. Desferrioxamine 130. Waxman, S. G. Mechanisms of disease: sodium 154. Sulkowski, G., Dabrowska-Bouta, B., Chalimoniuk, M.
in chronic progressive multiple sclerosis: a pilot study. channels and neuroprotection in multiple sclerosis – & Struzynska, L. Effects of antagonists of glutamate
Mult. Scler. 2, 157–160 (1996). current status. Nat. Clin. Pract. Neurol. 4, 159–169 receptors on pro-inflammatory cytokines in the brain
107. Lynch, S. G., Fonseca, T. & LeVine, S. M. A multiple (2008). cortex of rats subjected to experimental autoimmune
course trial of desferrioxamine in chronic progressive 131. Stys, P. K., Waxman, S. G. & Ransom, B. R. Ionic encephalomyelitis. J. Neuroimmunol. 261, 67–76
multiple sclerosis. Cell. Mol. Biol. 46, 865–869 (2000). mechanisms of anoxic injury in mammalian CNS (2013).
108. Faissner, S. et al. Systematic screening of generic white matter: role of Na+ channels and Na(+)-Ca2+ 155. Suhs, K. W. et al. N-methyl-D-aspartate receptor
drugs for progressive multiple sclerosis identifies exchanger. J. Neurosci. 12, 430–439 (1992). blockade is neuroprotective in experimental
clomipramine as a promising therapeutic. 132. Paling, D. et al. Sodium accumulation is associated autoimmune optic neuritis. J. Neuropathol. Exp.
Nat. Commun. 8, 1990 (2017). with disability and a progressive course in multiple Neurol. 73, 507–518 (2014).
This systematic screening of generic medications sclerosis. Brain 136, 2305–2317 (2013). 156. Simma, N. et al. NMDA-receptor antagonists block
identified several medications that are protective 133. Raftopoulos, R. et al. Phenytoin for neuroprotection B-cell function but foster IL-10 production in BCR/
against iron-mediated neurotoxicity. in patients with acute optic neuritis: a randomised, CD40-activated B cells. Cell Commun. Signal. 12, 75
109. Mahad, D., Ziabreva, I., Lassmann, H. & Turnbull, D. placebo-controlled, phase 2 trial. Lancet Neurol. 15, (2014).
Mitochondrial defects in acute multiple sclerosis 259–269 (2016). 157. Beeton, C. et al. Selective blockade of T lymphocyte
lesions. Brain 131, 1722–1735 (2008). 134. Kapoor, R. et al. Lamotrigine for neuroprotection in K(+) channels ameliorates experimental autoimmune
This study investigated mitochondrial defects in MS. secondary progressive multiple sclerosis: a randomised, encephalomyelitis, a model for multiple sclerosis.
110. Witte, M. E. et al. Enhanced number and activity of double-blind, placebo-controlled, parallel-group trial. Proc. Natl Acad. Sci. USA 98, 13942–13947 (2001).
mitochondria in multiple sclerosis lesions. J. Pathol. Lancet Neurol. 9, 681–688 (2010). 158. Nave, K. A. Myelination and support of axonal
219, 193–204 (2009). 135. Craner, M. J. et al. Sodium channels contribute to integrity by glia. Nature 468, 244–252 (2010).
111. Kiryu-Seo, S., Ohno, N., Kidd, G. J., Komuro, H. microglia/macrophage activation and function in EAE 159. Lee, Y. et al. Oligodendroglia metabolically support
& Trapp, B. D. Demyelination increases axonal and MS. Glia 49, 220–229 (2005). axons and contribute to neurodegeneration. Nature
stationary mitochondrial size and the speed of axonal 136. Miller, D. H. et al. MRI outcomes in a placebo-controlled 487, 443–448 (2012).
mitochondrial transport. J. Neurosci. 30, 6658–6666 trial of natalizumab in relapsing MS. Neurology 68, This study demonstrates the relevance of
(2010). 1390–1401 (2007). oligodendrocytes for trophic axonal support.
112. Zambonin, J. L. et al. Increased mitochondrial 137. Zivadinov, R. et al. Mechanisms of action of disease- 160. Irvine, K. A. & Blakemore, W. F. Remyelination
content in remyelinated axons: implications for modifying agents and brain volume changes in protects axons from demyelination-associated
multiple sclerosis. Brain 134, 1901–1913 (2011). multiple sclerosis. Neurology 71, 136–144 (2008). axon degeneration. Brain 131, 1464–1477
113. Campbell, G. R. et al. Mitochondrial DNA deletions 138. Friese, M. A. et al. Acid-sensing ion channel-1 (2008).
and neurodegeneration in multiple sclerosis. contributes to axonal degeneration in autoimmune 161. Lau, L. W., Cua, R., Keough, M. B., Haylock-Jacobs, S.
Ann. Neurol. 69, 481–492 (2011). inflammation of the central nervous system. Nat. Med. & Yong, V. W. Pathophysiology of the brain extracellular
114. Campbell, G. R. et al. Clonally expanded mitochondrial 13, 1483–1489 (2007). matrix: a new target for remyelination. Nat. Rev.
DNA deletions within the choroid plexus in multiple This study implicates ASIC1 as a potential Neurosci. 14, 722–729 (2013).
sclerosis. Acta Neuropathol. 124, 209–220 (2012). therapeutic target for progressive MS. 162. Stoffels, J. M. et al. Fibronectin aggregation in
115. Tranah, G. J. et al. Mitochondrial DNA sequence 139. Vergo, S. et al. Acid-sensing ion channel 1 is involved multiple sclerosis lesions impairs remyelination. Brain
variation in multiple sclerosis. Neurology 85, in both axonal injury and demyelination in multiple 136, 116–131 (2013).
325–330 (2015). sclerosis and its animal model. Brain 134, 571–584 163. Tepavcevic, V. et al. Early netrin-1 expression impairs
116. Witte, M. E., Geurts, J. J., de Vries, H. E., van der Valk, (2011). central nervous system remyelination. Ann. Neurol.
P. & van Horssen, J. Mitochondrial dysfunction: 140. Arun, T. et al. Targeting ASIC1 in primary progressive 76, 252–268 (2014).
a potential link between neuroinflammation and multiple sclerosis: evidence of neuroprotection with 164. Mi, S. et al. LINGO-1 negatively regulates myelination
neurodegeneration? Mitochondrion 10, 411–418 amiloride. Brain 136, 106–115 (2013). by oligodendrocytes. Nat. Neurosci. 8, 745–751
(2010). 141. Hundehege, P. et al. Targeting voltage-dependent (2005).
117. Lucchinetti, C. et al. Heterogeneity of multiple calcium channels with pregabalin exerts a direct This study identifies LINGO1, which is now being
sclerosis lesions: implications for the pathogenesis neuroprotective effect in an animal model of multiple targeted with the antibody opicinumab to enhance
of demyelination. Ann. Neurol. 47, 707–717 (2000). sclerosis. Neurosignals 26, 77–93 (2018). remyelination.

Nature Reviews | Drug Discovery


Reviews

165. Mi, S. et al. LINGO-1 antagonist promotes spinal cord of estrogen receptors. J. Neurosci. 39, 2184–2194 sclerosis. S.F. is grateful for grant support from the Medical
remyelination and axonal integrity in MOG-induced (2019). Faculty of Ruhr-University Bochum (FoRUM). V.W.Y is grateful
experimental autoimmune encephalomyelitis. 176. Deshmukh, V. A. et al. A regenerative approach to the for operating grant support from the Canadian Institutes of
Nat. Med. 13, 1228–1233 (2007). treatment of multiple sclerosis. Nature 502, 327–332 Health Research, the Multiple Sclerosis Society of Canada
166. Tran, J. Q. et al. Randomized phase I trials of the safety/ (2013). and the Alberta Innovates – Health Solutions CRIO Team
tolerability of anti-LINGO-1 monoclonal antibody 177. Moss, B. P., Rensel, M. R. & Hersh, C. M. Wellness program.
BIIB033. Neurol. Neuroimmunol. Neuroinflamm. 1, e18 and the role of comorbidities in multiple sclerosis.
(2014). Neurotherapeutics 14, 999–1017 (2017). Author contributions
167. Cadavid, D. et al. Safety and efficacy of opicinumab 178. Ontaneda, D., Fox, R. J. & Chataway, J. Clinical All authors contributed equally to the preparation of this
in acute optic neuritis (RENEW): a randomised, trials in progressive multiple sclerosis: lessons learned manuscript.
placebo-controlled, phase 2 trial. Lancet Neurol. 16, and future perspectives. Lancet Neurol. 14, 208–223
189–199 (2017). (2015).
Competing interests
168. Petrillo, J. et al. Initial impairment and recovery of An important and comprehensive discussion on
S.F. and V.W.Y. have filed provisional patent applications at
vision-related functioning in participants with acute optimal trial design in progressive MS.
the US Food and Drug Administration (US patent application
optic neuritis from the RENEW trial of opicinumab. 179. Stys, P. K., Zamponi, G. W., van Minnen, J. &
no. 62/412,555, entitled ‘Combination Therapy with
J. Neuroophthalmol. 39, 153–160 (2019). Geurts, J. J. Will the real multiple sclerosis please
Minocycline and Hydroxychloroquine for the Treatment of
169. Cadavid, D. et al. Efficacy analysis of opicinumab in stand up? Nat. Rev. Neurosci. 13, 507–514 (2012).
Multiple Sclerosis (MS)’ and US patent application no.
relapsing multiple sclerosis: the phase 2b SYNERGY 180. Lodygin, D. et al. β-Synuclein-reactive T cells induce
62/412,534, entitled ‘Treatment for Progressive Multiple
trial. ECTRIMS Online Library 147038, 192 (2016). autoimmune CNS grey matter degeneration. Nature
Sclerosis’). S.F. has received travel support from Biogen Idec
170. Gregg, C. et al. White matter plasticity and enhanced 566, 503–508 (2019).
and Genzyme, speaker’s honoraria from Novartis, board hon-
remyelination in the maternal CNS. J. Neurosci. 27, 181. Beard, J. L., Wiesinger, J. A. & Connor, J. R. Pre- and
oraria from Celgene and grant support from Novartis, unre-
1812–1823 (2007). postweaning iron deficiency alters myelination in
lated to this article. J.R.P. declares no competing interests.
171. Zhornitsky, S., Johnson, T. A., Metz, L. M., Weiss, S. Sprague-Dawley rats. Dev. Neurosci. 25, 308–315
R.G. has received speaker’s and board honoraria from Baxter,
& Yong, V. W. Prolactin in combination with interferon- (2003).
Bayer Schering, Biogen Idec, CLB Behring, Genzyme, Merck
beta reduces disease severity in an animal model of 182. Lange, S. J. & Que, L. Jr. Oxygen activating nonheme
Serono, Novartis, Stendhal, Talecris and Teva. His department
multiple sclerosis. J. Neuroinflammation 12, 55 (2015). iron enzymes. Curr. Opin. Chem. Biol. 2, 159–172
has received grant support from Bayer Schering, BiogenIdec,
172. Tourbah, A. et al. MD1003 (high-dose biotin) for (1998).
Genzyme, Merck Serono, Novartis and Teva. All of R.G.’s dec-
the treatment of progressive multiple sclerosis: 183. Todorich, B., Pasquini, J. M., Garcia, C. I., Paez, P. M.
larations are unrelated to the content of this article. V.W.Y.
a randomised, double-blind, placebo-controlled & Connor, J. R. Oligodendrocytes and myelination:
has received speaker’s and advisory board honoraria from
study. Mult. Scler. 22, 1719–1731 (2016). the role of iron. Glia 57, 467–478 (2009).
Biogen, EMD Serono, Roche, Novartis, Sanofi-Genzyme and
173. Mei, F. et al. Micropillar arrays as a high-throughput 184. de los Monteros, A. E. et al. Dietary iron and the
Teva, and educational grants from Biogen, EMD Serono,
screening platform for therapeutics in multiple integrity of the developing rat brain: a study with
Roche, Novartis, Sanofi-Genzyme and Teva.
sclerosis. Nat. Med. 20, 954–960 (2014). the artificially-reared rat pup. Cell. Mol. Biol. 46,
This high-throughput study identified clemastine 501–515 (2000).
as a potent remyelinating agent. 185. Schulz, K., Kroner, A. & David, S. Iron efflux from Publisher’s note
174. Green, A. J. et al. Clemastine fumarate as a astrocytes plays a role in remyelination. J. Neurosci. Springer Nature remains neutral with regard to jurisdictional
remyelinating therapy for multiple sclerosis 32, 4841–4847 (2012). claims in published maps and institutional affiliations.
(ReBUILD): a randomised, controlled, double-blind,
crossover trial. Lancet 390, 2481–2489 (2017). Acknowledgements Related links
175. Rankin, K. A. et al. Selective estrogen receptor The authors acknowledge the many trainees and collabora- ClinicalTrials.gov: https://clinicaltrials.gov
modulators enhance CNS remyelination independent tors who have contributed to our knowledge on multiple

www.nature.com/nrd

You might also like