Hauser 2020

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

ARTICLE IN PRESS

REVIEW

Treatment of Multiple Sclerosis: A Review


Stephen L. Hauser, MD, Bruce A.C. Cree, MD, PhD, MAS
UCSF Weill Institute for Neurosciences and Department of Neurology, University of California, San Francisco.

ABSTRACT

Multiple sclerosis (MS) is an autoimmune demyelinating and neurodegenerative disease of the central ner-
vous system, and the leading cause of nontraumatic neurological disability in young adults. Effective man-
agement requires a multifaceted approach to control acute attacks, manage progressive worsening, and
remediate bothersome or disabling symptoms associated with this illness. Remarkable advances in treat-
ment of all forms of MS, and especially for relapsing disease, have favorably changed the long-term out-
look for many patients. There also has been a conceptual shift in understanding the immune pathology of
MS, away from a purely T-cell-mediated model to recognition that B cells have a key role in pathogenesis.
The emergence of higher-efficacy drugs requiring less frequent administration have made these preferred
options in terms of tolerability and adherence. Many experts now recommend use of these as first-line
treatment for many patients with early disease, before permanent disability is evident.
Ó 2020 Elsevier Inc. All rights reserved.  The American Journal of Medicine (2020) 000:1−11

KEYWORDS: B-cell therapy; MS therapy; Multiple sclerosis

INTRODUCTION of neurological dysfunction followed by partial, complete, or


The autoimmune disease multiple sclerosis (MS) is the lead- no remission. Over time, relapses usually decrease in fre-
ing cause of nontraumatic neurological disability arising in quency but a gradual worsening often supervenes, resulting in
young adults.1,2 MS is characterized by 2 pathological hall- uninterrupted progression (termed secondary progressive MS
marks: 1) inflammation with demyelination, and 2) astroglial [SPMS]) (Figure 1).3 Less than 10% of patients with MS
proliferation (gliosis) and neurodegeneration. Tissue damage experience progression from onset, a category termed primary
in MS is restricted to the central nervous system (CNS), spar- progressive MS (PPMS).3 Despite these distinctions, all clini-
ing the peripheral nervous system. Clinically, MS can follow cal forms of MS appear to reflect the same underlying disease
2 paths: relapsing or progressive. Most commonly, onset is a process. And although inflammation is typically associated
relapsing form of MS (RMS), manifested as discrete episodes with relapses, and neurodegeneration with progression, it is
now recognized that both pathologies are present in essen-
Funding: This work was supported by grants from the National Insti- tially all patients across the entire disease continuum.
tute of Neurological Disorders and Stroke (R35NS111644), the National MS is a global problem, and its prevalence is on the
Multiple Sclerosis Society (RR 2005-A-13), and the Valhalla Foundation. rise.4 The prevalence is highest in North America, Western
Conflict of Interest: Outside of the submitted work, BACC reports Europe, and Australasia (>100 cases per 100,000 popula-
personal fees for consulting from Akili, Alexion, Atara, Biogen, EMD
Serono, Novartis, Sanofi, and TG Therapeutics. Outside of the submitted
tion), and lowest in countries centered around the equator
work, SLH reports stock options received for serving on the Board of (<30 cases per 100,000 population).4 In the United States, a
Directors for Neurona; Scientific Advisory Board for Alector, Annexon, recent study estimated that nearly 1 million individuals are
Bionure, and Molecular Stethoscope. He has also received nonfinancial affected. In RMS, women are affected nearly 3 times more
support (travel reimbursement and writing support for anti-CD20-related often than men, and the mean age of onset is ~30 years,
meetings) from F. Hoffmann-La Roche Ltd and Novartis AG.
Authorship: Both authors contributed equally to the literature search,
whereas in PPMS the rates of men and women affected are
figures, study design, data collection, data analysis, data interpretation, similar and the mean age of onset is ~40 years.5-7
and writing of this manuscript. The development of increasingly effective therapies for
Requests for reprints should be addressed to Stephen L. Hauser, MD, RMS, and partially effective therapy for PPMS and SPMS,
UCSF Weill Institute for Neurosciences and Department of Neurology, represents a profound success that has substantially
University of California, San Francisco, 675 Nelson Rising Lane, Suite
NS-217, San Francisco, CA 94143-3208.
improved prospects for lives free from disability. For
E-mail address: stephen.hauser@ucsf.edu patients with RMS, the mean time to development of SPMS

0002-9343/© 2020 Elsevier Inc. All rights reserved.


https://doi.org/10.1016/j.amjmed.2020.05.049
ARTICLE IN PRESS
2 The American Journal of Medicine, Vol 000, No 000, && 2020

was historically estimated at approximately 19 years after contribute to “dissemination in space.” Although MS pla-
onset, but in the treatment era has been lengthened substan- ques can involve subcortical white matter, lesions in this
tially. On highly effective therapy, relapses are markedly location are considered nondiagnostic because other pathol-
reduced or eliminated. However, control of RMS has ogies are associated with similar lesions.
uncovered a relapse-independent “silent” progression that Lumbar puncture is helpful, especially in uncertain cases
was previously obscured by attacks and remissions in and in all cases of suspected PPMS. CSF abnormalities
RMS.8,9 This recognition has also led to an increasing reli- include a mononuclear cell pleocytosis and an increased
ance on highly effective therapies level of intrathecally synthesized
early in the course of MS in order CLINICAL SIGNIFICANCE immunoglobulin G. Oligoclonal
to maximally control both relapses bands reflect the products of a
and progression. In this review we  Multiple sclerosis (MS), a common highly focused immune response by
summarize recent advances in MS autoimmune demyelinating disease of activated B cells in the CNS. The
treatment and speculate on future the central nervous system, has 2 abnormal intrathecal synthesis of
directions. major components: inflammation caus- gamma globulins, measured by an
ing episodic attacks, and neurodegen- elevated immunoglobulin G index
DIAGNOSIS eration responsible for progressive or 2 or more discrete oligoclonal
bands not present in a paired serum
worsening.
Clinical Manifestations  Recently developed MS therapies are sample, is present in >90% of MS
MS symptoms vary according to patients. Elevated intrathecal anti-
highly effective in preventing attacks body production can also be used to
location and severity of lesions and can partially protect against pro- fulfill “dissemination in time” crite-
occurring within the CNS. Clinical gression.
features of RMS may present ria in patients presenting with their
 Many experts now recommend the first clinical manifestation of MS.
acutely or subacutely over hours to
days, sometimes followed by grad-
use of these therapies early in the Although sensitive, elevated CSF
ual spontaneous remission over disease course to protect against antibody production is not specific
weeks to months. Conversely, late disability. for MS, and also occurs with CNS
PPMS is characterized by slowly infections. More than 50 cells/mm3
progressive symptoms from onset. are rare in MS, and polymorphonu-
Table 1 summarizes common clinical and laboratory fea- clear leukocytes, eosinophils, or a markedly elevated total
tures of MS. Symptoms may be severe at onset or begin protein level should call the diagnosis into question.
insidiously, sometimes unnoticed for months or years. Other useful tests include evoked potentials to assess
Once the patient seeks medical attention, and if MS is sus- nerve conduction in CNS pathways, and retinal imaging by
pected, prompt referral to a specialist is indicated. optical coherence tomography.

Investigation ADVANCES IN TREATMENT


Diagnosis requires objective evidence of inflammatory Current management strategies are focused on treating
CNS injury, and often, additional details of dissemination acute attacks, ameliorating symptoms, and reducing bio-
of the disease process “in space and time,” that is, affecting logic activity through disease-modifying therapies. The
more than one CNS location with evolution over time approach to treating acute attacks and symptom-based man-
(Table 2).10 Symptoms must last for >24 hours and occur agement is summarized in the Appendix (available online).
as distinct episodes separated by at least 1 month. The main
tests used to support diagnoses are magnetic resonance Disease-Modifying Therapies
imaging (MRI) and cerebrospinal fluid (CSF) analysis. Disease-modifying therapies modify the course of MS
In most patients an abnormal MRI is observed.11 Leak- through suppression or modulation of immune function.
age of intravenous gadolinium is caused by breakdown in They exert anti-inflammatory activity primarily in the
the blood−brain barrier that occurs early in the develop- relapsing phase of MS; they reduce the rate of relapses,
ment of an MS lesion and is a marker of acute inflamma- reduce accumulation of MRI lesions, and stabilize, delay,
tion. Gadolinium enhancement typically persists for <1 and in some cases modestly improve, disability.
month; however, the residual MS plaque remains visible The first approved therapies, the interferons and glatir-
indefinitely as a focal area of hyperintensity (a lesion) on amer acetate, were fortuitous discoveries. These well-toler-
fluid-attenuated inversion recovery or T2-weighted MRI ated drugs modestly reduce the frequency of MS relapses
scans. A periventricular (adjacent to the ventricles) distribu- and soon became commonly prescribed.12 Preclinical studies
tion of lesions, indicative of perivenular inflammation, is in experimental autoimmune encephalomyelitis, an animal
characteristic. Lesions in juxtacortical (adjacent to the cere- model of MS, advanced our understanding of crucial steps in
bral cortex) white matter, infratentorial white matter, and the pathogenesis of CNS autoimmune diseases, specifically,
within the spinal cord are also suggestive of MS and early peripheral expansion of autoreactive immune cells in
ARTICLE IN PRESS
Hauser and Cree Treatment of Multiple Sclerosis 3

Figure 1 The new natural history of multiple sclerosis (MS). The top half of the figure illustrates the natural history of
relapse-onset MS in the pretreatment era. During the relapsing phase, disability accumulation was thought to result from
incomplete recovery from relapses, until relapse-independent disability, designated SPMS, supervened. The “new” natural
history of MS in the current treatment era is shown in the bottom half. With use of highly effective therapies, attacks are
abolished in most patients, but insidious progression independent of relapse activity, termed “silent progression,” is now
evident during the relapsing phase.
CIS = clinically isolated syndrome; EDSS = extended disability status score; SPMS = secondary progressive multiple
sclerosis.

secondary lymphoid organs, subsequent infiltration of acti- monoclonal antibody rituximab and followed by the more
vated cells into the CNS, and generation of inflammatory successful development of ocrelizumab and ofatumumab.
lesions in white matter.13,14 These studies showed that T cells Anti-CD20-mediated B-cell depletion demonstrated a high
play a critical role in experimental autoimmune encephalo- level of success in limiting new relapses and silent progres-
myelitis;13 however, clinical trials with purely T-cell-based sion in RMS,19-21 and in reducing disability progression in
treatment approaches were ineffective in RMS.15,16 In con- PPMS.22 Thus, the past decade has seen a shift in the concep-
trast, strategies that inhibited lymphocyte access to the CNS tual framework of MS pathophysiology, from the earlier
by blocking adhesion (natalizumab) or sequestering lympho- model that MS is a T-cell-mediated autoimmune disorder, to
cytes in primary lymphoid organs (the sphingosine-1-phos- the understanding of a central role for B cells.23 Although
phate [S1P] receptor modulators fingolimod, siponimod, and substantial progress has been made against RMS, the devel-
ozanimod) were found to be effective in both MS and experi- opment of highly effective therapies against progressive MS
mental autoimmune encephalomyelitis. A major advance remains an unmet need (Figure 2).24-26
was the development of disease models that more closely
replicated the pattern of tissue damage in MS, leading to a
new appreciation of the importance of humoral immunity in APPROACH TO TREATING PATIENTS WITH MS
MS pathogenesis.17,18 This paved the way for clinical trials Table 319,22,27-46 provides an overview of the characteristics
of B-cell-depleting therapies, initially with the anti-CD20 and pivotal data associated with approved agents, stratified
ARTICLE IN PRESS
4 The American Journal of Medicine, Vol 000, No 000, && 2020

Table 1 Approach to the Diagnosis of Multiple Sclerosis


Symptoms Tests
 Sensory loss or paresthesias (tingling) Magnetic Resonance Imaging (MRI)
 Unilateral painful visual loss (optic neuritis)  Multiple lesions
 Limb weakness (hyperreflexia, Babinski sign)  White matter
 Facial weakness resembling Bell’s palsy  Cerebral hemispheres, brainstem, spinal cord
 Visual blurring due to diplopia  Recent lesions enhance with gadolinium
 Ataxia  Lesions perpendicular to ventricular surface and juxtacortical
 Vertigo Cerebrospinal fluid
 Paroxysmal symptoms  Oligoclonal immunoglobulin
○ Lhermitte’s symptom (electric shock-like sensations evoked  Modest inflammation (mononuclear cells)
by neck flexion)
Evoked potentials
○ Trigeminal neuralgia, hemifacial spasm, and glossophar-
 Detect conduction delay in visual, auditory, and sensory
yngeal neuralgia
pathways
○ Facial myokymia (rapid flickering contractions of the facial
muscles)
 Heat sensitivity
 Bladder dysfunction
 Pain
 Cognitive dysfunction, usually mild, “brain fog,” difficulty with
multitasking
 Sexual dysfunction
 Fatigue
Uncommon symptoms (red flags)
 Seizure
 Dementia
 Movement disorder

according to frequency of use and perceived level of effi- epidemiologic expectations. Postmarketing studies are gen-
cacy. Given this large array of available therapies, it may erally consistent with the clinical trials, although serious
be prudent for clinicians to become conversant with a few herpes virus infections have become a recognized compli-
agents. As such, the following section is focused on detail- cation.50 Rituximab is a chimeric anti-CD20 monoclonal
ing MS disease-modifying therapies that are widely used in antibody that appears to be similarly effective against RMS
clinical practice. and PPMS in preliminary trials and real-world experience,
and is widely used despite never having received approval
from any regulatory body. Ofatumumab27 is a fully human
Treating RMS anti-CD20 monoclonal antibody administered by monthly
Frequently used disease-modifying therapies for RMS. Tag- subcutaneous injection at home and has an efficacy profile
gedPOcrelizumab, a humanized monoclonal antibody against similar to ocrelizumab, with a high degree of safety in the
the CD20 molecule on the surface of mature B cells,47 has phase 3 trials for RMS; ofatumumab is expected to become
been widely used since its approval in 2017. Ocrelizumab available in 2020.
is highly effective against relapses and silent progression in Natalizumab, a humanized monoclonal antibody, is an
RMS patients, and has substantial benefits in halting devel- inhibitor of the a4b1 integrin, an adhesion molecule
opment of new white matter lesions detected by MRI. Ocre- expressed on the surface of lymphocytes and involved in
lizumab selectively depletes CD20-expressing B cells, transmigration across endothelia into the CNS.51 Natalizu-
preserving preexisting humoral immunity and the capacity mab is administered as an intravenous infusion once every
for B-cell reconstitution. B-cell depletion is associated with 4 weeks. Natalizumab is highly effective in reducing relap-
potent interruption in B-cell trafficking from the periphery ses and slowing disease progression in RMS patients com-
to the CNS, reduced B-cell antigen presentation to T cells, pared with placebo or interferon beta (IFN-b)-1a,28,52
modulation of proinflammatory cytokine secretion by B benefits sustained over a longer term in real-world stud-
cells, and reduced activation and differentiation to immuno- ies.53 Natalizumab is generally well tolerated; however,
globulin-secreting plasmablasts.48,49 Ocrelizumab is admin- long-term treatment carries risk of progressive multifocal
istered as an intravenous infusion every 24 weeks. Initial leukoencephalopathy (an opportunistic brain infection
findings from the phase 3 trials indicated a possible low caused by the John Cunningham virus), occurring in ~0.4%
risk of increased malignancies, including breast cancer, of natalizumab-treated patients.54 Progressive multifocal
although longer follow-up revealed cancer rates identical to leukoencephalopathy incidence increases with natalizumab
ARTICLE IN PRESS
Hauser and Cree Treatment of Multiple Sclerosis 5

Table 2 Diagnostic Criteria for Multiple Sclerosis10  Evidence for dissemination in space in the spinal cord
based on ≥2 T2+ lesions in the cord
Criteria for Diagnosis of Multiple Sclerosis in Patients with an  Positive cerebrospinal fluid (isoelectric focusing evidence
Attack at Onset
of oligoclonal bands or elevated immunoglobulin G index)
≥2 attacks; objective clinical evidence of ≥2 lesions or objec-
tive clinical evidence of 1 lesion with reasonable historical CNS = central nervous system; MRI = magnetic resonance imaging;
evidence of a prior attack MS = multiple sclerosis.
≥2 attacks; objective clinical evidence of 1 lesion
Dissemination in space, demonstrated by:
 ≥1 T2 lesion on MRI in at least 2 of 4 MS-typical regions
exposure and this risk can be stratified according to the lev-
of the CNS (periventricular, juxtacortical, infratentorial,
els of John Cunningham virus antibodies in serum. In anti-
or
body-negative patients, progressive multifocal
spinal cord)
OR leukoencephalopathy risk is minimal, <1:10,000. Con-
 Await a further clinical attack implicating a different CNS versely, in antibody-positive patients, risk may be as high
site as ≥1.1% annually.54 Thus, natalizumab is generally rec-
1 attack; objective clinical evidence of ≥2 lesions ommended only for antibody-negative patients. Extending
Dissemination in time, demonstrated by: the dosing interval from 4 to 6 weeks appears to reduce the
 Simultaneous presence of asymptomatic gadolinium- risk of progressive multifocal leukoencephalopathy by
enhancing and nonenhancing lesions at any time more than 90% in seropositive patients, providing a useful
OR strategy in reducing risk.55 A disadvantage of natalizumab
 A new T2 or gadolinium-enhancing lesion(s) on follow-up is that discontinuation can trigger “rebound” disease activ-
MRI, irrespective of its timing with reference to a baseline ity, a problem that may be encountered in patients who are
scan noncompliant, or John Cunningham virus seroconvert, or
OR
who discontinue prior to attempting to conceive.
 Await a second clinical attack
Fingolimod was the first oral therapy approved for RMS.
1 attack; objective clinical evidence of 1 lesion (clinically iso-
It is an S1P inhibitor that prevents egress of lymphocytes
lated syndrome)
Dissemination in space and time, demonstrated by: from secondary lymphoid organs, blocking infiltration of
For dissemination in space autoreactive lymphocytes into the CNS. Fingolimod is well
 ≥1 T2 lesion in at least 2 of 4 MS-typical regions of the tolerated, although adverse events include mild abnormali-
CNS (periventricular, juxtacortical, infratentorial, or spi- ties on routine laboratory evaluation (eg, elevated liver
nal cord) function tests or lymphopenia).29,30,56,57 Heart block and
OR bradycardia can also occur when therapy is initiated, thus, a
 Await a second clinical attack implicating a different CNS 6-hour observation period (including electrocardiogram
site monitoring) is recommended for all patients receiving their
AND first dose.58 Other side effects include macular edema and,
For dissemination in time rarely, disseminated varicella-zoster virus, cryptococcal
 Simultaneous presence of asymptomatic gadolinium- infections, and progressive multifocal leukoencephalop-
enhancing and nonenhancing lesions at any time athy; prior to initiating therapy, an ophthalmic examination
OR
and varicella-zoster virus vaccination for seronegative indi-
 A new T2 or gadolinium-enhancing lesion(s) on follow-up
viduals are indicated. Ozanimod, a recently approved selec-
MRI, irrespective of its timing with reference to a baseline
scan tive S1P receptor modulator, was also shown to be effective
OR in RMS, with favorable safety and tolerability.31,32
 Await a second clinical attack Dimethyl fumarate33,34 exerts anti-inflammatory and
Criteria for diagnosis of MS in patients with a disease course cytoprotective effects through activation of the nuclear fac-
characterized by progression from onset (primary progressive tor (erythroid-derived 2)−like 2 (Nrf2) pathway and Nrf2-
MS) independent pathways.59,60 Dimethyl fumarate is generally
Insidious neurologic progression suggestive of primary pro- well tolerated, but treatment has been linked with some pro-
gressive multiple sclerosis gressive multifocal leukoencephalopathy risk.61 Most of
1 year of disease progression (retrospectively or prospec- these cases were lymphopenic, thus, monitoring for lym-
tively determined) phopenia every 6-12 months is recommended. Diroximel
AND
fumarate was recently approved and, like dimethyl fuma-
2 of the 3 following criteria:
rate, is metabolized to mono-methyl fumarate.62
 Evidence for dissemination in space in the brain based on
Teriflunomide35 is the active metabolite of leflunomide,
≥1 T2+ lesions in the MS-characteristic periventricular, jux-
tacortical, or infratentorial regions an immune suppressant medication used in rheumatoid
arthritis. Teriflunomide inhibits dihydroorotate dehydroge-
nase, an enzyme involved in pyrimidine synthesis. Teriflu-
nomide inhibits proliferation of activated lymphocytes
presumed to be autoreactive. Boxed warnings include risk
ARTICLE IN PRESS
6 The American Journal of Medicine, Vol 000, No 000, && 2020

Figure 2 Five distinctive pathologies likely to contribute to progressive multiple sclerosis (MS). (1) Classic inflamma-
tory white matter plaques are typically associated with relapses but also occur in patients who progress gradually without
acute attacks. Hallmarks are perivenous inflammation dominated by lymphocytes and macrophages (detected by MRI as
gadolinium enhancement indicating blood−brain barrier disruption); demyelination associated with activated microglia;
sharply demarcated plaques with glial scarring; and axonal loss with secondary retrograde and anterograde degeneration.
(2) B-cell rich lymphoid aggregates in the meninges, often in deep sulci, with underlying cortical demyelination and neu-
ronal loss. (3) Slowly enlarging lesions due to gradual concentric expansion of chronic plaques, characterized by a rim of
activated microglia at the leading edge, astrocytosis, stressed oligodendrocytes, and progressive axonal injury. (4) Wide-
spread diffuse microglial inflammation and astrogliosis throughout the CNS white matter, associated with decreased mye-
lin density and ongoing axonal damage. (5) Age-related neurodegeneration. For a more detailed discussion, readers may
consult references24-26.
CNS = central nervous system; MRI = magnetic resonance imaging.

of hepatotoxicity and teratogenesis. Common adverse laboratory evaluation, as well as injection-site reactions
events include headache, diarrhea, nausea, alopecia, and with subcutaneous administration.36,66 Side effects can usu-
increase in hepatic alanine transferase. When necessary, ter- ally be managed with nonsteroidal anti-inflammatory medi-
iflunomide can be rapidly eliminated by ingestion of chole- cations.
styramine. Glatiramer acetate is the acetate salt of a mixture of ran-
IFN-b is a class I interferon whose mechanism of action dom polypeptides composed of 4 amino acids. Its mecha-
likely involves immunomodulation through downregulating nism of action may involve favorably altering the balance
expression of major histocompatibility complex molecules between proinflammatory and regulatory cytokines.38,67,68
on antigen-presenting cells, decreasing proinflammatory It modestly reduces relapse rates and some disease severity
and increasing anti-inflammatory cytokines, inhibiting T- measures, and can be considered as an equally effective
cell proliferation, and blocking trafficking of inflammatory alternative to IFN-b in RMS.38,67,69 Common adverse
cells to the CNS.63 IFN-b modestly reduces the relapse rate events include injection-site reactions, flushing, chest tight-
and MRI measures of disease activity and slows accumula- ness, dyspnea, palpitations, anxiety after injection, and less
tion of disability.36,37,64-66 Common adverse events include commonly, lipoatrophy that can rarely be disfiguring and
flu-like symptoms and mild abnormalities on routine require treatment cessation.38,67
Hauser and Cree
Table 3 Summary of Approved Disease-Modifying Therapies for Multiple Sclerosis
Drug Name Mechanism of Action Indication Route of and Pivotal Efficacy Data Common Adverse Events
Frequency of
Administration
Highly effective
Ocrelizumab19,22 Anti-CD20 mAb RMS and PPMS (first IV infusion, every 6 RMS: Relative reduction in ARR RMS: Infusion-related reaction, nasopharyngitis,

Treatment of Multiple Sclerosis


line) months compared with IFN-b-1a: 47% upper respiratory tract infection, headache, and
PPMS: Relative reduction in 12- urinary tract infection PPMS: Infusion-related
week CDP compared with pla- reaction, upper respiratory tract infection, and
cebo: 24% oral herpes infection
Ofatumumab27 Anti-CD20 mAb RMS (first line) SC injection, every Relative reduction in ARR com- Injection-related reaction, nasopharyngitis,
4 weeks pared with teriflunomide: 54% headache, upper respiratory tract infection, and
urinary tract infection
Natalizumab28 a4b1 integrin inhibitor RMS (second line) IV infusion, every 4 Relative reduction in ARR com- Fatigue and allergic reaction

ARTICLE IN PRESS
weeks pared with placebo: 68%
Relative reduction in sustained
disease progression compared
with placebo: 42%
Alemtuzumab40-42 Anti-CD52 mAb RMS (first line) IV infusion, once Relative reduction in ARR com- Headache, rash, nausea, and pyrexia
daily pared with placebo: 49%-69%
Mitoxantrone39 DNA intercalator RMS, SPMS IV infusion, every Relative reduction in relapses Dose-related cardiomyopathy, promyelocytic
(second or third month or 3 compared with placebo: 61% leukemia
line) months
Moderately effective
Fingolimod29,30 Sphingosine-1- phosphate RMS (second line) Oral, once daily Relative reduction in ARR com- Bradycardia, atrioventricular conduction block,
inhibitor pared with placebo: 48%-60% macular edema, elevated liver-enzyme levels,
and mild hypertension
Siponimod44 Sphingosine 1-phosphate CIS, RMS, active Oral, once daily Relative reduction in 12-week Headache, nasopharyngitis, urinary tract infec-
receptor modulator SPMS (first Line) CDP compared with placebo: tion, and falls
21%
Ozanimod31,32 Sphingosine 1-phosphate CIS, RMS, active Oral, once daily Relative reduction in ARR com- Headache and elevated liver aminotransferase
receptor modulator SPMS pared with placebo: 48%
Dimethyl fumarate Nuclear factor (erythroid- RMS (first line) Oral, twice daily Relative reduction in ARR com- Flushing, diarrhea, nausea, upper abdominal pain,
and diroximel derived 2)−like 2 path- pared with placebo: 48%-53% decreased lymphocyte counts, and elevated
fumarate33,34 way inhibitor liver aminotransferase
Cladribine43 Not fully known RMS (second or Oral, 4-5 days over Relative reduction in ARR com- Headache, lymphocytopenia, nasopharyngitis,
third line) 2-week treatment pared with placebo: 55%-58% upper respiratory tract infection, and nausea
courses
Modestly effective
Teriflunomide35 Dihydroorotate dehydro- RMS (first line) Oral, once daily Relative reduction in ARR com- Nasopharyngitis, headache, diarrhea, and alanine
genase inhibitor pared with placebo: 32%-36% aminotransferase increase
Not fully known RMS (first line) Injection-site reactions

7
ARTICLE IN PRESS
8 The American Journal of Medicine, Vol 000, No 000, && 2020

Flu-like symptoms, muscle aches, asthenia, chills,

ARR = annualized relapse rate; CDP = confirmed disability progression; CIS = clinically isolated syndrome; IFN-b-1a = interferon beta 1a; IM = intramuscular; IV = intravenous; mAb = monoclonal antibody;
Infrequently used disease-modifying therapies for RMS.

Lymphopenia, flu-like symptoms, and injection-


Injection-site inflammation, flu-like symptoms,

Injection-site erythema, influenza-like illness,


These include mitoxantrone,39 alemtuzumab,40-42 and cla-
dribine.43 All are of proven benefit in RMS; however, seri-
ous treatment-emergent adverse events limit their use.
Finally, a growing body of evidence indicates that autolo-
gous hematopoietic stem cell transplantation can induce a
prolonged remission in many RMS patients. Randomized
rhinitis, and headache trials comparing this procedure with other highly effective

pyrexia, and headache


Common Adverse Events

therapies are now underway. For the time being, autologous


hematopoietic stem cell transplantation is recommended

site reactions
only for RMS patients who have exhausted all other reason-
able treatment options, and is not recommended for patients
and fever

with progressive MS because available data suggest that


progression often continues despite this treatment.70

PPMS = primary progressive multiple sclerosis; RMS = relapsing forms of multiple sclerosis; SC = subcutaneous; SPMS = secondary progressive multiple sclerosis. Treating Progressive MS
Relative reduction in ARR com-

Relative reduction in ARR com-

Relative reduction in ARR com-

Relative reduction in ARR com-

SPMS. Siponimod44 is a selective S1P modulator that is


CDP compared with placebo:
Relative reduction in 24-week

approved for relapsing forms of MS, including active


pared with placebo: 29%

pared with placebo: 33%

pared with placebo: 39%

pared with placebo: 31%

SPMS, meaning patients with SPMS who have had recent


Pivotal Efficacy Data

clinical relapses or evidence of new or enlarging MRI


lesions. Ocrelizumab, cladribine, and diroximel fumarate
can also be used for patients with active SPMS.

PPMS. Ocrelizumab is the only approved disease-modify-


37%

ing therapy for the treatment of PPMS. Dosing is the same


as for RMS. Ocrelizumab reduces progression of clinical
SC injection, 3 times

disability by approximately one-quarter, and improves


SC injection, every

SC injection, every
IM injection, once
SC injection, once
daily or 3 times

other clinical and MRI markers of inflammatory and degen-


Administration

erative disease activity in this population.22


Route of and
Frequency of

other day
2 weeks
weekly

weekly

weekly

Decision-Making for Treatment of MS


Given the increasingly complex landscape of therapeutics
for MS, the treatment algorithm in Figure 3 may aid in deci-
sion-making. In general, therapy should be initiated in
CIS and RMS (first

CIS and RMS (first

CIS and RMS (first

CIS and RMS (first

patients with a first attack who are at high risk for MS or in


patients diagnosed with RMS.10
Indication

Although historically, high-dose IFN-b-1a and glatir-


line)

line)

line)

line)

amer acetate were the preferred first-line options, we now


recommend the use of highly effective disease-modifying
therapies as first-line options for the majority of patients
with active MS. This approach supervenes the traditional
“treat to target” method in which a treatment of modest or
Mechanism of Action

moderate effectiveness is first used, and therapy advanced


Not fully known

Not fully known

Not fully known

Not fully known

to a more effective agent when breakthrough disease (evi-


dent clinically or by MRI) occurs. Observational studies
suggest that early use of high-efficacy therapy improves
long-term outcomes. We favor initiating therapy for many
patients with ocrelizumab or another anti-CD20 drug, or
with natalizumab in John Cunningham virus-negative
IFN-b-1a (Rebif)45

patients. Anti-CD20 therapies represent an attractive option


Table 3 (Continued)

(Betaseron)36

given their high level of efficacy, infrequent infusions or


(Plegridy)46
(Avonex)37

PegIFN-b-1a

injections, favorable safety profile, and absence of rebound


acetate38
Glatiramer

IFN-b-1b
IFN-b-1a

following discontinuation. Patients with PPMS with new or


Drug Name

enlarging MRI lesions should also be considered for ocreli-


zumab treatment. Switching therapies may be required in
the following situations: suboptimal response, experiencing
ARTICLE IN PRESS
Hauser and Cree Treatment of Multiple Sclerosis 9

Figure 3 A therapeutic algorithm for disease-modifying therapy use in multiple sclerosis (MS). A simplified MS treat-
ment algorithm based on the authors’ practice pattern. Several drugs with proven effectiveness for MS are not mentioned,
including: diroximel fumarate (presumably comparable with dimethyl fumerate); cladribine (reserved for MS refractory
to at least 2 other treatments); alemtuzumab and mitoxantrone (not generally recommended due to severe toxicities);
hematopoietic stem cell transplantation (reserved for severe treatment refractory disease unresponsive to high efficacy
treatments); ozanimod (approved but not yet commercially available).
DMT = disease-modifying therapy; JCV = John Cunningham virus; MRI = magnetic resonance imaging.

more than one relapse with active MRI scans in the prior ACKNOWLEDGMENT
year while on treatment, and safety issues including devel- The authors thank Natalie Nwkor for invaluable editorial
opment of persistent high-titer neutralizing antibodies in assistance, and Andrew Barnecut for superb work creating
patients receiving IFN-b. Discontinuation of disease-modi- figures and finalizing the manuscript.
fying therapies is required in cases of serious adverse events
that may be drug related and for many disease-modifying
therapies in women who become pregnant while on treat-
ment. Exceptions include glatiramer acetate that can be References
continued during pregnancy, and in some cases, prior use of 1. Cree BAC, Hauser SL. Multiple sclerosis. In: Jameson JL, Fauci AS,
ocrelizumab, alemtuzumab, and cladribine that have pro- Kasper DL, Hauser SL, Longo DL, Loscalzo J, eds. Harrison’s Princi-
ples of Internal Medicine, 20th ed New York, NY: McGraw-Hill Edu-
longed pharmacodynamic effects that persist after the drug cation; 2018.
has been eliminated. 2. Browne P, Chandraratna D, Angood C, et al. Atlas of Multiple Sclero-
sis 2013: a growing global problem with widespread inequity. Neurol-
ogy 2014;83(11):1022–4.
3. Lublin FD, Reingold SC, Cohen JA, et al. Defining the clinical course
of multiple sclerosis: the 2013 revisions. Neurology 2014;83(3):278–
CONCLUSION 86.
Spectacular progress has been made in the treatment of MS 4. Global, regional, and national burden of multiple sclerosis 1990-2016:
as a result of advances in understanding of the pathogenesis a systematic analysis for the Global Burden of Disease Study 2016.
and course of this disease. The development of highly Lancet Neurol 2019;18(3):269–85.
5. Kamm CP, Uitdehaag BM, Polman CH. Multiple sclerosis: current
effective therapies has produced near-complete control of
knowledge and future outlook. Eur Neurol 2014;72(3-4):132–41.
relapsing disease and focal brain inflammation. However, 6. Confavreux C, Vukusic S. Natural history of multiple sclerosis: a uni-
effective treatment of progression remains an unmet need fying concept. Brain 2006;129(Pt 3):606–16.
because current therapies confer only partial protection 7. Wallin MT, Culpepper WJ, Campbell JD, et al. The prevalence of MS
against the neurodegenerative component of MS. Although in the United States: a population-based estimate using health claims
natural history studies suggest that the long-term course of data. Neurology 2019;92(10):e1029–40.
8. Cree BAC, Hollenbach JA, Bove R, et al. Silent progression in disease
disease has been substantially improved in the treatment activity-free relapsing multiple sclerosis. Ann Neurol 2019;85(5):653–
era, further clinical and real-world assessments are needed 66.
to gather long-term efficacy and safety evidence for these 9. Kappos L, Butzkueven H, Wiendl H, et al. Greater sensitivity to multi-
therapies. Additional studies of the value of highly effective ple sclerosis disability worsening and progression events using a rov-
ing versus a fixed reference value in a prospective cohort study. Mult
agents for early treatment and determination of patients
Scler 2018;24(7):963–73.
who benefit most will also be essential as we seek to 10. Thompson AJ, Banwell BL, Barkhof F, et al. Diagnosis of multiple
achieve evidence-based and personalized approaches to MS sclerosis: 2017 revisions of the McDonald criteria. Lancet Neurol
treatment and management. 2018;17(2):162–73.
ARTICLE IN PRESS
10 The American Journal of Medicine, Vol 000, No 000, && 2020

11. Filippi M, Rocca MA, Ciccarelli O, et al. MRI criteria for the diagno- (RADIANCE): a multicentre, randomised, 24-month, phase 3 trial.
sis of multiple sclerosis: MAGNIMS consensus guidelines. Lancet Lancet Neurol 2019;18(11):1021–33.
Neurol 2016;15(3):292–303. 33. Fox RJ, Miller DH, Phillips JT, et al. Placebo-controlled phase 3 study
12. Galetta SL, Markowitz C, Lee AG. Immunomodulatory agents for the of oral BG-12 or glatiramer in multiple sclerosis. N Engl J Med
treatment of relapsing multiple sclerosis: a systematic review. Arch 2012;367(12):1087–97.
Intern Med 2002;162(19):2161–9. 34. Gold R, Kappos L, Arnold DL, et al. Placebo-controlled phase 3 study
13. Steinman L, Zamvil SS. How to successfully apply animal studies in of oral BG-12 for relapsing multiple sclerosis. N Engl J Med 2012;367
experimental allergic encephalomyelitis to research on multiple scle- (12):1098–107.
rosis. Ann Neurol 2006;60(1):12–21. 35. O’Connor P, Wolinsky JS, Confavreux C, et al. Randomized trial of
14. Martin R. Immunological aspects of experimental allergic encephalo- oral teriflunomide for relapsing multiple sclerosis. N Engl J Med
myelitis and multiple sclerosis and their application for new therapeu- 2011;365(14):1293–303.
tic strategies. J Neural Transm Suppl 1997;49:53–67. 36. Interferon beta-1b is effective in relapsing-remitting multiple sclero-
15. van Oosten BW, Lai M, Hodgkinson S, et al. Treatment of multiple sis. I. Clinical results of a multicenter, randomized, double-blind, pla-
sclerosis with the monoclonal anti-CD4 antibody cM-T412: results of cebo-controlled trial. The IFNB Multiple Sclerosis Study Group.
a randomized, double-blind, placebo-controlled, MR-monitored phase Neurology 1993;43(4):655–61.
II trial. Neurology 1997;49(2):351–7. 37. Jacobs LD, Cookfair DL, Rudick RA, et al. Intramuscular interferon
16. Weinshenker BG, Bass B, Karlik S, Ebers GC, Rice GP. An open trial beta-1a for disease progression in relapsing multiple sclerosis. The
of OKT3 in patients with multiple sclerosis. Neurology 1991;41 Multiple Sclerosis Collaborative Research Group (MSCRG). Ann
(7):1047–52. Neurol 1996;39(3):285–94.
17. Massacesi L, Genain CP, Lee-Parritz D, Letvin NL, Canfield D, 38. Johnson KP, Brooks BR, Cohen JA, et al. Copolymer 1 reduces
Hauser SL. Active and passively induced experimental autoimmune relapse rate and improves disability in relapsing-remitting multiple
encephalomyelitis in common marmosets: a new model for multiple sclerosis: results of a phase III multicenter, double-blind placebo-con-
sclerosis. Ann Neurol 1995;37(4):519–30. trolled trial. The Copolymer 1 Multiple Sclerosis Study Group. Neu-
18. Hauser SL. The Charcot Lecture | beating MS: a story of B cells, with rology 1995;45(7):1268–76.
twists and turns. Mult Scler 2015;21(1):8–21. 39. Goodin DS, Arnason BG, Coyle PK, Frohman EM, Paty DW. The use
19. Hauser SL, Bar-Or A, Comi G, et al. Ocrelizumab versus interferon of mitoxantrone (Novantrone) for the treatment of multiple sclerosis:
beta-1a in relapsing multiple sclerosis. N Engl J Med 2017;376 report of the Therapeutics and Technology Assessment Subcommittee
(3):221–34. of the American Academy of Neurology. Neurology 2003;61
20. Hauser SL, Waubant E, Arnold DL, et al. B-cell depletion with rituxi- (10):1332–8.
mab in relapsing-remitting multiple sclerosis. N Engl J Med 2008;358 40. Coles AJ, Compston DA, Selmaj KW, et al. Alemtuzumab vs. inter-
(7):676–88. feron beta-1a in early multiple sclerosis. N Engl J Med 2008;359
21. Kappos L, Wolinsky JS, Giovannoni G, et al. Contribution of relapse- (17):1786–801.
independent progression vs relapse-associated worsening to overall 41. Cohen JA, Coles AJ, Arnold DL, et al. Alemtuzumab versus interferon
confirmed disability accumulation in typical relapsing multiple sclero- beta 1a as first-line treatment for patients with relapsing-remitting
sis in a pooled analysis of 2 randomized clinical trials:[e-pub ahead of multiple sclerosis: a randomised controlled phase 3 trial. Lancet
print] JAMA Neurol 2020 June 8. https://doi.org/10.1001/jama- 2012;380(9856):1819–28.
neurol.2020.1568. 42. Coles AJ, Twyman CL, Arnold DL, et al. Alemtuzumab for patients
22. Montalban X, Hauser SL, Kappos L, et al. Ocrelizumab versus pla- with relapsing multiple sclerosis after disease-modifying therapy: a
cebo in primary progressive multiple sclerosis. N Engl J Med 2017; randomised controlled phase 3 trial. Lancet 2012;380(9856):1829–39.
376(3):209–20. 43. Giovannoni G, Comi G, Cook S, et al. A placebo-controlled trial of
23. Greenfield AL, Hauser SL. B-cell therapy for multiple sclerosis: enter- oral cladribine for relapsing multiple sclerosis. N Engl J Med
ing an era. Ann Neurol 2018;83(1):13–26. 2010;362(5):416–26.
24. Lassmann H. Pathogenic mechanisms associated with different clini- 44. Kappos L, Bar-Or A, Cree BAC, et al. Siponimod versus placebo in
cal courses of multiple sclerosis. Front Immunol 2018;9:3116. secondary progressive multiple sclerosis (EXPAND): a double-blind,
25. Frischer JM, Bramow S, Dal-Bianco A, et al. The relation between randomised, phase 3 study. Lancet 2018;391(10127):1263–73.
inflammation and neurodegeneration in multiple sclerosis brains. 45. Randomised double-blind placebo-controlled study of interferon beta-
Brain 2009;132(Pt 5):1175–89. 1a in relapsing/remitting multiple sclerosis. PRISMS (Prevention of
26. Haider L, Zrzavy T, Hametner S, et al. The topograpy of demyelin- Relapses and Disability by Interferon beta-1a Subcutaneously in Mul-
ation and neurodegeneration in the multiple sclerosis brain. Brain tiple Sclerosis) Study Group. Lancet 1998;352(9139):1498–504.
2016;139(Pt 3):807–15. 46. Calabresi PA, Kieseier BC, Arnold DL, et al. Pegylated interferon
27. Hauser SL, Bar-Or A, Cohen JA, et al. Ofatumumab versus terifluno- beta-1a for relapsing-remitting multiple sclerosis (ADVANCE): a
mide in multiple sclerosis. N Engl J Med 2020;383(6):546–57. randomised, phase 3, double-blind study. Lancet Neurol 2014;13
28. Polman CH, O’Connor PW, Havrdova E, et al. A randomized, pla- (7):657–65.
cebo-controlled trial of natalizumab for relapsing multiple sclerosis. N 47. Sorensen PS, Blinkenberg M. The potential role for ocrelizumab in the
Engl J Med 2006;354(9):899–910. treatment of multiple sclerosis: current evidence and future prospects.
29. Calabresi PA, Radue EW, Goodin D, et al. Safety and efficacy of fin- Ther Adv Neurol Disord 2016;9(1):44–52.
golimod in patients with relapsing-remitting multiple sclerosis (FREE- 48. Li R, Patterson KR, Bar-Or A. Reassessing B cell contributions in
DOMS II): a double-blind, randomised, placebo-controlled, phase 3 multiple sclerosis. Nat Immunol 2018;19(7):696–707.
trial. Lancet Neurol 2014;13(6):545–56. 49. Sabatino JJ Jr, Probstel AK, Zamvil SS. B cells in autoimmune and
30. Kappos L, Radue EW, O’Connor P, et al. A placebo-controlled trial of neurodegenerative central nervous system diseases. Nat Rev Neurosci
oral fingolimod in relapsing multiple sclerosis. N Engl J Med 2019;20(12):728–45.
2010;362(5):387–401. 50. Hauser SL, Kappos L, Montalban X, et al. Safety of ocrelizumab in mul-
31. Comi G, Kappos L, Selmaj KW, et al. Safety and efficacy of ozanimod tiple sclerosis: updated analysis in patients with relapsing and primary
versus interferon beta-1a in relapsing multiple sclerosis (SUNBEAM): progressive multiple sclerosis. Eur J Neurol 2019;26(suppl 1):239.
a multicentre, randomised, minimum 12-month, phase 3 trial. Lancet 51. Yednock TA, Cannon C, Fritz LC, Sanchez-Madrid F, Steinman L,
Neurol 2019;18(11):1009–20. Karin N. Prevention of experimental autoimmune encephalomyelitis
32. Cohen JA, Comi G, Selmaj KW, et al. Safety and efficacy of ozani- by antibodies against alpha 4 beta 1 integrin. Nature 1992;356
mod versus interferon beta-1a in relapsing multiple sclerosis (6364):63–6.
ARTICLE IN PRESS
Hauser and Cree Treatment of Multiple Sclerosis 11

52. Rudick RA, Stuart WH, Calabresi PA, et al. Natalizumab plus inter- with dimethyl fumarate in patients with relapsing-remitting multiple
feron beta-1a for relapsing multiple sclerosis. N Engl J Med 2006;354 sclerosis: results from the randomized, double-blind, phase III
(9):911–23. EVOLVE-MS-2 study. CNS Drugs 2020;34(2):185–96.
53. Myhr K-M, Sch€ uler S, Alstadhaug KB, et al. Long-term, real-world 63. Kieseier BC. The mechanism of action of interferon-beta in relapsing
effectiveness of natalizumab treatment in relapsing-remitting multiple multiple sclerosis. CNS Drugs 2011;25(6):491–502.
sclerosis (RRMS): data from ≥6 years in the TYSABRIÒ Observa- 64. Kappos L, Kuhle J, Multanen J, et al. Factors influencing long-term
tional Program (TOP) Nordic and global cohorts. Eur J Neurol outcomes in relapsing-remitting multiple sclerosis: PRISMS-15. J
2019;26(suppl 1):680. Neurol Neurosurg Psychiatry 2015;86(11):1202–7.
54. Ho PR, Koendgen H, Campbell N, Haddock B, Richman S, Chang I. 65. Rudick RA, Goodkin DE, Jacobs LD, et al. Impact of interferon beta-
Risk of natalizumab-associated progressive multifocal leukoencephal- 1a on neurologic disability in relapsing multiple sclerosis. The Multi-
opathy in patients with multiple sclerosis: a retrospective analysis of ple Sclerosis Collaborative Research Group (MSCRG). Neurology
data from four clinical studies. Lancet Neurol 2017;16(11):925–33. 1997;49(2):358–63.
55. Ryerson LZ, Foley J, Chang I, et al. Risk of natalizumab-associated 66. PRISMS-4: long-term efficacy of interferon-beta-1a in relapsing MS.
PML in patients with MS is reduced with extended interval dosing. Neurology 2001;56(12):1628–36.
Neurology 2019;93(15):e1452–62. 67. Johnson KP, Brooks BR, Cohen JA, et al. Extended use of glatiramer
56. Kappos L, Cohen J, Collins W, et al. Fingolimod in relapsing multiple acetate (Copaxone) is well tolerated and maintains its clinical
sclerosis: an integrated analysis of safety findings. Mult Scler Relat effect on multiple sclerosis relapse rate and degree of disability.
Disord 2014;3(4):494–504. Copolymer 1 Multiple Sclerosis Study Group. Neurology 1998;50
57. Cohen JA, Barkhof F, Comi G, et al. Oral fingolimod or intramuscular (3):701–8.
interferon for relapsing multiple sclerosis. N Engl J Med 2010;362 68. Lalive PH, Neuhaus O, Benkhoucha M, et al. Glatiramer acetate in the
(5):402–15. treatment of multiple sclerosis: emerging concepts regarding its mech-
58. Laroni A, Brogi D, Morra VB, et al. Safety of the first dose of fingoli- anism of action. CNS Drugs 2011;25(5):401–14.
mod for multiple sclerosis: results of an open-label clinical trial. BMC 69. Cadavid D, Wolansky LJ, Skurnick J, et al. Efficacy of treatment of
Neurol 2014;14:65. MS with IFNbeta-1b or glatiramer acetate by monthly brain MRI in
59. Scannevin RH, Chollate S, Jung MY, et al. Fumarates promote cyto- the BECOME study. Neurology 2009;72(23):1976–83.
protection of central nervous system cells against oxidative stress via 70. Cohen JA, Baldassari LE, Atkins HL, et al. Autologous hematopoietic
the nuclear factor (erythroid-derived 2)-like 2 pathway. J Pharmacol cell transplantation for treatment-refractory relapsing multiple sclero-
Exp Ther 2012;341(1):274–84. sis: position statement from the American Society for Blood and Mar-
60. Linker RA, Lee DH, Ryan S, et al. Fumaric acid esters exert neuropro- row Transplantation. Biol Blood Marrow Transplant 2019;25(5):845–
tective effects in neuroinflammation via activation of the Nrf2 antioxi- 54.
dant pathway. Brain 2011;134(Pt 3):678–92.
61. Mills EA, Mao-Draayer Y. Aging and lymphocyte changes by immu-
nomodulatory therapies impact PML risk in multiple sclerosis
patients. Mult Scler 2018;24(8):1014–22. SUPPLEMENTARY DATA
62. Naismith RT, Wundes A, Ziemssen T, et al. Diroximel fumarate dem- Supplementary data to this article can be found online at
onstrates an improved gastrointestinal tolerability profile compared https://doi.org/10.1016/j.amjmed.2020.05.049.
ARTICLE IN PRESS
11.e1 The American Journal of Medicine, Vol 000, No 000, && 2020

SUPPLEMENTARY APPENDIX because of the cooling effect of cold water in heat-sensitive


individuals). Because vitamin D deficiency is considered a
Advances in Treatment risk factor for MS and osteopenia, it is reasonable also to
Treating Acute Attacks. The terms “acute attack”, “acute correct with oral vitamin D. Table Supplementary Table
exacerbations”, and “relapses” are used interchangeably Supplementary Table 13-9 summarizes treatment options
and refer to the onset or worsening of neurologic deficits for common symptoms in MS.
lasting ≥24 hours in the absence of fever or infection.1
When acute deterioration results from an increase in ambi-
Supplementary References
ent temperature, fever, or infection, this is considered a
“pseudo exacerbation.”1 Glucocorticoids are used as first- 1. Berkovich RR. Acute multiple sclerosis relapse. Contin-
line treatment for attacks, as they provide short-term clini- uum (Minneap, Minn). 2016;22(3):799-814.
cal benefit by reducing the severity and shortening the dura- 2. Le Page E, Veillard D, Laplaud DA, et al. Oral versus
tion of attacks. Typically, intravenous (IV) intravenous high-dose methylprednisolone for treatment
methylprednisolone 1 g/day for 3-5 days is given, often fol- of relapses in patients with multiple sclerosis (COPOU-
lowed by an oral course of prednisone beginning at a dose SEP): a randomised, controlled, double-blind, non-infe-
of 60-80 mg/d and then tapered over 2 weeks.1 Other gluco- riority trial. Lancet. 2015;386(9997):974-981.
corticoid considerations are dexamethasone1 and high-dose 3. Chang E, Ghosh N, Yanni D, Lee S, Alexandru D,
oral prednisone (in equivalent doses to high-dose IV meth- Mozaffar T. A review of spasticity treatments: pharma-
ylprednisolone) that appears to be equally effective.2 cological and interventional approaches. Crit Rev Phys
Second-line treatment for patients resistant or refractory Rehabil Med. 2013;25(1-2):11-22.
to glucocorticoid treatment includes plasmapheresis, IV 4. Henze T, Rieckmann P, Toyka KV. Symptomatic treat-
immunoglobulin, and adrenocorticotropic hormone ment of multiple sclerosis. Multiple Sclerosis Therapy
(ACTH). The use of plasmapheresis (plasma exchange) is Consensus Group (MSTCG) of the German Multiple
reserved for cases of severe symptoms refractory to gluco- Sclerosis Society. Eur Neurol. 2006;56(2):78-105.
corticoids and generally involves 5-7 exchanges (40- 5. Toosy A, Ciccarelli O, Thompson A. Symptomatic treat-
60 mL/kg per exchange) every other day for 14 days.1 IV ment and management of multiple sclerosis. Handb Clin
immunoglobulin is not approved in this indication but is Neurol. 2014;122:513-562.
sometimes used off-label in steroid-unresponsive patients 6. Joy JE, Johnston RB Jr, eds. Multiple Sclerosis: Current
as second- or third-line treatment; notably, this is the pre- Status and Strategies for the Future. Washington, DC:
ferred treatment for postpartum patients. ACTH is another National Academies Press; 2001.
US Food and Drug Administration-approved option, but is 7. Jensen HB, Ravnborg M, Dalgas U, Stenager E. 4-Ami-
rarely used due to high cost and uncertain advantages over nopyridine for symptomatic treatment of multiple scle-
glucocorticoids; in individuals with poor intravenous access rosis: a systematic review. Ther Adv Neurol Disord.
or who are unable to tolerate oral corticosteroids, ACTH is 2014;7(2):97-113.
given intramuscularly at 80-120 units for 2-3 weeks and 8. Turner-Stokes L, Ashford S, Esquenazi A, et al. A com-
can be tapered. prehensive person-centered approach to adult spastic
paresis: a consensus-based framework. Eur J Phys Reha-
Symptomatic Therapies. For all patients, it is useful to bil Med. 2018;54(4):605-617.
encourage attention to a healthy lifestyle, including main- 9. Schneider SA, Deuschl G. The treatment of tremor. Neu-
taining an optimistic outlook, a healthy diet, and regular rotherapeutics. 2014;11(1):128-138.
exercise as tolerated (swimming is often well tolerated
ARTICLE IN PRESS
Hauser and Cree Treatment of Multiple Sclerosis 11.e2

Supplementary Table Symptomatic Treatment Approaches in Multiple Sclerosis (MS)


MS Symptom Approximate Nonpharmacological Treatments: Pharmacological Treatments: Key
Frequency in MS Key Recommendations Recommendations
Spasticity and spasms3-5 90%  Physiotherapy (including stretching)  Baclofen (oral [10-120 mg/d] and intrathe-
 Occupational therapy cal)
 Tizanidine (2-36 mg/d)
 Gabapentin (300-3600 mg/d)
 Clonazepam (0.25-2 mg/d)
 Diazepam (6-15 mg/d)
 Cannabinoids
 Botulinum toxin injection
Impaired gait6-8 80%  Adaptive devices  4-Aminopyridine (Dalfampridine) (20 mg/d)
 Physiotherapy
 Functional electrical stimulation
Pain4,5 Up to 86%  Pain management  Gabapentin (300-2400 mg/d)
 Pregabalin (150-600 mg/d)
 Duloxetine (20-120 mg/d)
 Amitriptyline (25-150 mg/d)
 Carbamazepine (100-1600 mg/d)
 Lamotrigine (200-400 mg/d)
 Topiramate (200-300 mg/d)
Ataxia/tremor4,5,9 80%  Physiotherapy  Carbamazepine (400-600 mg/d)
 Occupational therapy  Propranolol (40-240 mg/d)
 Wrist weights  Topiramate (100-333 mg/d)
 Thalamotomy  Cannabinoids
 Deep-brain stimulation  Primidone (up to 750 mg/d)
 Ondansetron (8 mg/d)
 Clonazepam (36 mg/d)
 (These agents have been tried with mixed
success; response is generally poor)
Bladder dysfunction4,5 70%-80% Assessments:  Mirabegron
 Urodynamic testing  Oxybutynin
Treatments:  Tolterodine
 Pelvic floor exercises  Solifenacin
 Electrical stimulation  Trospium chloride (40-60 mg/d)
 Fluid intake management  Desmopressin (up to 20 mg)
 Urinary aids
Depression4,5 50%  Psychotherapy  Fluoxetine
 Counseling  Sertraline
 Escitalopram
 Bupropion
 Venlafaxine
Fatigue4,5 75%  Cooling  Modafinil (200-400 mg/d)
 Regular exercise  Armodafinil
 Physiotherapy  Methylphenidate
 Sleep hygiene
Cognitive dysfunction4,5 40%-70%  Attention training  Lisdexamfetamine
 Memory training
 Cognitive rehabilitation
Paroxysmal symptoms4,5 10%-20%  Thermocoagulation  Carbamazepine (100-300 mg/d)
 Radiotherapy  Oxcarbazepine
 Lamotrigine (up to 400 mg/d)
 Gabapentin (up to 1600 mg/d)
 Topiramate (up to 300 mg/d)

You might also like