415 2017 Article 8594
415 2017 Article 8594
DOI 10.1007/s00415-017-8594-9
REVIEW
Received: 24 May 2017 / Revised: 11 August 2017 / Accepted: 12 August 2017 / Published online: 6 September 2017
© The Author(s) 2017. This article is an open access publication
Abstract The treatment landscape for relapsing forms of involved in treating people with RMS throughout the entire
multiple sclerosis (RMS) has expanded considerably over course of their disease.
the last 10 years with the approval of multiple new disease-
modifying therapies (DMTs), and others in late-stage clini- Keywords Multiple sclerosis · Therapeutic drug
cal development. All DMTs for RMS are believed to reduce monitoring · Selection for treatment · Re-treatment ·
central nervous system immune-mediated inflammatory pro- Treatment effectiveness
cesses, which translate into demonstrable improvement in
clinical and radiologic outcomes. However, some DMTs are
associated with long-lasting effects on the immune system Introduction
and/or serious adverse events, both of which may compli-
cate the use of subsequent therapies. When customizing a The unpredictable nature of multiple sclerosis (MS) clinical
treatment program, a benefit–risk assessment must consider manifestations within and between patients with apparently
multiple factors, including the efficacy of the DMT to reduce similar characteristics is brought about by a complex and
disease activity, the short- and long-term safety and immu- dynamic pathophysiology involving inflammatory-based
nologic profiles of each DMT, the criteria used to define mechanisms of demyelination and axon loss [1–3]. A range
switching treatment, and the risk tolerance of each patient. of genetic [4, 5], immunopathologic [6–8], and environmen-
A comprehensive benefit–risk assessment can only be tal/epigenetic [9] factors drive the tremendous variability in
achieved by evaluating the immunologic, safety, and efficacy the type, frequency, and severity of signs and symptoms that
data for DMTs in the controlled clinical trial environment may present during the course of MS [10, 11].
and the postmarketing clinical practice setting. This review Despite the heterogeneity in MS disease course, select-
is intended to help neurologists make informed decisions ing an appropriate therapy for relapsing forms of MS
when treating RMS by summarizing the known data for each (RMS) before the approval of fingolimod in 2010 [12, 13]
DMT and raising awareness of the multiple considerations was relatively simple because neurologists had two main
treatment options: interferon beta/glatiramer acetate or
natalizumab. The beta interferons and glatiramer acetate
* Gabriel Pardo have comparable long-term safety profiles and efficacy,
gabriel‑[email protected] reducing the frequency of relapses by ~30% over a 2- to
David E. Jones 3-year treatment period, as evaluated in clinical trials [1,
[email protected] 2, 14]. However, a high proportion of patients experi-
1
ence breakthrough disease or have persistent clinical or
OMRF Multiple Sclerosis Center of Excellence, Oklahoma
radiologic disease activity within 2 years of treatment ini-
Medical Research Foundation, 820 NE 15th Street,
Oklahoma City, OK 73104, USA tiation of these agents [14, 15]. Conversely, the safety of
2 interferon beta and glatiramer acetate over two decades
Department of Neurology, University of Virginia School
of Medicine, PO Box 800394, Charlottesville, VA 22908, is highly favorable and the relative risk for immunologic
USA complications is low [14, 16–18]. Natalizumab [19, 20] is
13
Vol.:(0123456789)
2352 J Neurol (2017) 264:2351–2374
more efficacious than interferon beta and glatiramer acetate General principles of treatment sequencing
[21, 22], but has a complex safety profile due to the risk in RMS
of progressive multifocal leukoencephalopathy (PML) [19,
20]. For this reason, historically, natalizumab was gener- The primary aim of treatment is to reduce disease activity to
ally reserved for patients requiring higher efficacy than optimize neurologic reserve, cognition, and physical func-
interferon beta and glatiramer acetate, and, more recently tion [27]. Meeting this goal requires a concordant relation-
in the US, only when the expected benefit is sufficient to ship between the health care professional and patient so that
offset PML risk [19, 20, 23]. the personal preferences of the patient are considered when
Since 2010, new disease-modifying therapies (DMTs) developing or revising the treatment plan (shared decision
have emerged [18]; 13 approved DMTs are currently avail- making). Patients should be informed that different DMTs
able to treat RMS worldwide (Table 1), which target dif- may be required at different times because of suboptimal
ferent pathways of the immune system (Table 2). Several response, safety concerns, intolerable side effects to the
of the new DMTs have demonstrated superior efficacy over DMT, or a change in the risk tolerance of the patient. Within
either interferon beta or glatiramer acetate in Phase III stud- this context, patients could be made aware of commonly
ies of patients with RMS (Table 3). While there may be used criteria necessitating a treatment switch due to subop-
differences in their relative efficacy, additional head-to-head timal response. For interferon beta recipients specifically,
clinical trials, particularly comparator studies between oral the Rio score estimated after 1 year of treatment is prog-
DMTs, are required to confirm and quantify this assertion nostic for ongoing disease activity in the ensuing 3 years
[24]. Oral DMTs such as teriflunomide, dimethyl fumarate, [28, 29]. For DMTs more generally, the Canadian Multiple
and fingolimod offer a convenient route of administration, Sclerosis Working Group recommends changing treatment
while peginterferon beta, daclizumab beta, ocrelizumab, and when there is a low level of concern in all three domains of
alemtuzumab provide a lower dosing frequency, being dosed the MS disease status triad [relapses, disability progression,
once every 2 weeks, monthly, every 6 months, and annually, and magnetic resonance imaging (MRI)], a medium level of
respectively. concern in any two domains, or a high level of concern in
Although the expansion in treatment options for RMS any one domain [30]. Another example is the multifactorial
is welcome, health care professionals are now faced with MS decision model that, by grading the four domains of
complicated decisions on how to individualize initial ther- relapse, disability progression, MRI, and neuropsychology,
apy for patients (see Table 4 for prognostic features in MS) aims to support early treatment decisions and detect treat-
and then select subsequent therapies, based on incomplete ment failure in a timely manner [31].
benefit–risk assessments of the current and potentially Regardless of the clinical criteria used to identify sub-
unknown long-term immunologic and safety risks. The optimally controlled RMS [30, 32], treatment sequencing
most important knowledge deficit is the long-term safety is often necessary to maintain disease control, which may
of newly approved DMTs for RMS, which may not have introduce an additional safety risk [30]. Neurologists have
been fully elucidated during their Phase III clinical trial two major decisions regarding the prescription of DMTs: (1)
programs, and thus may place some patients at risk for choosing the initial DMT expected to reduce disease activity
complications yet to be defined. For instance, some DMTs while recognizing the potential need for alternative later-line
for RMS have been associated with adverse events (AEs) DMTs if the response is suboptimal; and (2) selection of
that only came to light during postmarketing surveillance subsequent treatment choices based on previous DMT use.
[16, 25], culminating in the development of intensive The appropriate course of action selected depends on a thor-
risk reduction strategies to optimize patient safety such ough benefit–risk evaluation for each candidate DMT after
as classifications of PML risk [26]. Other generic factors accounting for specific disease- and patient-related factors
preventing the extrapolation of data to a real-world set- at a specific point in time, as well as the patient’s access to
ting include strict patient selection and high motivation DMTs through their health insurance plans.
in clinical trials. Treatment algorithms for RMS that rank DMTs as first
The purpose of this review is to raise awareness of the and second line have been proposed [33, 34], as have the
issues involved in sequencing RMS therapies by discuss- pros and cons of induction (starting highly effective therapy
ing the immunologic effects and known safety profiles of earlier in the course of the disease) versus escalation treat-
available DMTs. In doing so, the treating neurologist may ment paradigms [35, 36]. However, the initial DMT should
be better able to inform patients on the likely benefits and provide the most favorable benefit–risk profile given the
risks of treatment. level of disease activity over the last 6–12 months, taking
13
J Neurol (2017) 264:2351–2374 2353
IFN beta-1b (Betaseron®; Subcutaneous injection Every other day 250 mcg 1993a RMS [134]
Extavia®)
IFN beta-1a ( Avonex®) Intramuscular injection Once a week 30 mcg 1996 RMS [135]
IFN beta-1a ( Rebif®) Subcutaneous injection Three times per week 22 and 44 mcg 2002 RMS [136]
Peginterferon beta-1a Subcutaneous injection Once every 2 weeks 125 mcg 2014 RMS [137]
(Plegridy®)
Glatiramer acetate Subcutaneous injection Daily 20 mg 1996b RMS [138]
(Copaxone®; Glatopa®) Three times per week 40 mg 2014
Dimethyl fumarate Oral capsule Twice a day 240 mg 2013 US: RMS [67]
(Tecfidera®) Europe: RRMS [139]
Teriflunomide Oral tablets Daily 14 and 7 mg 2012 US: RMS [72]
(Aubagio®) Europe: RRMS [73]
Fingolimod (Gilenya®) Oral capsule Daily 0.5 mg 2010 US: RMS [13]
Europe: second-line treat-
ment or rapidly evolving
severe RRMS [12]
Daclizumab beta Subcutaneous injection Once monthly 150 mg 2016 US: RMS, generally after
(Zinbryta®)d an inadequate response
to ≥2 D MTsc [82]
Europe: RMS and failure
to respond/unsuitable
for other treatments [81,
140]
Alemtuzumab Intravenous infusion First course: daily for 12 mg 2014 US: generally reserved for
(Lemtrada®) 5 days; second course: patients who have had
daily for 3 days, 1 year an inadequate response
after the first course to ≥2 drugs for RMSc
[100]
Europe: active RRMS
defined by clinical or
imaging features [141]
Natalizumab (Tysabri®) Intravenous infusion Every 4 weeks 300 mg Approved 2004; US: RMS when the
reintroduced expected benefit is
2006 sufficient to offset PML
riskc [20]
Europe: high disease
activity despite IFN beta
or glatiramer acetate;
rapidly evolving severe
RRMS [19]
Ocrelizumab (Ocrevus®) Intravenous infusion Twice a year 600 mg 2017 US: RMS or PPMS [118]
Mitoxantrone Intravenous infusion Every 3 months 12 mg/m2 2000 US: secondary (chronic)
(Novantrone®; Every 3 months 5 mg/m2 progressive, progressive-
Onkotrone®) relapsing, or worsening
RMS (but not PPMS) as
an early, transient, high-
efficacy strategy [142]
DMT disease-modifying therapy, IFN interferon, MS multiple sclerosis, PML progressive multifocal leukoencephalopathy, PPMS primary pro-
gressive MS, RMS relapsing forms of multiple sclerosis, RRMS relapsing–remitting multiple sclerosis
a
Extavia available in the US since 2009
b
Glatopa available in the US since 2015
c
Only available through a restricted distribution program
d
INBRYTA®), which has a different form and structure than an earlier form of dacli-
Formerly daclizumab high-yield process (approved as Z
zumab
13
2354 J Neurol (2017) 264:2351–2374
into account the MS type, long-term prognostic factors 2–5 years and short first interattack interval) [33, 35, 38],
(Table 4), patient-related factors, and the fact that the risk and people of African descent [39]. Before an induction
of AEs associated with some DMTs can change over time. strategy is initiated, physicians must consider the appropri-
Importantly, due to the heterogeneity of RMS, the choice of ate maintenance DMT postinduction but, in practice, this is
starting DMT should take into consideration potential future not always possible and data to guide postinduction choices
treatment needs by keeping subsequent treatment options are very limited.
open (e.g., reserving the need for DMTs with a long-lasting The range of available induction therapies is fairly nar-
immunoablative impact on the immune system until a later row, and includes mitoxantrone, alemtuzumab, and to a lim-
time if appropriate). Like a good chess player who thinks ited extent, hematopoietic stem cell transplantation (within
several moves ahead, performing multiple benefit–risk a clinical trial setting). Off-label cyclophosphamide also
assessments across several DMTs for RMS is required when has been investigated in patients with highly active dis-
reviewing medication at any point in time. ease [40]. There are positive neuroradiologic data for the
brief use of immunosuppressive induction therapy with
mitoxantrone before maintenance therapy with glatiramer
Escalation versus induction acetate in patients with highly active RMS [41, 42]. How-
ever, the safety profile of the induction agent may preclude
Clinicians may deliberate on the relative value of an escala- many patients from receiving this treatment strategy. Use of
tion or induction treatment approach. A treatment escala- either immunoablative chemotherapy or immune-depleting
tion approach is based on starting with a relatively safer antibodies followed by autologous hematopoietic stem cell
agent and reactive treatment switches due to breakthrough transplantation has been successful in treating patients with
disease. At every stage in the patient’s disease course, there MS [43–45]. The immune-depleting antibody alemtuzumab
can be lasting effects of previous DMTs on the patient’s may be considered as an induction therapy because its effects
immune system, especially with medications that have pro- on the immune system persist long after treatment cessation,
longed immunologic sequelae. When switching from the enabling dosing on a one-off or annual basis [37]. In con-
initial DMT, it is imperative to consider the mechanism of trast, the high-efficacy DMTs fingolimod and natalizumab
action and duration of pharmacodynamic and immune sys- should not be considered as induction therapies because
tem effects because these can impact the efficacy and safety their rapidly reversible mechanisms of action predispose
of the next agent. Patients with moderate disease activity patients to a quick return of disease activity following treat-
[one disabling relapse in the last year and/or two new gad- ment cessation [46–53]. For a similar reason, it is likely
olinium-enhancing (Gd+) lesions, or accumulation of two that daclizumab beta should be used as a maintenance high-
new T2 lesions per year, indicating multifocal attacks] or efficacy DMT rather than as an induction therapy [54, 55].
high disease activity (at least one disabling relapse in 1 year
plus at least three new Gd+ lesions, or accumulation of three
new T2 lesions per year) may be placed on a high-efficacy Clinical pharmacology, safety, and monitoring
treatment early in the disease course and continue with that of DMTs
DMT [24, 30]. Some therapies with reversible mechanisms
of action facilitate escalation of therapy towards other agents Most DMTs have a clear pharmacodynamic drug–drug inter-
within a relatively short time frame of discontinuation if action by virtue of their temporal effects on immune cell
safety considerations allow, whereas other DMTs with long- counts and functions (Table 2). Because different DMTs
term effects on the immune system after treatment cessation exert distinct immunologic effects that persist for variable
can limit the scope of subsequent pharmacotherapy. The lat- periods of time after discontinuation, the immune system
ter DMTs also have been used as induction therapies. may not have fully recovered to its pretreatment baseline
Induction involves short-term use of a high-efficacy treat- physiologic composition during transition from one DMT
ment to obtain rapid control of highly active disease and to another. The type and duration of effect of the previous
to increase the likelihood of beneficial long-term outcomes DMT not only influences the selection of the subsequent
[35, 37], justifying an increased risk of serious AEs. The DMT, but also the known and unknown risk of an AE with
induction strategy is generally intended for younger patients the later-line DMT. As currently understood, the armamen-
(<40 years of age) with aggressive RMS who may have tarium of approved and investigational agents for MS can
already received immunomodulatory drugs, with frequent be grouped into DMTs that exert near-term effects on the
(at least two) and severe relapses within the last 12 months, immune system [day-to-week timescale: interferon beta-1a
neuroradiologic activity (at least two additional Gd+ lesions and 1b, peginterferon beta-1a, glatiramer acetate, dimethyl
on recent T2 MRI), and who are at increased risk of rapid fumarate, teriflunomide (if an accelerated elimination pro-
accumulation of disability (e.g., high relapse rate in the first cedure with activated charcoal or cholestyramine is used)],
13
Table 2 Mechanism of action and effects on the immune system of DMTs for RMS
DMT Molecular mode of action Effect on immune cells and mediators Time taken for immune system reconstitution after
DMT cessation
SC IFN beta-1b (Betaseron; Extavia) Exert autocrine and paracrine actions via activa- Reduces inflammatory cell migration across the Effects on the immune system endure for five
IM IFN beta-1a (Avonex) tion of the IFN receptor on leucocytes blood–brain barrier, reduces the production of times the serum elimination half-life (i.e., 40 min
SC IFN beta-1a (Rebif) proinflammatory cytokines, and induces anti- to 21.5 h for SC IFN beta-1b [134], 95 h for IM
IM peginterferon beta-1a (Plegridy) inflammatory cytokines [18] IFN beta-1a [135], 345 h for SC IFN beta-1a
[136], and 390 h for SC peginterferon beta-1a)
[137]
J Neurol (2017) 264:2351–2374
Glatiramer acetate (Copaxone) MBP mimetic; thus competes with MBP antigens Protects neurons by diverting T cell responses Effects on the immune system endure for five
to bind with MHC II [58] away from myelin in a dose-dependent manner. times the elimination half-life [138]
Increases production of anti-inflammatory
cytokines and reduces the production of proin-
flammatory cytokines [18]
Dimethyl fumarate (Tecfidera) Activates the nuclear factor (erythroid-derived Mean absolute lymphocyte count decreased >4 weeks for lymphocyte counts to increase, but
2)-like 2 pathway to protect against oxida- by 30% during the first year then remained did not return to baseline values [67]
tive stress–induced cellular injury and loss in stable above the lower limit of normal (i.e.,
neurons and astrocytes [61] 0.91 × 109/L) [63]
6% of patients had lymphocyte counts
<0.5 × 109/L [67]
Effective immune response to recall antigens,
neoantigens, and T cell-independent antigens
similarly to nonpegylated IFN therapy [143]
Teriflunomide (Aubagio) Inhibits proliferation of activated T and B Reduces neutrophils and lymphocytes by 15%, Unknown; reduced white blood cell counts of 15%
lymphocytes via mitochondrial dihydroorotate with mean counts remaining in the normal may be related to bone marrow suppression [72]
dehydrogenase inhibition [144] range [71]
16% and 12% of patients had neutrophil and lym-
phocyte counts <1.5 × 109 and <0.8 × 109/L
with 14 mg dose, respectively [72]
Effective immune response to H1N1, H3N2, and
B influenza strains [145]
Fingolimod (Gilenya) Binds the sphingosine-1-phospate receptor, Dose-dependent reduction in peripheral lympho- ≤2 months to return to normal range [13]; average
blocking lymphocyte egress from lymph nodes cyte count to 20–30% of baseline values [13] lymphocyte counts were 80% of baseline values
[146, 147] Lymphopenia incidence was 7% in placebo- after 3 months [149]
controlled trials [13]
Attenuated response to influenza vaccine in some
patients [148]
Daclizumab beta (Zinbryta)a Humanized monoclonal antibody that selectively Fivefold expansion in CD56bright NK cells at Pharmacodynamics are related to the half-life of
blocks high-affinity IL-2 receptor formation on 1 year. Total lymphocyte, CD4+ and CD8+ daclizumab beta (21 days) and are reversible [55,
activated T cells. Modulation of the IL-2 signal T cell, and B cell counts decrease ≤10% from 81, 82]
leads to selective antagonism of activated T cell baseline during the first year of treatment [81, Total lymphocyte counts return to baseline levels
responses and expansion of immunoregulatory 82, 92] ~8–12 weeks after the last dose [81, 82]
CD56bright NK cells [81, 87] Effective immune responses to influenza vaccine Treg and CD56bright NK cell numbers return to
[150] baseline levels within 24 weeks [81, 93, 95]
13
2355
Table 2 (continued)
2356
DMT Molecular mode of action Effect on immune cells and mediators Time taken for immune system reconstitution after
DMT cessation
13
Alemtuzumab (Lemtrada) Targets CD52 on lymphocytes and monocytes. Decrease in the level of circulating T and Lymphocytes repopulate within 8 months, but
It readily depletes B cells, T cells, monocytes, B lymphocytes very rapidly, with the lowest T cell populations take >1 year to fully
macrophages, and dendritic cells, leading to values observed within days posttreatment [99] repopulate [100, 101]
long-lasting changes in adaptive immunity, T cell populations do not recover to baseline levels
and reduces the pathogenesis of inflammatory [101]
response in MS [99]
Natalizumab (Tysabri) Monoclonal antibody that selectively inhibits Increases the number of circulating leukocytes ≤16 weeks to return to baseline levels [20]
VLA-4 (α4β1) integrins, preventing leukocyte (including lymphocytes, monocytes, basophils,
migration across the BBB [58] and eosinophils) [20]. Natalizumab does not
affect the absolute count of circulating neutro-
phils [20]
Effective immune responses to recall antigen
(tetanus toxoid) and neoantigen (keyhole limpet
hemocyanin) [151]
Ocrelizumab (Ocrevus) Targets CD20 on B cells through mechanisms CD20+ targeted by ocrelizumab include Median (range) time to B cell repletion to either
that include antibody-dependent cell-mediated pre-B cells, mature B cells, and memory baseline or lower limit of normal was 72 weeks
cytotoxicity, complement-dependent cytotoxic- B cells; lymphoid stem cells and plasma cells (range 27–175 weeks); within 2.5 years after last
ity, and/or the induction of apoptosis [118, 152] are unaffected [119, 152] infusion, B cell counts rose to either baseline
or the lower limit of normal in 90% of patients
[118]
Mitoxantrone (Novantrone) Intercalates with DNA, causing strand breaks, Reduction of leukocytes primarily affecting neu- Unknown
and inhibits DNA repair via inhibition of topoi- trophils and most lymphocyte subsets except
somerase II, leading to cytotoxicity [153] for naive and activated T lymphocytes [154]
BBB blood–brain barrier, DMT disease-modifying therapy, IFN interferon, IL-2 interleukin 2, IM, intramuscular, MBP myelin basic protein, MHC II class II major histocompatibility complex,
MS multiple sclerosis, NK natural killer, RMS relapsing forms of multiple sclerosis, SC, subcutaneous, Treg regulatory T cell, VLA-4 very late activation antigen 4
a
Formerly daclizumab high-yield process (approved as Z
INBRYTA®), which has a different form and structure than an earlier form of daclizumab
J Neurol (2017) 264:2351–2374
Table 3 Efficacy of DMTs for MS
DMT (trade name) ARR reduction Disability progressiona Neuroradiologic outcomes
IFN beta-1b (Betaseron; Extavia) 0.25 mg 34% No effect 0.9% reduction in lesion area from baseline
dose [155] versus a 21.4% increase with placebo [134]
IFN beta-1a (Avonex) [156] 18% for ITT population; 32% for completer 37% relative reduction in 24-week CDP by Reduction in number of G d+ lesions (0.80 vs.
population end of year 2 [135] 1.65) and volume of G d+ lesions per patient
(74.1 vs. 122.4 mm3)
IFN beta-1a (Rebif) [157] 29% (22 mcg); 32% (44 mcg) 23% (22 mcg) and 31% (44 mcg) relative Median 1.2% (22 mcg) and 3.8% (44 mcg)
J Neurol (2017) 264:2351–2374
13
2357
Table 3 (continued)
2358
13
Alemtuzumab (Lemtrada) Treatment-naive patients
CAMMS223: 69 and 66% in relapse risk CAMMS223: 75 and 69% risk reduction Improvement in lesion load on T2-weighted
over 3 and 5 years, respectively, versus IFN over 3 and 5 years, respectively, versus IFN MRI, and cerebral volume on T1-weighted
beta-1a 44 mcg SC three times per week beta-1a 44 mcg SC three times per week MRI in CAMMS223 [169]
[37, 169] [37, 169]
CARE-MS I: 55% in relapse risk over 2 years CARE-MS I: No statistically significant effect CARE-MS I: reduced the proportions of
versus IFN beta-1a 44 mcg SC three times over 2 years versus IFN beta-1a 44 mcg SC patients with G
d+ lesions (7% alemtuzumab
per week [170] three times per week [170] vs. 19% IFN beta-1a 44 mcg SC three times
per week) and new or newly enlarging T2
hyperintense lesions (48 vs. 58%)
Treatment-experienced patients
CARE-MS II: 49% in relapse risk versus IFN CARE-MS II: 42% reduction in 6-month risk No between-group difference on T2 lesion
beta-1a 44 mcg SC three times per week versus IFN beta-1a 44 mcg SC three times volume; reduced the proportions of patients
[105] per week with Gd+ lesions (9% alemtuzumab vs. 23%
IFN beta-1a 44 mcg SC three times per week)
and new or newly enlarging T2 hyperintense
lesions (46 vs. 68%)
Natalizumab (Tysabri) [171] AFFIRM: 68% AFFIRM: 42% risk reduction in 12-week AFFIRM: 92% fewer Gd+ lesions and 83%
CDP; 54% risk reduction in 24-week CDP reduction in new or newly enlarging T2
hyperintense lesions
Ocrelizumab (Ocrevus) [120] OPERA I: 46% versus IFN beta-1a OPERA I and II: 40% risk reduction in 12- OPERA I: 94% reduction in G d+ lesions; 77%
OPERA II: 47% versus IFN beta-1a and 24-week CDP versus IFN beta-1a reduction in new or newly enlarging T2
hyperintense lesions versus IFN beta-1a
OPERA II: 95% reduction in G d+ lesions;
83% reduction in new or newly enlarging T2
hyperintense lesions versus IFN beta-1a
Mitoxantrone (Novantrone) [172, 173] 63% of relapse risk [172] 70% reduction in 24-week CDP [173] Gd+ lesions (0 vs. 16% on placebo); mean
increase in T2 lesions: 0.29 in the mitox-
antrone group and 1.94 in the placebo group
[172]
Efficacy data are over 2 years and relative to placebo unless otherwise stated
ARR annualized relapse rate, CDP confirmed disability progression, DMT disease-modifying therapy, Gd+ gadolinium-enhancing, IFN interferon, IM intramuscular, ITT intention-to-treat, MRI
magnetic resonance imaging, MS multiple sclerosis, SC subcutaneous
a
Disability progression data are significant unless stated otherwise
b
Formerly daclizumab high-yield process (approved as Z
INBRYTA®), which has a different form and structure than an earlier form of daclizumab
J Neurol (2017) 264:2351–2374
J Neurol (2017) 264:2351–2374 2359
Gd+ gadolinium-enhancing, Ig immunoglobulin, MRI magnetic resonance imaging, MS multiple sclerosis, NEDA no evidence of disease activity
mid-term immunologic effects (week-to-month timescale: suggesting that better outcomes may be seen with shorter
fingolimod, natalizumab, and daclizumab beta), and long- washout. Another point for consideration is whether a pre-
term immunologic effects [month-to-year timescale: mitox- vious drug could potentially nullify or attenuate the mode
antrone, teriflunomide (if the rapid elimination procedure of action of later-line therapies: would the B and T cell-
is not implemented), alemtuzumab, and ocrelizumab]. A depleting action of alemtuzumab occur immediately after
more precise classification of the immunologic half-lives use of fingolimod if lymphocytes have not yet exited from
of DMTs will be possible once their effects on lymphocyte secondary lymphoid tissue? Hence, the diverse interactions
subpopulations are better understood. The safety profile and of DMTs with the immune system underscore their efficacy
immunologic effects of interferon beta and glatiramer ace- and safety profile, which, in turn, guides patient monitoring.
tate are such that an immediate transition to another DMT The increased efficacy and/or patient convenience asso-
is possible, providing there are no obvious abnormalities on ciated with newer DMTs relative to interferon beta and
clinical biochemistry. glatiramer acetate must be balanced against known and
Arguably, a transition period between stopping the exist- unknown safety issues. DMTs have the potential to produce
ing DMT and initiating a new DMT may be unnecessary on- and/or off-target-based toxicities that manifest as unex-
with some of the newer DMTs, but there is a fine balance pected serious AEs. Safety concerns for some therapies only
between the duration of washout (if any) and risk of dis- became evident during extension studies and postmarketing
ease. For instance, the immunologic effect of alemtuzumab surveillance studies, requiring ongoing changes to several
persists long after cessation of therapy and is unrelated to DMT product labels [16]. It follows that a more accurate
its biologic half-life, which may expose patients to immune- benefit–risk assessment is possible for a DMT scrutinized
mediated risks when they are switched to subsequent DMTs. by postmarketing surveillance than for a DMT that has com-
However, aggressive rebound disease activity can resume pleted Phase III clinical development, but has yet to undergo
shortly after stopping one agent and initiating another, as has safety evaluation in a large patient population over a pro-
been observed with natalizumab and fingolimod [46–48, 51], tracted period of drug exposure. For instance, a prospective
13
2360 J Neurol (2017) 264:2351–2374
descriptive study of patients with MS between 1995 and antioxidant responses (Table 2) [60]. It is likely that dime-
2006 found an increased malignancy risk with the sequenc- thyl fumarate has several additional immunomodulatory
ing of multiple immunomodulatory and immunosuppressant effects underpinning its efficacy in MS, including directing
therapies, and also with the number of immunosuppressant the immune response away from Th1 [62].
courses [56]. Integrated analyses of patient-level data (N = 2470)
from Phase IIb, Phase III, and long-term extension studies
Beta interferons, peginterferon beta‑1a, and glatiramer of dimethyl fumarate showed that mean absolute lympho-
acetate cyte count decreased by 30%, but generally remained above
the lower limit of normal during the first year of treatment,
The mechanism(s) of action of the beta interferons are not before stabilizing [63]. Separate observational data indicated
yet fully established, although these therapies have been that the dynamics of the absolute lymphocyte count gener-
available since the 1990s. All beta interferons are known ally correlate with CD4+ and CD8+ counts [64], with the
to exert autocrine and paracrine actions via activation of reduction of CD8+ T cells greater than that of CD4+ T cells
the interferon receptor on leukocytes (Table 2). Production (−55 vs. −39%) reflected in a 36% increase in the CD4/
of proinflammatory cytokines is reduced, and production CD8 ratio [65]. In patients without severe lymphopenia (i.e.,
of anti-inflammatory cytokines is induced [18]. Attachment <0.5 × 109 cells/L), there is evidence of improvement in
of a polyethylene glycol side chain to the parent interferon lymphocyte counts following discontinuation of dimethyl
beta-1a molecule yields peginterferon beta-1a, which, when fumarate, but full restoration takes >4 weeks [63, 66, 67].
administered subcutaneously, has a longer half-life, higher For patients who become severely lymphopenic on dimethyl
systemic exposure, and lower immunogenicity potential than fumarate, lymphocyte counts may take a long time to recover
intramuscular interferon beta-1a [57]. Glatiramer acetate, a [66]; this delay in lymphocyte recovery may complicate the
synthetic polymer of four amino acids (l-glutamate, l-lysine, switch to a subsequent DMT with myelosuppressive effects.
l-alanine, and l-tyrosine), is a mimetic for the MS autoan- Six percent of patients experienced lymphocyte counts
tigen myelin basic protein (MBP), and thus competes with <0.5 × 109/L (grade ≥3 lymphopenia) in placebo-controlled
MBP antigens for binding to class II major histocompat- trials [67]. The risk of developing moderate to severe lym-
ibility complex (MHC II) [58]. With formation of the MBP phopenia while on dimethyl fumarate may be increased by
antigen/MHC II complex impeded by glatiramer, helper T the following: increasing age, lower baseline absolute lym-
cells have less opportunity for activation and potential to phocyte count, and recent natalizumab exposure (there is a
destroy myelin [58]. In addition, glatiramer binding to MHC greater percentage reduction in absolute lymphocyte count
II inhibits the interaction of MHC II with C D4+ molecules due to the lymphocytosis induced by prior natalizumab) [64,
located on the surface of helper T cells (Th1 and Th2). Con- 66]. Although the risk may be slightly different in patients of
sequently, there is reduced production of proinflammatory older age or with lower baseline absolute lymphocyte count,
cytokines (interferon gamma) by Th1 cells, increased pro- all patients remain at a small risk of lymphopenia. Based
duction of anti-inflammatory cytokines (interleukin 10) by on 7250 cumulative patient-years of exposure, the overall
Th2 cells (promoting a less inflammatory state), and induc- incidence of serious infections was low, and there was no
tion of antigen-specific expansion of F OXP3+ regulatory T apparent correlation between the incidence of infection and
cells [58, 59]. grade of lymphopenia [63].
The effects of beta interferons and glatiramer acetate on Of >230,000 patients treated with dimethyl fumarate
the immune system likely only endure for as long as a patient globally in the 3 years following commercial availability
is exposed to therapeutic drug concentrations (i.e., less than (representing >330,000 patient-years), there have been five
five times the elimination half-life; Table 2). The safety pro- cases of PML in the setting of moderate to severe prolonged
files of the beta interferons and glatiramer acetate are well lymphopenia (absolute lymphocyte count <0.8 × 10 9/L),
established (Table 5). a rare opportunistic brain infection caused by John Cun-
ningham (JC) virus that is normally harmless in immuno-
Dimethyl fumarate competent hosts [25, 68]. There are established risk strati-
fication and mitigation strategies for patients on dimethyl
Following oral administration, dimethyl fumarate is rapidly fumarate in light of its safety profile. In the US, complete
metabolized to its active metabolite monomethyl fumarate, blood counts at baseline and every 6 months thereafter are
which is primarily responsible for its efficacy in MS [60, mandatory to identify patients who may have developed
61]. Similar to other fumarate derivatives, administration severe prolonged lymphopenia [67]. In Europe, a baseline
of dimethyl fumarate activates nuclear factor-erythroid MRI also should be performed within 3 months of initiating
2-related factor 2, resulting in differential effects involving therapy [69].
13
Table 5 Tolerability, safety, and monitoring issues of DMTs for MS
DMT Common AEs Safety issues Contraindications
IFN beta-1b (Betaseron/Extavia) [134] Injection site reaction, lymphopenia, flu-like Hepatic injury, anaphylaxis, depression (and Pregnancy (increased risk of spontaneous abor-
IFN beta-1a (Avonex) [135] symptoms, myalgia, leukopenia (asymp- suicidal ideation), injection site necrosis, tion) and breastfeeding
tomatic), neutropenia, increased liver congestive heart failure, leukopenia throm- Hypersensitivity reactions to active ingredient
IFN beta-1a (Rebif) [136]
enzymes, headache, hypertonia, pain, rash, botic microangiopathy, flu-like symptom or formulation excipients
Peginterferon beta-1a (Plegridy) [137] insomnia, abdominal pain, asthenia, depres- complex, seizures, autoimmune disorders,
sion, hematologic abnormalities, arthralgia decreased peripheral blood counts
Glatiramer acetate (Copaxone) [138] Injection site reactions, postinjection reaction Cutaneous necrosis Use during pregnancy only if clearly needed
J Neurol (2017) 264:2351–2374
13
2361
Table 5 (continued)
2362
13
Daclizumab-beta (Zinbryta) [81, 82]a Nasopharyngitis, upper respiratory tract infec- Hepatic injury (including autoimmune hepati- Hypersensitivity reactions to active ingredient
tion, rash, influenza, dermatitis, oropharyn- tis), other immune-mediated disorders (skin or formulation excipients
geal pain, bronchitis, eczema and lymphad- reactions, lymphadenopathy, noninfectious Preexisting hepatic disease or hepatic impair-
enopathy, depression, pharyngitis, increased colitis), infections, and depression ment, including ALT or AST ≥2 times the
alanine aminotransferase ULN
History of autoimmune hepatitis or other auto-
immune condition involving the liver
Alemtuzumab (Lemtrada) [100, 141, Rash, headache, pyrexia, nasopharyngitis, Infusion-associated reactions and anaphylaxis HIV infection
188–192] nausea, vomiting, infection (urinary tract, (including bradycardia), thyroid disor- Hypersensitivity reactions to active ingredient
upper respiratory tract, viral including ders and other autoimmune cytopenias, or formulation excipients
herpes, fungal), fatigue, insomnia, urticaria, glomerulonephritis (Goodpasture’s disease),
pruritus, thyroid gland disorders, arthralgia, malignancy (thyroid cancer, melanoma, and
pain in extremity, back pain, oropharyngeal lymphoproliferative disorders), infections
pain, abdominal pain, diarrhea, sinusitis, (including opportunistic such as herpes
paresthesia, dizziness, flushing virus, human papilloma virus, fungal infec-
tions, listeria, and nocardiosis)
Natalizumab (Tysabri) [19, 20, 193, 194] Headache, fatigue, arthralgia, urinary tract PML, hypersensitivity reactions (including Patients who have or have had PML
infection, lower respiratory tract infection, anaphylaxis), immunosuppression/infections Patients at risk of opportunistic infections
gastroenteritis, vaginitis, depression, pain in (including herpes simplex virus, meningitis, Concomitant beta IFN or glatiramer acetate
extremity, abdominal discomfort, diarrhea, and hepatitis B virus infection with acute therapy
rash fatal liver injury), hepatic injury Known active malignancies (except cutaneous
basal cell carcinoma)
Hypersensitivity reactions to active ingredient
or formulation excipients
Ocrelizumab (Ocrevus) [118] Infusion-related reactions and upper respira- Infusion-related reactions, infections, neo- Active hepatitis B virus infection and a his-
tory tract infections plasms tory of life-threatening infusion reaction to
ocrelizumab
Mitoxantrone (Novantrone) [142] Nausea, alopecia, urinary tract infection, Congestive heart failure (can result in death) Baseline LVEF below the lower limit of normal
menstrual disorders (including amenorrhea), may occur either during or months to years Pregnancy and breastfeeding
asthenia after termination of therapy. Secondary Hypersensitivity reactions to active ingredient
acute myeloid leukemia, infection, leuko- or formulation excipients
penia, depression, bone pain, emesis, renal
failure
AE adverse event, AV atrioventricular, DMT disease-modifying therapy, HIV human immunodeficiency virus, IFN interferon, LEVF left ventricular ejection fraction, MS multiple sclerosis, PML
progressive multifocal leukoencephalopathy, ULN upper limit of normal
a
Formerly daclizumab high-yield process (approved as Z
INBRYTA®), which has a different form and structure than an earlier form of daclizumab
J Neurol (2017) 264:2351–2374
J Neurol (2017) 264:2351–2374 2363
13
2364 J Neurol (2017) 264:2351–2374
blood counts, and examination of the fundus for macular from real-world use, although the product label provides
edema [13]. Monitoring for signs of infection and suspicious specific warnings regarding hepatic injury—elevations of
skin lesions in case of basal cell carcinoma also should be serum transaminases and serious events, including fatal
conducted, as per the fingolimod product label [13]. Case cases of autoimmune hepatitis and liver failure—and other
reports of severe disease reactivation following fingolimod immune-mediated disorders (including skin reactions, lym-
withdrawal [46–48] also must be considered when treat- phadenopathy, and noninfectious colitis) as well as depres-
ing patients with highly active disease and in women who sion, infections, autoimmune hemolytic anemia, gastroin-
wish to become pregnant (in whom it is advised to continue testinal AEs, and lymphopenia [81, 82]. These warnings are
therapy only if the potential benefit justifies the potential based on safety and tolerability information from an inte-
risk to the fetus) [12]. grated analysis of six clinical studies (primarily randomized
controlled trials and their extensions) encompassing 2236
Daclizumab beta patients with 5214 patient-years of daclizumab beta expo-
sure [98]. Although this database was not large enough to
Daclizumab beta is a humanized monoclonal antibody detect rare events, the analysis did show that daclizumab
approved for the treatment of RMS as a monthly subcuta- beta had an acceptable safety profile without evidence of
neous self-injectable [81–83]. Daclizumab beta works in cumulative toxicity [98]. Daclizumab beta should be dis-
the periphery by binding CD25 (alpha subunit of the high- continued in cases of significant transaminase elevation [i.e.,
affinity interleukin 2 receptor mostly expressed on activated alanine transaminase (ALT) or aspartate transaminase (AST)
T cells) to modulate interleukin 2 signaling (Table 2) >5 × the upper limit of normal (ULN) only; or total biliru-
[84–87]. Blockade of CD25 by daclizumab beta limits inter- bin greater >2 × ULN; or ALT or AST ≥3 but <5 × ULN
leukin 2 consumption by activated T cells and facilitates cells and total bilirubin >1.5 and <2 × ULN] [82]. Discontinu-
that express the intermediate-affinity interleukin 2 receptor ing daclizumab beta should be considered if severe depres-
[i.e., natural killer (NK) cells and precursors of innate lymphoid sion or suicidal ideation occurs [81, 82]. If serious infection
cells] to receive more interleukin 2 signal, as this receptor develops, daclizumab beta should be withheld until the epi-
does not feature CD25 [88, 89]. Consequently, there is a sub- sode resolves [81, 82].
stantial expansion of immunoregulatory CD56bright NK cells
that penetrate the blood–brain barrier and eliminate impor- Alemtuzumab
tant mediators of MS immunopathology, activated T cells,
leaving resting T cells intact (Table 2) [84, 86, 90, 91]. The humanized monoclonal antibody alemtuzumab targets
The pharmacodynamic effects of daclizumab beta are sus- the extracellular glycoprotein CD52, resulting in antibody-
tained in patients with relapsing–remitting MS, as evidenced dependent cytolysis and complement-mediated lysis of T and
by a fivefold expansion of CD56bright NK cell levels that pla- B lymphocytes, monocytes, NK cells, macrophages, and den-
teau by the end of the first year of treatment [92, 93]. Modest dritic cells (Table 2) [99, 100]. Alemtuzumab elicits rapid,
10% reductions in circulating total lymphocytes, CD4+ and profound, and prolonged B and T cell lymphopenia followed
CD8+ T cells, and B cells were observed in patients with MS by a reconstituted immune system different in composition
after 1 year of daclizumab beta 150 mg treatment, and regu- from that before treatment, which may rationalize its long-
latory T cell levels were reduced by approximately 50% after term efficacy, given patients only receive two medication
8 weeks [92–94]. The remaining regulatory T cells are func- cycles that are 1 year apart (Table 3) [99, 100]. It can take
tionally active, as evidenced by stable cytokine production, ~8 months for B cells and up to 3 years for T cell subsets to
maintained active cell cycling, and retention of a regulatory recover to the lower limits of the normal range after a single
T cell-specific demethylated region in the FOXP3 promoter, course of alemtuzumab, and T cells may not recover fully to
albeit with a significant decrease in CD25 expression [94]. baseline values [101]. It is worth noting that B cell recovery
The effects of daclizumab beta are reversible; after treatment was rapid in one study; levels of ‘mature naive’ B cells (CD19
cessation, total lymphocyte counts return to baseline levels and CD23 positive but CD27 negative) returned to baseline by
within 12 weeks, and CD56bright NK cell and regulatory T 3 months and rose to 165% of baseline values by 12 months
cell counts return to baseline levels within 24 weeks [93, after the first course of alemtuzumab treatment [102]. Con-
95]. There was no apparent impact of antidrug antibodies or versely, CD27-positive memory B cell recovery was slow,
neutralizing antibodies on the pharmacodynamics, efficacy, reaching only 25% of baseline levels by month 12 [102]. The
or safety of daclizumab beta [82, 96]. immunosuppressive effects of alemtuzumab on C D4+ T cell
The safety profile of daclizumab beta was determined subsets lasted for up to 4 years in 29 patients who participated
over a 5-year period and was consistent with that from in CARE-MS I and CARE-MS II [103]. Differential lympho-
the pivotal Phase III trial (Table 5) [97]. Given the recent cyte reconstitution after alemtuzumab treatment may be a bio-
approvals for daclizumab beta, there are only limited data marker for MS relapse, as patients with active disease showed
13
J Neurol (2017) 264:2351–2374 2365
an accelerated recovery of CD4+ cells (p = 0.001), with a transmigration out of the vascular space), but does not affect
difference in absolute CD4+ counts at 24 months (p = 0.009), the absolute count of circulating neutrophils [20].
while CD4+ counts <388 × 106 cells/mL predicted MRI sta- Natalizumab has an elimination half-life of 11 days
bility [104]. Anti-alemtuzumab antibodies reduce plasma [20], although plasma natalizumab concentrations can be
alemtuzumab concentrations during course 2 but not course reduced by 92% within 1 week of plasma exchange sessions
1, although they do not appear to affect clinical outcomes, to treat PML if required [106]. The reversibility of natali-
total lymphocyte count, or AEs [100]. Alemtuzumab induces zumab’s pharmacologic effects on peripheral immune cells
long-term immunodepleting effects, which must be consid- is evident starting at weeks 8–12, with levels returning to
ered when planning subsequent therapies for maintenance those observed or expected in non-natalizumab patients by
treatment or if a patient does not respond adequately. There 16–20 weeks after the last natalizumab dose [107]. This is
are no data on sequencing therapies after alemtuzumab use consistent with the reduction in plasma natalizumab con-
to guide the clinician, who must, therefore, rely on intensive centrations to below the limit of detection by 16 weeks post-
patient monitoring to individualize care. dose [107]. Lymphocyte counts remain within the normal
Although the advantages of long-lasting efficacy and range at all times both for patients receiving natalizumab and
extremely high patient adherence are positive attributes of for those who have stopped natalizumab treatment [107].
alemtuzumab, Table 5 shows that this DMT is associated Patients who develop anti-natalizumab antibodies are more
with several serious AEs that may arise years after starting likely to have hypersensitivity reactions during drug admin-
treatment and are, therefore, not reflective of alemtuzum- istration [20].
ab’s pharmacokinetic profile (elimination half-life, 2 weeks) An intensive risk stratification program is in place to help
[100]. Alemtuzumab is usually reserved for patients with prescribers weigh the clear efficacy benefits of natalizumab
unfavorable prognostic indicators because it is difficult to against the development of PML [19, 20]. Three main fac-
reconcile its superior efficacy over interferon beta with tors drive the risk of developing PML in patients undergo-
exposure to serious AEs in patients with less severe disease. ing natalizumab therapy: (1) therapy ≥24 months; (2) previ-
Even in patients with highly active disease, diligent patient ous use of immunosuppressant treatment; and (3) JC virus
selection and strict adherence to risk monitoring programs antibody positivity [108]. The anti-JC virus antibody index
is required. The alemtuzumab product label recommends value and duration of natalizumab treatment are two key fac-
regular laboratory monitoring up to 4 years after the last tors that enable clinicians and JC virus-positive immunosup-
alemtuzumab dose (and beyond if warranted) for the detec- pressant-naive patients with MS to make informed treatment
tion of secondary autoimmune conditions (e.g., immune and monitoring decisions [109–111].
thrombocytopenia, antiglomerular basement membrane Natalizumab withdrawal often leads to an MS relapse and
disease, and thyroid disorders) [100]. Laboratory testing return of inflammatory disease activity on MRI [49–53].
includes differential blood count, serum creatinine, and urine Younger patients (<40 years of age) were 3.8-fold more
analysis before administration and monthly thereafter [100]. likely to have increased MRI activity during 24 weeks of
Pretreatment thyroid-stimulating hormone level is manda- natalizumab treatment interruption, as were those with one
tory and requires rechecking every 3 months until 4 years d+ lesions (2.7-fold increase) and >five G
to five G d+ lesions
after the last infusion [100]. Patients with active or uncon- (6.2-fold) before natalizumab initiation (vs. no lesions) [52].
trolled infections are not candidates for therapy [100]. Pro- Initiating interferon beta within 30 days postnatalizumab
phylactic oral acyclovir should be taken until C D4+ count dosing in patients who had been free of disease activity
3
is >200 cell/mm to reduce the risk for herpes infections [112], or initiating fingolimod ~4 months postnatalizumab
[100, 105]. dosing in patients with stable Expanded Disability Status
Scale scores [113], was associated with clinical and radio-
Natalizumab logic disease recurrence. A therapeutic gap of no more than
3 months between discontinuing natalizumab and initiating
Natalizumab is a humanized monoclonal antibody that fingolimod appears to minimize the risk of relapse [113].
binds to the integrin molecule very late activation antigen 4 Switching from natalizumab to alemtuzumab (n = 16) [114]
(α4β1), a glycoprotein surface molecule found on all leuko- or off-label rituximab (n = 114 [115] and n = 118 [116])
cytes except neutrophils (Table 2) [58]. Blockade of α4β1 may be a feasible option to maintain disease control, includ-
prevents adhesion of leukocytes to vascular cell adhesion ing in those at high risk of PML. It is currently unknown if
molecule 1, a protein expressed on the surface of vascu- the switch-to therapy selection impacts the risk of PML in
lar endothelial cells in the brain and spinal cord, and thus this context. Data on treatment selection after a PML event
blocks entry of leukocytes into the central nervous system on natalizumab are limited, but there are reports of success-
across the blood–brain barrier [58]. Natalizumab increases ful use of both dimethyl fumarate and fingolimod in this
the number of circulating leukocytes (due to inhibition of situation [117].
13
2366 J Neurol (2017) 264:2351–2374
13
J Neurol (2017) 264:2351–2374 2367
the sequencing of these therapies might impact the overall European Academy of Neurology, European Committee for
PML risk of each patient. Treatment and Research in Multiple Sclerosis, and American
The immunization status of the patient also is important Academy of Neurology [132, 133]. Because both documents
because of interactions between some DMTs and vaccine are highly data driven, there is no recommendation provided
response. The National Multiple Sclerosis Society does for selecting one DMT over another. Instead, the appropri-
not recommend use of live vaccinations in people with MS ate choice of DMT is the one that the practicing neurologist
[121], and respective product labels for teriflunomide, fin- rationalizes will provide the level of efficacy warranted by
golimod, daclizumab beta, or alemtuzumab advise avoiding the recent disease activity, balanced by patient safety and
use of live attenuated vaccines during and for prespecified preferences.
time periods after stopping therapy [13, 72, 82, 100]. No The long-term immunologic and safety risks of sequenc-
product-specific information is available on the effects of ing multiple therapies are still unknown. Prescribing DMTs
vaccination in patients receiving peginterferon beta-1a, glati- in RMS depends on a thorough benefit–risk analysis, which
ramer acetate, dimethyl fumarate, and natalizumab. There- is inconclusive if the patient’s characteristics are not reflec-
fore, complete or partial prevention of infection by influenza, tive of clinical trial populations, and if the long-term effects
tuberculosis, varicella zoster virus, and hepatitis A and B of DMT in the clinical practice setting are unknown. Pro-
may be considered by vaccination before starting immu- spective industry-sponsored switching studies, patient reg-
nomodulatory therapy. Availability of non-live vaccines, istries, and robust analysis of real-world data are needed
such as the herpes zoster subunit vaccine (HZ/su) contain- to collect data tailored to the therapeutic agent and vari-
ing recombinant varicella zoster virus glycoprotein E and ous patient scenarios. Until such evidence-based medical
the AS01B adjuvant system [122], may represent a welcome information is available, decisions on sequencing DMTs for
and more flexible addition to efforts to prevent infection in RMS will depend heavily on the clinical acumen of the neu-
patients with MS receiving DMTs. rologist. In the meantime, sequencing the most appropriate
For elderly patients, a potential benefit–risk consideration therapies for patients with RMS is usually determined by
for the DMTs dimethyl fumarate, teriflunomide, fingolimod, a combination of factors such as disease activity, patient-
alemtuzumab, and ocrelizumab is their variable effect on related factors, and drug-related factors (e.g., pharmacody-
lymphocyte counts, which occur as a consequence of their namic profile).
on- or off-target pharmacology (Table 2). In elderly patients, Treatment should be selected to address the immediate
age-related immunosenescence, characterized by diminished clinical issue, and to keep alternative therapeutic options
levels and functionalities of B and T lymphocytes, may lead available for later-line therapies. This consideration is
to a theoretically greater likelihood of lymphopenia with particularly important early on in the disease course and
these DMTs [123]. One also could hypothesize that elderly even more relevant in today’s therapeutic landscape, which
patients would be less likely to experience breakthrough MS includes DMT options with potentially long-lasting effects
activity for the same reason. Hence, older age affects the on the immune system that can persist for months or even
benefit–risk ratio of DMTs and acts as a prompt for neu- years following discontinuation of therapy. Patients should
rologists to consider hematologic monitoring when making be made aware of these issues so that a shared care decision
prescribing decisions. can be reached, which is driven by matching the level of risk
Progress toward development of pharmacogenetics- and a patient is willing to accept with their prognostic factors.
biomarker-based approaches to individualize treatments
according to patient and DMT characteristics is in its infancy Acknowledgements Biogen provided funding for medical writing
support in the development of this paper; Malcolm Darkes, Ph.D.,
[124, 125]. In the meantime, other factors on which to base and Juliet Bell, Ph.D., from Excel Scientific Solutions wrote and
these decisions include patient preferences, lifestyle and updated the manuscript drafts based on input from the authors, and
beliefs, comorbidities and concomitant medications, immu- Kristen DeYoung from Excel Scientific Solutions copyedited and
nization status, family planning, and age. The first three styled the manuscript per journal requirements. Biogen reviewed
and provided feedback on the paper to the authors. The authors had
factors have a profound influence on adherence to medica- full editorial control of the paper and provided their final approval
tion; poor adherence predisposes the patient to suboptimal of all content.
clinical, neuroradiologic, health-related quality of life, and
pharmacoeconomic outcomes [126–131]. Compliance with ethical standards
13
2368 J Neurol (2017) 264:2351–2374
Genzyme, and received research or salary support from Biogen, the of subcutaneous interferon beta-1a with glatiramer acetate in
Consortium of Multiple Sclerosis Centers, and the National Multiple patients with relapsing multiple sclerosis (the REbif vs Glati-
Sclerosis Society over the past 2 years. ramer Acetate in Relapsing MS Disease [REGARD] study): a
multicentre, randomised, parallel, open-label trial. Lancet Neurol
Open Access This article is distributed under the terms of the 7:903–914. doi:10.1016/S1474-4422(08)70200-X
Creative Commons Attribution 4.0 International License (http://crea- 15. Limmroth V, Malessa R, Zettl UK, Koehler J, Japp G, Haller P,
tivecommons.org/licenses/by/4.0/), which permits unrestricted use, QUASIMS Study Group et al (2007) Quality Assessment in Mul-
distribution, and reproduction in any medium, provided you give appro- tiple Sclerosis Therapy (QUASIMS): a comparison of interferon
priate credit to the original author(s) and the source, provide a link to beta therapies for relapsing-remitting multiple sclerosis. J Neurol
the Creative Commons license, and indicate if changes were made. 254:67–77. doi:10.1007/s00415-006-0281-1
16. Yadav V, Bourdette D (2012) New disease-modifying ther-
apies and new challenges for MS. Curr Neurol Neurosci Rep
12:489–491. doi:10.1007/s11910-012-0295-2
References 17. Goodin DS, Ebers GC, Cutter G, Cook SD, O’Donnell T, Reder
AT et al (2012) Cause of death in MS: long-term follow-up of a
1. Compston A, Coles A (2008) Multiple sclerosis. Lancet randomised cohort, 21 years after the start of the pivotal IFNbeta-
372:1502–1517. doi:10.1016/S0140-6736(08)61620-7 1b study. BMJ Open 30:2. doi:10.1136/bmjopen-2012-001972
2. Noseworthy JH, Lucchinetti C, Rodriguez M, Weinshenker 18. Du Pasquier RA, Pinschewer DD, Merkler D (2014) Immuno-
BG (2000) Multiple sclerosis. N Engl J Med 343:938–952. logical mechanism of action and clinical profile of disease-mod-
doi:10.1056/NEJM200009283431307 ifying treatments in multiple sclerosis. CNS Drugs 28:535–558.
3. Barten LJ, Allington DR, Procacci KA, Rivey MP (2010) New doi:10.1007/s40263-014-0160-8
approaches in the management of multiple sclerosis. Drug Des 19. European Medicines Agency (2006) Tysabri 300 mg concen-
Devel Ther 4:343–366. doi:10.2147/DDDT.S9331 trate for solution for infusion [summary of product characteris-
4. Muñoz-Culla M, Irizar H, Otaegui D (2013) The genetics of tics]. http://www.ema.europa.eu/docs/en_GB/document_library/
multiple sclerosis: review of current and emerging candidates. EPAR_-_Product_Information/human/000603/WC500044686.
Appl Clin Genet 6:63–73. doi:10.2147/TACG.S29107 pdf. Accessed 15 April 2016
5. Sawcer S, Hellenthal G, Pirinen M, Spencer CC, Patsopoulos 20. Biogen (2016) Tysabri [prescribing information]. Biogen, Cam-
NA, Moutsianas L, International Multiple Sclerosis Genet- bridge, MA, USA
ics Consortium, Wellcome Trust Case Control Consortium 2 21. Spelman T, Kalincik T, Jokubaitis V, Zhang A, Pellegrini F,
et al (2011) Genetic risk and a primary role for cell-mediated Wiendl H et al (2016) Comparative efficacy of first-line natali-
immune mechanisms in multiple sclerosis. Nature 476:214–219. zumab vs IFN-β or glatiramer acetate in relapsing MS. Neurol
doi:10.1038/nature10251 Clin Pract 6:102–115. doi:10.1212/cpj.0000000000000227
6. Lucchinetti C, Bruck W, Parisi J, Scheithauer B, Rodriguez M, 22. Institute for Clinical and Economic Review (2017) Disease-mod-
Lassmann H (2000) Heterogeneity of multiple sclerosis lesions: ifying therapies for relapsing-remitting and primary-progressive
implications for the pathogenesis of demyelination. Ann Neurol multiple sclerosis: effectiveness and value. https://icer-review.
47:707–717. doi:10.1002/1531-8249(200006)47:6<707:AID- org/. Accessed 4 July 2017
ANA3>3.0.CO;2-Q 23. European Medicines Agency (2016) Conditions or restrictions
7. Dos Passos GR, Sato DK, Becker J, Fujihara K (2016) Th17 cells with regard to the safe and effective use of the medicinal prod-
pathways in multiple sclerosis and neuromyelitis optica spec- uct to be implemented by the member states. http://www.ema.
trum disorders: pathophysiological and therapeutic implications. europa.eu/docs/en_GB/document_library/EPAR_-_Condi-
Mediators Inflamm 2016:5314541. doi:10.1155/2016/5314541 tions_imposed_on_member_states_for_safe_and_effective_use/
8. Lovett-Racke AE, Yang Y, Racke MK (2011) Th1 versus Th17: human/000603/WC500044687.pdf. Accessed 21 Aug 2016
are T cell cytokines relevant in multiple sclerosis? Biochim Bio- 24. Ziemssen T, De Stefano N, Pia Sormani M, Van Wijmeersch B,
phys Acta 1812:246–251. doi:10.1016/j.bbadis.2010.05.012 Wiendl H, Kieseier BC (2015) Optimizing therapy early in mul-
9. Koch MW, Metz LM, Kovalchuk O (2013) Epigenetic changes tiple sclerosis: an evidence-based view. Mult Scler Relat Disord
in patients with multiple sclerosis. Nat Rev Neurol 9:35–43. 4:460–469. doi:10.1016/j.msard.2015.07.007
doi:10.1038/nrneurol.2012.226 25. Van Schependom J, Gielen J, Laton J, Nagels G (2016) Assess-
10. Disanto G, Berlanga AJ, Handel AE, Para AE, Burrell AM, Fries ing PML risk under immunotherapy: if all you have is a ham-
A et al (2010) Heterogeneity in multiple sclerosis: scratching the mer, everything looks like a nail. Mult Scler 22:389–392.
surface of a complex disease. Autoimmune Dis 2011:932351. doi:10.1177/1352458515596458
doi:10.4061/2011/932351 26. Berger JR (2017) Classifying PML risk with disease modify-
11. Lublin FD, Reingold SC, National Multiple Sclerosis Society ing therapies. Mult Scler Relat Disord 12:59–63. doi:10.1016/j.
(USA) Advisory Committee on Clinical Trials of New Agents in msard.2017.01.006
Multiple Sclerosis (1996) Defining the clinical course of multiple 27. Giovannoni G, Butzkueven H, Dhib-Jalbut S, Hobart J, Kobelt G,
sclerosis: results of an international survey. Neurology 46:907–911. Pepper G et al (2016) Brain health: time matters in multiple scle-
doi:10.1212/WNL.46.4.907 rosis. Mult Scler Relat Disord 9(suppl 1):S5–S48. doi:10.1016/j.
12. European Medicines Agency (2015) Gilenya 0.5 mg hard cap- msard.2016.07.003
sules [summary of product characteristics]. http://www.ema. 28. Rio J, Castillo J, Rovira A, Tintore M, Sastre-Garriga J, Horga
europa.eu/docs/en_GB/document_library/EPAR_-_Product_ A et al (2009) Measures in the first year of therapy predict the
Information/human/002202/WC500104528.pdf. Accessed 15 response to interferon beta in MS. Mult Scler 15:848–853.
April 2016 doi:10.1177/1352458509104591
13. Novartis (2016) Gilenya [prescribing information]. Novartis 29. Sormani MP, Rio J, Tintore M, Signori A, Li D, Cor-
Pharmaceuticals Corporation, East Hanover, NJ, USA nelisse P et al (2013) Scoring treatment response in patients
14. Mikol DD, Barkhof F, Chang P, Coyle PK, Jeffery DR, with relapsing multiple sclerosis. Mult Scler 19:605–612.
Schwid SR, REGARD study group et al (2008) Comparison doi:10.1177/1352458512460605
13
J Neurol (2017) 264:2351–2374 2369
30. Freedman MS, Selchen D, Arnold DL, Prat A, Banwell B, Yeung autologous hematopoietic cell transplantation for relapsing-
M, Canadian Multiple Sclerosis Working Group et al (2013) remitting multiple sclerosis (HALT-MS): a 3-year interim report.
Treatment optimization in MS: Canadian MS Working Group JAMA Neurol 72:159–169. doi:10.1001/jamaneurol.2014.3780
updated recommendations. Can J Neurol Sci 40:307–323 46. Berger B, Baumgartner A, Rauer S, Mader I, Luetzen N, Faren-
31. Stangel M, Penner IK, Kallmann BA, Lukas C, Kieseier BC kopf U, Stich O (2015) Severe disease reactivation in four
(2015) Towards the implementation of ‘no evidence of dis- patients with relapsing-remitting multiple sclerosis after fin-
ease activity’ in multiple sclerosis treatment: the multiple golimod cessation. J Neuroimmunol 282:118–122. doi:10.1016/j.
sclerosis decision model. Ther Adv Neurol Disord 8:3–13. jneuroim.2015.03.022
doi:10.1177/1756285614560733 47. Siva A, Uygunoglu U, Tutunca M, Altintas A, Saip S (2016)
32. Freedman MS, Cohen B, Dhib-Jalbut S, Jeffery D, Reder AT, Rebound of disease activity after fingolimod discontinuation:
Sandberg-Wollheim M, Weinstock-Guttman B (2009) Recogniz- report of six cases. Neurology 86(16 suppl):P2.081
ing and treating suboptimally controlled multiple sclerosis: steps 48. Hatcher SE, Waubant E, Nourbakhsh B, Crabtree-Hartman E,
toward regaining command. Curr Med Res Opin 25:2459–2470. Graves JS (2016) Rebound syndrome in patients with multiple
doi:10.1185/03007990903158364 sclerosis after cessation of fingolimod treatment. JAMA Neurol
33. Gajofatto A, Benedetti MD (2015) Treatment strategies for multi- 73:790–794. doi:10.1001/jamaneurol.2016.0826
ple sclerosis: when to start, when to change, when to stop? World 49. Clerico M, Schiavetti I, De Mercanti SF, Piazza F, Gned D,
J Clin Cases 3:545–555. doi:10.12998/wjcc.v3.i7.545 Brescia Morra V et al (2014) Treatment of relapsing-remitting
34. Ingwersen J, Aktas O, Hartung HP (2016) Advances in and algo- multiple sclerosis after 24 doses of natalizumab: evidence from
rithms for the treatment of relapsing-remitting multiple sclerosis. an Italian spontaneous, prospective, and observational study
Neurotherapeutics 13:47–57. doi:10.1007/s13311-015-0412-4 (the TY-STOP Study). JAMA Neurol 71:954–960. doi:10.1001/
35. Edan G, Le Page E (2013) Induction therapy for patients with jamaneurol.2014.1200
multiple sclerosis: why? When? How? CNS Drugs 27:403–409. 50. Fox RJ, Cree BA, De Sèze J, Gold R, Hartung HP, Jeffery D,
doi:10.1007/s40263-013-0065-y RESTORE et al (2014) MS disease activity in RESTORE: a ran-
36. Giovannoni G, Turner B, Gnanapavan S, Offiah C, Schmierer K, domized 24-week natalizumab treatment interruption study. Neu-
Marta M (2015) Is it time to target no evident disease activity rology 82:1491–1498. doi:10.1212/WNL.0000000000000355
(NEDA) in multiple sclerosis? Mult Scler Relat Disord 4:329–333. 51. Giovannoni G, Naismith RT (2014) Natalizumab to fingoli-
doi:10.1016/j.msard.2015.04.006 mod washout in patients at risk of PML: when good intentions
37. Coles AJ, Fox E, Vladic A, Gazda SK, Brinar V, Selmaj KW yield bad outcomes. Neurology 82:1196–1197. doi:10.1212/
et al (2012) Alemtuzumab more effective than interferon ß-1a WNL.0000000000000296
at 5-year follow-up of CAMMS223 clinical trial. Neurology 52. Kaufman M, Cree BA, De Sèze J, Fox RJ, Gold R, Hartung
78:1069–1078. doi:10.1212/WNL.0b013e31824e8ee7 HP et al (2015) Radiologic MS disease activity during natali-
38. Scalfari A, Neuhaus A, Degenhardt A, Rice GP, Muraro PA, zumab treatment interruption: findings from RESTORE. J Neurol
Daumer M, Ebers GC (2010) The natural history of multiple 262:326–336. doi:10.1007/s00415-014-7558-6
sclerosis: a geographically based study 10: relapses and long- 53. Kerbrat A, Le Page E, Leray E, Anani T, Coustans M, Des-
term disability. Brain 133(Pt 7):1914–1929. doi:10.1093/brain/ ormeaux C et al (2011) Natalizumab and drug holiday in clini-
awq118 cal practice: an observational study in very active relapsing
39. Klineova S, Nicholas J, Walker A (2012) Response to disease remitting multiple sclerosis patients. J Neurol Sci 308:98–102.
modifying therapies in African Americans with multiple sclero- doi:10.1016/j.jns.2011.05.043
sis. Ethn Dis 22:221–225 54. Giovannoni G, Radue EW, Havrdova E, Riester K, Green-
40. Rinaldi L, Perini P, Calabrese M, Gallo P (2009) Cyclophos- berg S, Mehta L, Elkins J (2014) Effect of daclizumab high-
phamide as second-line therapy in multiple sclerosis: benefits yield process in patients with highly active relapsing-remit-
and risks. Neurol Sci 30(suppl 2):S171–S173. doi:10.1007/ ting multiple sclerosis. J Neurol 261:316–323. doi:10.1007/
s10072-009-0145-4 s00415-013-7196-4
41. Vollmer T, Panitch H, Bar-Or A, Dunn J, Freedman MS, Gazda SK 55. Giovannoni G, Gold R, Selmaj K, Havrdova E, Montalban
et al (2008) Glatiramer acetate after induction therapy with mitox- X, Radue EW, SELECTION Study Investigators et al (2014)
antrone in relapsing multiple sclerosis. Mult Scler 14:663–670. Daclizumab high-yield process in relapsing-remitting multiple
doi:10.1177/1352458507085759 sclerosis (SELECTION): a multicentre, randomised, double-
42. Arnold DL, Campagnolo D, Panitch H, Bar-Or A, Dunn J, blind extension trial. Lancet Neurol 13:472–481. doi:10.1016/
Freedman MS et al (2008) Glatiramer acetate after mitoxantrone S1474-4422(14)70039-0
induction improves MRI markers of lesion volume and perma- 56. Lebrun C, Debouverie M, Vermersch P, Clavelou P, Rumbach
nent tissue injury in MS. J Neurol 255:1473–1478. doi:10.1007/ L, de Seze J et al (2008) Cancer risk and impact of disease-
s00415-008-0911-x modifying treatments in patients with multiple sclerosis. Mult
43. Atkins HL, Bowman M, Allan D, Anstee G, Arnold DL, Bar- Scler 14:399–405. doi:10.1177/1352458507083625
Or A et al (2016) Immunoablation and autologous haemopoi- 57. Hu X, Miller L, Richman S, Hitchman S, Glick G, Liu S et al
etic stem-cell transplantation for aggressive multiple sclerosis: (2012) A novel PEGylated interferon beta-1a for multiple scle-
a multicentre single-group phase 2 trial. Lancet 388:576–585. rosis: safety, pharmacology, and biology. J Clin Pharmacol
doi:10.1016/S0140-6736(16)30169-6 52:798–808. doi:10.1177/0091270011407068
44. Mancardi GL, Sormani MP, Gualandi F, Saiz A, Carreras E, 58. Damal K, Stoker E, Foley JF (2013) Optimizing therapeutics
Merelli E, ASTIMS Haemato-Neurological Collaborative Group in the management of patients with multiple sclerosis: a review
on behalf of the Autoimmune Disease Working Party (ADWP) of drug efficacy, dosing, and mechanisms of action. Biologics
of the European Group for Blood and Marrow Transplanta- 7:247–258. doi:10.2147/BTT.S53007
tion (EBMT) et al (2015) Autologous hematopoietic stem cell 59. Toker A, Slaney CY, Backstrom BT, Harper JL (2011) Glati-
transplantation in multiple sclerosis: a phase II trial. Neurology ramer acetate treatment directly targets CD11b+Ly6G− mono-
84:981–988. doi:10.1212/WNL.0000000000001329 cytes and enhances the suppression of autoreactive T cells in
45. Nash RA, Hutton GJ, Racke MK, Popat U, Devine SM, Grif- experimental autoimmune encephalomyelitis. Scand J Immunol
fith LM et al (2015) High-dose immunosuppressive therapy and 74:235–243. doi:10.1111/j.1365-3083.2011.02575.x
13
2370 J Neurol (2017) 264:2351–2374
60. Linker RA, Lee DH, Ryan S, van Dam AM, Conrad R, Bista P teriflunomide treatment for relapsing multiple sclerosis. Mult
et al (2011) Fumaric acid esters exert neuroprotective effects in Scler 21:1476–1477. doi:10.1177/1352458515596601
neuroinflammation via activation of the Nrf2 antioxidant path- 76. Gajofatto A, Turatti M, Monaco S, Benedetti MD (2015) Clinical
way. Brain 134:678–692. doi:10.1093/brain/awq386 efficacy, safety, and tolerability of fingolimod for the treatment
61. Scannevin RH, Chollate S, Jung MY, Shackett M, Patel H, Bista of relapsing-remitting multiple sclerosis. Drug Healthc Patient
P et al (2012) Fumarates promote cytoprotection of central nerv- Saf 7:157–167. doi:10.2147/DHPS.S69640
ous system cells against oxidative stress via the nuclear factor 77. Johnson TA, Evans BL, Durafourt BA, Blain M, Lapierre Y,
(erythroid-derived 2)-like 2 pathway. J Pharmacol Exp Ther Bar-Or A, Antel JP (2011) Reduction of the peripheral blood
341:274–284. doi:10.1124/jpet.111.190132 CD56bright NK lymphocyte subset in FTY720-treated multi-
62. Bomprezzi R (2015) Dimethyl fumarate in the treatment of ple sclerosis patients. J Immunol 187:570–579. doi:10.4049/
relapsing-remitting multiple sclerosis: an overview. Ther Adv jimmunol.1003823
Neurol Disord 8:20–30. doi:10.1177/1756285614564152 78. Johnson TA, Shames I, Keezer M, Lapierre Y, Haegert DG, Bar-
63. Fox RJ, Chan A, Gold R, Phillips JT, Selmaj K, Chang I et al Or A, Antel J (2010) Reconstitution of circulating lymphocyte
(2016) Characterizing absolute lymphocyte count profiles in counts in FTY720-treated MS patients. Clin Immunol 137:15–20.
dimethyl fumarate-treated patients with MS: patient manage- doi:10.1016/j.clim.2010.06.005
ment considerations. Neurol Clin Pract 6:220–229. doi:10.1212/ 79. Alroughani R, Almulla A, Lamdhade S, Thussu A (2014) Mul-
CPJ.0000000000000238 tiple sclerosis reactivation postfingolimod cessation: is it IRIS?
64. Buckle G, Bandari D, Greenstein J, Gudesblatt M, Khatri B, Kita BMJ Case Rep. doi:10.1136/bcr-2014-206314
M et al (2016) Effect of delayed-release dimethyl fumarate on 80. FDA Center for Drug Evaluation and Research (2016) Gilenya
lymphocyte subsets in patients with relapsing multiple sclero- (fingolimod) capsules 0.5 mg. Safety labeling changes. https://
sis: a retrospective, multicentre, observational study (REALIZE). wayback.archive-it.org/7993/20170112171747/http://www.
Mult Scler 22(3 suppl):EP1495 fda.gov/Safety/MedWatch/SafetyInformation/ucm266123.htm.
65. Spencer CM, Crabtree-Hartman EC, Lehmann-Horn K, Cree Accessed 20 April 2016
BA, Zamvil SS (2015) Reduction of CD8+ T lymphocytes in 81. European Medicines Agency (2016) Zinbryta 150 mg solution
multiple sclerosis patients treated with dimethyl fumarate. for injection in pre-filled syringe, Zinbryta 150 mg solution for
Neurol Neuroimmunol Neuroinflamm 2:e76. doi:10.1212/ injection in pre-filled pen [summary of product characteristics].
NXI.0000000000000076 http://www.ema.europa.eu/docs/en_GB/document_library/
66. Longbrake EE, Naismith RT, Parks BJ, Wu GF, Cross AH (2015) EPAR_-_Product_Information/human/003862/WC500210598.
Dimethyl fumarate-associated lymphopenia: risk factors and pdf. Accessed 20 July 2017
clinical significance [published online 31 July 2015]. Mult Scler 82. Biogen (2017) Zinbryta [prescribing information]. Biogen, Cam-
J Exp Transl Clin. doi:10.1177/2055217315596994 bridge, MA, USA
67. Biogen (2017) Tecfidera [prescribing information]. Biogen, Cam- 83. Therapeutic Goods Administration (2017) Prescription medi-
bridge, MA, USA cines: registration of new chemical entities in Australia, 2016.
68. Rosenkranz T, Novas M, Terborg C (2015) PML in a patient with https://www.tga.gov.au/prescription-medicines-registration-new-
lymphocytopenia treated with dimethyl fumarate. N Engl J Med chemical-entities-australia-2016. Accessed 21 April 2017
372:1476–1478. doi:10.1056/NEJMc1415408 84. Bielekova B (2013) Daclizumab therapy for multiple sclerosis.
69. European Medicines Agency (2015) Updated recommenda- Neurotherapeutics 10:55–67. doi:10.1007/s13311-012-0147-4
tions to minimise the risk of the rare brain infection PML with 85. Malek TR (2008) The biology of interleukin-2. Annu Rev Immu-
Tecfidera. http://www.ema.europa.eu/docs/en_GB/document_ nol 26:453–479. doi:10.1146/annurev.immunol.26.021607.
library/Press_release/2015/10/WC500196017.pdf. Accessed 7 090357
April 2016 86. Milo R (2014) The efficacy and safety of daclizumab and its
70. Zeyda M, Poglitsch M, Geyeregger R, Smolen JS, Zlabinger GJ, potential role in the treatment of multiple sclerosis. Ther Adv
Hörl WH et al (2005) Disruption of the interaction of T cells Neurol Disord 7:7–21. doi:10.1177/1756285613504021
with antigen-presenting cells by the active leflunomide metabolite 87. Wiendl H, Gross CC (2013) Modulation of IL-2Rα with dacli-
teriflunomide: involvement of impaired integrin activation and zumab for treatment of multiple sclerosis. Nat Rev Neurol
immunologic synapse formation. Arthritis Rheum 52:2730–2739. 9:394–404. doi:10.1038/nrneurol.2013.95
doi:10.1002/art.21255 88. Martin JF, Perry JS, Jakhete NR, Wang X, Bielekova B (2010)
71. Comi G, Freedman MS, Kappos L, Olsson TP, Miller AE, Wolin- An IL-2 paradox: blocking CD25 on T cells induces IL-2–driven
sky JS et al (2016) Pooled safety and tolerability data from four activation of C D56bright NK cells. J Immunol 185:1311–1320.
placebo-controlled teriflunomide studies and extensions. Mult doi:10.4049/jimmunol.0902238
Scler Relat Disord 5:97–104. doi:10.1016/j.msard.2015.11.006 89. Perry JS, Han S, Xu Q, Herman ML, Kennedy LB, Csako G,
72. Genzyme (2016) Aubagio [prescribing information]. Genzyme Bielekova B (2012) Inhibition of LTi cell development by CD25
Corporation, Cambridge, MA, USA blockade is associated with decreased intrathecal inflammation
73. European Medicines Agency (2013) Aubagio 14 mg film-coated in multiple sclerosis. Sci Transl Med 4:145ra106. doi:10.1126/
tablets [summary of product characteristics]. http://www.ema. scitranslmed.3004140
europa.eu/docs/en_GB/document_library/EPAR_-_Product_ 90. Bielekova B, Catalfamo M, Reichert-Scrivner S, Packer A,
Information/human/002514/WC500148682.pdf. Accessed 18 Cerna M, Waldmann TA et al (2006) Regulatory CD56bright nat-
July 2016 ural killer cells mediate immunomodulatory effects of IL-2Rα-
74. FDA Center for Drug Evaluation and Research (2016) Teriflu- targeted therapy (daclizumab) in multiple sclerosis. Proc Natl
nomide (Aubagio). Safety labelling changes. https://wayback. Acad Sci USA 103:5941–5946. doi:10.1073/pnas.0601335103
archive-it.org/7993/20161023083238/http://www.fda.gov/Safety/ 91. Bielekova B, Howard T, Packer AN, Richert N, Blevins G,
MedWatch/SafetyInformation/ucm423101.htm. Accessed 22 Ohayon J et al (2009) Effect of anti-CD25 antibody daclizumab
Aug 2016 in the inhibition of inflammation and stabilization of disease
75. Gerschenfeld G, Servy A, Valeyrie-Allanore L, de Prost N, Cec- progression in multiple sclerosis. Arch Neurol 66:483–489.
chini J (2015) Fatal toxic epidermal necrolysis in a patient on doi:10.1001/archneurol.2009.50
13
J Neurol (2017) 264:2351–2374 2371
92. Gold R, Giovannoni G, Selmaj K, Havrdova E, Montalban X, reversible by 16–20 weeks after treatment discontinuation. Neu-
Radue EW, SELECT study Investigators et al (2013) Daclizumab rology 86(16 suppl):P5.408
high-yield process in relapsing-remitting multiple sclerosis 108. Bloomgren G, Richman S, Hotermans C, Subramanyam M,
(SELECT): a randomised, double-blind, placebo-controlled trial. Goelz S, Natarajan A et al (2012) Risk of natalizumab-associ-
Lancet 381:2167–2175. doi:10.1016/S0140-6736(12)62190-4 ated progressive multifocal leukoencephalopathy. N Engl J Med
93. Amaravadi L, Mokliatchouk O, Mehta D, Riester K, Sheridan 366:1870–1880. doi:10.1056/NEJMoa1107829
J, Elkins J (2015) Early, sustained, and reversible pharmacody- 109. Koendgen H, Chang I, Sperling B, Bloomgren G, Richman S,
namic effects of daclizumab HYP in MS support mechanism of Ho PR, Campbell N (2016) New algorithm to estimate risk of
action via modulation of the IL-2 pathway. Neurology 84(14 natalizumab-associated progressive multifocal leukoencepha-
suppl):P1.149 lopathy (PML) in anti-JCV antibody positive patients: analyses
94. Huss DJ, Mehta DS, Sharma A, You X, Riester KA, Sheridan of clinical trial data to provide further temporal precision and
JP et al (2015) In vivo maintenance of human regulatory T cells inform clinical practice. Mult Scler 22(3 suppl):P1249
during CD25 blockade. J Immunol 194:84–92. doi:10.4049/ 110. Plavina T, Subramanyam M, Bloomgren G, Richman S, Pace
jimmunol.1402140 A, Lee S et al (2014) Anti-JC virus antibody levels in serum or
95. Fam S, Mokliatchouk O, Mehta D, Riester K, Sheridan S, plasma further define risk of natalizumab-associated progres-
McCroskery P, Elkins J (2016) Reversible effects of daclizumab sive multifocal leukoencephalopathy. Ann Neurol 76:802–812.
HYP on lymphocyte counts in RRMS patients: data from the doi:10.1002/ana.24286
SELECT trilogy studies. Neurology 86(16 suppl):P5.281 111. Kuesters G, Plavina T, Lee S, Campagnolo D, Richman S,
96. Diao L, Hang Y, Othman AA, Nestorov I, Tran JQ (2016) Popu- Belachew S, Subramanyam M (2015) Anti-JC virus (JCV) anti-
lation pharmacokinetics of daclizumab high-yield process in body index differentiates risk of progressive multifocal leukoen-
healthy volunteers and subjects with multiple sclerosis: analysis cephalopathy (PML) in natalizumab-treated multiple sclerosis
of phase I-III clinical trials. Clin Pharmacokinet 55:943–955. (MS) patients with no prior immunosuppressant (IS) use: an
doi:10.1007/s40262-016-0366-7 updated analysis. Neurology 84(14 suppl):P4.031
97. Kappos L, Cohan S, Arnold DL, Mokliatchouk O, Greenberg SJ, 112. Gobbi C, Meier DS, Cotton F, Sintzel M, Leppert D, Guttmann
McCroskery P, Lima G (2016) Interim report on the safety and CR, Zecca C (2013) Interferon beta 1b following natalizumab
efficacy of long-term daclizumab HYP treatment for up to 5 years discontinuation: one year, randomized, prospective, pilot trial.
in EXTEND. Mult Scler 22(3 suppl):P653 BMC Neurol 13:101. doi:10.1186/1471-2377-13-101
98. Giovannoni G, Kappos L, Gold R, Khatri BO, Selmaj K, Umans 113. Cohen M, Maillart E, Tourbah A, De Seze J, Vukusic S, Brassat
K et al (2016) Safety and tolerability profile of daclizumab in D, Club Francophone de la Sclerose en Plaques Investigators et al
patients with relapsing-remitting multiple sclerosis: an integrated (2014) Switching from natalizumab to fingolimod in multiple
analysis of clinical studies. Mult Scler Relat Disord 9:36–46. sclerosis: a French prospective study. JAMA Neurol 71:436–441.
doi:10.1016/j.msard.2016.05.010 doi:10.1001/jamaneurol.2013.6240
99. Babij R, Perumal JS (2015) Comparative efficacy of alemtu- 114. Malucchi S, Capobianco M, Lo Re M, Malentacchi M, di
zumab and established treatment in the management of multiple Sapio A, Matta M et al (2016) High-risk PML patients switch-
sclerosis. Neuropsychiatr Dis Treat 11:1221–1229. doi:10.2147/ ing from natalizumab to alemtuzumab: an observational study
NDT.S60518 [published online 3 December 2016]. Neurol Ther. doi:10.1007/
100. Genzyme (2016) Lemtrada [prescribing information]. Genzyme s40120-016-0058-0
Corporation, Cambridge, MA, USA 115. Alping P, Frisell T, Novakova L, Islam-Jakobsson P, Salzer J,
101. Hill-Cawthorne GA, Button T, Tuohy O, Jones JL, May K, Bjorck A et al (2016) Rituximab versus fingolimod after natali-
Somerfield J et al (2012) Long term lymphocyte reconstitution zumab in multiple sclerosis patients. Ann Neurol 79:950–958.
after alemtuzumab treatment of multiple sclerosis. J Neurol Neu- doi:10.1002/ana.24651
rosurg Psychiatry 83:298–304. doi:10.1136/jnnp-2011-300826 116. Alvarez E, Vollmer B, Jace B, Corboy J, Vollmer T, Sillou
102. Thompson SA, Jones JL, Cox AL, Compston DA, Coles AJ SH et al (2015) Effectiveness of switching to rituximab over
(2010) B-cell reconstitution and BAFF after alemtuzumab fingolimod or dimethyl fumarate after natalizumab in prevent-
(Campath-1H) treatment of multiple sclerosis. J Clin Immunol ing disease activity in multiple sclerosis. Neurology 84(14
30:99–105. doi:10.1007/s10875-009-9327-3 suppl):P3.288
103. Durelli L, De Mercanti S, Rolla S, Cucci A, Bardina V, Cocco E 117. Maillart E, Vidal JS, Brassat D, Stankoff B, Fromont A, de Seze
et al (2016) Alemtuzumab long term immunological study: the J et al (2017) Natalizumab-PML survivors with subsequent MS
immunosuppressive effect does not last more than 48 months. treatment: clinico-radiologic outcome. Neurol Neuroimmunol
Neurology 86(S2):008 Neuroinflamm 4:e346. doi:10.1212/NXI.0000000000000346
104. Cossburn MD, Harding K, Ingram G, El-Shanawany T, Heaps A, 118. Genentech (2017) Ocrevus [prescribing information]. Genentech,
Pickersgill TP et al (2013) Clinical relevance of differential lym- Inc., South San Francisco, CA, USA
phocyte recovery after alemtuzumab therapy for multiple sclero- 119. Sorensen PS, Blinkenberg M (2016) The potential role for
sis. Neurology 80:55–61. doi:10.1212/WNL.0b013e31827b5927 ocrelizumab in the treatment of multiple sclerosis: current evi-
105. Coles AJ, Twyman CL, Arnold DL, Cohen JA, Confavreux C, dence and future prospects. Ther Adv Neurol Disord 9:44–52.
Fox EJ, CARE-MS II Investigators et al (2012) Alemtuzumab for doi:10.1177/1756285615601933
patients with relapsing multiple sclerosis after disease-modify- 120. Hauser SL, Bar-Or A, Comi G, Giovannoni G, Hartung HP,
ing therapy: a randomised controlled phase 3 trial. Lancet 380: Hemmer B, OPERA I and OPERA II Clinical Investigators
1829–1839. doi:10.1016/S0140-6736(12)61768-1 et al (2017) Ocrelizumab versus interferon beta-1a in relapsing
106. Khatri BO, Man S, Giovannoni G, Koo AP, Lee JC, Tucky B multiple sclerosis. N Engl J Med 376:221–234. doi:10.1056/
et al (2009) Effect of plasma exchange in accelerating natali- NEJMoa1601277
zumab clearance and restoring leukocyte function. Neurology 121. National Multiple Sclerosis Society (2017) Vaccinations. http://
72:402–409. doi:10.1212/01.wnl.0000341766.59028.9d www.nationalmssociety.org/Living-Well-With-MS/Health-Well-
107. Plavina T, Muralidharan KK, Kuesters G, Mikol D, Evans K, ness/Vaccinations. Accessed 22 April 2017
Subramanyam M et al (2016) Natalizumab’s effects on periph- 122. Cunningham AL, Lal H, Kovac M, Chlibek R, Hwang SJ,
eral immune cells in patients with multiple sclerosis (MS) are Diez-Domingo J et al (2016) Efficacy of the herpes zoster
13
2372 J Neurol (2017) 264:2351–2374
subunit vaccine in adults 70 years of age or older. N Engl J Med 139. European Medicines Agency (2014) Tecfidera 120 mg gastro-
375:1019–1032. doi:10.1056/NEJMoa1603800 resistant hard capsules, Tecfidera 240 mg gastro-resistant hard
123. Gruver AL, Hudson LL, Sempowski GD (2007) Immunosenes- capsules [summary of product characteristics]. http://www.
cence of ageing. J Pathol 211:144–156. doi:10.1002/path.2104 ema.europa.eu/docs/en_GB/document_library/EPAR_-_Prod-
124. Axtell RC, de Jong BA, Boniface K, van der Voort LF, Bhat R, uct_Information/human/002601/WC500162069.pdf. Accessed
De Sarno P et al (2010) T helper type 1 and 17 cells determine 15 April 2016
efficacy of interferon-ß in multiple sclerosis and experimental 140. European Medicines Agency (2017) EMA restricts use of multi-
encephalomyelitis. Nat Med 16:406–412. doi:10.1038/nm.2110 ple sclerosis medicine Zinbryta. http://www.ema.europa.eu/docs/
125. Grossman I, Avidan N, Singer C, Goldstaub D, Hayardeny L, en_GB/document_library/Press_release/2017/07/WC500230922.
Eyal E et al (2007) Pharmacogenetics of glatiramer acetate pdf. Accessed 20 July 2017
therapy for multiple sclerosis reveals drug–response mark- 141. European Medicines Agency (2013) Lemtrada 12 mg concen-
ers. Pharmacogenet Genomics 17:657–666. doi:10.1097/ trate for solution for infusion [summary of product characteris-
FPC.0b013e3281299169 tics]. http://www.ema.europa.eu/docs/en_GB/document_library/
126. Steinberg SC, Faris RJ, Chang CF, Chan A, Tankers- EPAR_-_Product_Information/human/003718/WC500150521.
ley MA (2010) Impact of adherence to interferons in the pdf. Accessed 15 April 2016
treatment of multiple sclerosis: a non-experimental, ret- 142. EMD Serono (2008) Novantrone [prescribing information]. EMD
rospective, cohort study. Clin Drug Investig 30:89–100. Serono, Inc., Rockland, MA, USA
doi:10.2165/11533330-000000000-00000 143. von Hehn C, Howard J, Liu S, Meka V, Pultz J, Sheikh S (2016)
127. Devonshire V, Lapierre Y, Macdonell R, Ramo-Tello C, Patti An open-label study to assess the immune response to vacci-
F, Fontoura P, GAP Study Group et al (2011) The Global nation in patients with relapsing forms of multiple sclerosis
Adherence Project (GAP): a multicenter observational study treated with delayed-release dimethyl fumarate compared to
on adherence to disease-modifying therapies in patients with non-pegylated interferon. Mult Scler 22(3 suppl):P633
relapsing-remitting multiple sclerosis. Eur J Neurol 18:69–77. 144. Bar-Or A, Pachner A, Menguy-Vacheron F, Kaplan J, Wiendl H
doi:10.1111/j.1468-1331.2010.03110.x (2014) Teriflunomide and its mechanism of action in multiple
128. Cerghet M, Dobie E, Lafata J, Schultz L, Elias S, Pladevall M, sclerosis. Drugs 74:659–674. doi:10.1007/s40265-014-0212-x
Reuther J (2010) Adherence to disease-modifying agents and 145. Bar-Or A, Freedman MS, Kremenchutzky M, Menguy-Vacheron
association with quality of life among patients with relapsing- F, Bauer D, Jodl S et al (2013) Teriflunomide effect on immune
remitting multiple sclerosis. Int J MS Care 12:51–58 response to influenza vaccine in patients with multiple sclerosis.
129. Uitdehaag B, Constantinescu C, Cornelisse P, Jeffery D, Neurology 81:552–558. doi:10.1212/WNL.0b013e31829e6fbf
Kappos L, Li D et al (2011) Impact of exposure to interferon 146. Mandala S, Hajdu R, Bergstrom J, Quackenbush E, Xie J, Mil-
beta-1a on outcomes in patients with relapsing-remitting ligan J et al (2002) Alteration of lymphocyte trafficking by sphin-
multiple sclerosis: exploratory analyses from the PRISMS gosine-1-phosphate receptor agonists. Science 296:346–349.
long-term follow-up study. Ther Adv Neurol Disord 4:3–14. doi:10.1126/science.1070238
doi:10.1177/1756285610391693 147. Matloubian M, Lo CG, Cinamon G, Lesneski MJ, Xu Y, Brink-
130. Marrie RA, Hanwell H (2013) General health issues in mul- mann V et al (2004) Lymphocyte egress from thymus and periph-
tiple sclerosis: comorbidities, secondary conditions, and eral lymphoid organs is dependent on S1P receptor 1. Nature
health behaviors. Continuum (Minneap Minn) 19:1046–1057. 427:355–360. doi:10.1038/nature02284
doi:10.1212/01.CON.0000433284.07844.6b 148. Kappos L, Mehling M, Arroyo R, Izquierdo G, Selmaj K,
131. Marrie RA, Miller A, Sormani MP, Thompson A, Waubant Curovic-Perisic V et al (2015) Randomized trial of vaccination
E, Trojano M, Attendees of the International Workshop on in fingolimod-treated patients with multiple sclerosis. Neurology
Comorbidity in Multiple Sclerosis et al (2016) The challenge of 84:872–879. doi:10.1212/WNL.0000000000001302
comorbidity in clinical trials for multiple sclerosis. Neurology 149. Francis G, Kappos L, O’Connor P, Collins W, Tang D, Mercier
86:1437–1445. doi:10.1212/WNL.0000000000002471 F, Cohen JA (2014) Temporal profile of lymphocyte counts and
132. Otero-Romero S, Amato MP, Chandraratna D, Clanet M, Comi relationship with infections with fingolimod therapy. Mult Scler
G, Derfuss T et al (2016) ECTRIMS-EAN clinical practice 20:471–480. doi:10.1177/1352458513500551
guideline on pharmacological management of multiple sclerosis. 150. Mehta L, Umans K, Ozen G, Robinson RR, Elkins J (2017)
Mult Scler 22(3 suppl):255 Immune response to seasonal influenza vaccine in patients with
133. Rae-Grant A, Rabinstein A, Cree B, Dubinsky R, Day G, Haboubi relapsing-remitting multiple sclerosis receiving long-term dacli-
M et al (2015) Disease-modifying therapies for multiple sclerosis: zumab beta: a prospective, open-label, single-arm study. Int J MS
a guideline project protocol. https://www.aan.com/uploadedFiles/ Care 19:141–147. doi:10.7224/1537-2073.2016-026
Website_Library_Assets/Documents/2.Clinical_Guidelines/3. 151. Kaufman M, Pardo G, Rossman H, Sweetser MT, Forrestal
Browse_By_Status/4.Guidelines_Under_Development/Protocol- F, Duda P (2014) Natalizumab treatment shows no clinically
Draft-MS-DMT-7-9-15-Final.pdf. Accessed 19 Nov 2016 meaningful effects on immunization responses in patients with
134. Bayer (2016) Betaseron [prescribing information]. Bayer Health- relapsing-remitting multiple sclerosis. J Neurol Sci 341:22–27.
Care Pharmaceuticals Inc., Whippany, NJ, USA doi:10.1016/j.jns.2014.03.035
135. Biogen (2016) Avonex [prescribing information]. Biogen, Cam- 152. Sorensen PS (2014) New management algorithms in multi-
bridge, MA, USA ple sclerosis. Curr Opin Neurol 27:246–259. doi:10.1097/
136. Alwan S, Chambers CD, Armenti VT, Sadovnick AD (2015) WCO.0000000000000096
The need for a disease-specific prospective pregnancy registry 153. Bellosillo B, Colomer D, Pons G, Gil J (1998) Mitoxantrone,
for multiple sclerosis (MS). Mult Scler Relat Disord 4:6–17. a topoisomerase II inhibitor, induces apoptosis of B-chronic
doi:10.1016/j.msard.2014.10.001 lymphocytic leukaemia cells. Br J Haematol 100:142–146.
137. Biogen (2015) Plegridy [prescribing information]. Biogen, Cam- doi:10.1046/j.1365-2141.1998.00520.x
bridge, MA, USA 154. Gbadamosi J, Buhmann C, Tessmer W, Moench A, Haag F,
138. Teva (2016) Copaxone [prescribing information]. Teva Neurosci- Heesen C (2003) Effects of mitoxantrone on multiple sclero-
ence, Inc., Overland Park, KS, USA sis patients’ lymphocyte subpopulations and production of
13
J Neurol (2017) 264:2351–2374 2373
immunoglobulin, TNF-alpha and IL-10. Eur Neurol 49:137–141. 168. Kappos L, Wiendl H, Selmaj K, Arnold DL, Havrdova E, Boyko
doi:10.1159/000069082 A et al (2015) Daclizumab HYP versus interferon beta-1a in
155. The IFNB Multiple Sclerosis Study Group (1993) Interferon relapsing multiple sclerosis. N Engl J Med 373:1418–1428.
beta-1b is effective in relapsing-remitting multiple sclerosis. doi:10.1056/NEJMoa1501481
I. Clinical results of a multicenter, randomized, double-blind, 169. Coles AJ, Compston DA, Selmaj KW, Lake SL, Moran S, Mar-
placebo-controlled trial. Neurology 43:655–661. doi:10.1212/ golin DH, CAMMS223 Trial Investigators et al (2008) Alemtu-
WNL.43.4.655 zumab vs. interferon beta-1a in early multiple sclerosis. N Engl
156. Jacobs LD, Cookfair DL, Rudick RA, Herndon RM, Richert JR, J Med 359:1786–1801. doi:10.1056/NEJMoa0802670
Salazar AM et al (1996) Intramuscular interferon beta-1a for 170. Cohen JA, Coles AJ, Arnold DL, Confavreux C, Fox EJ, Har-
disease progression in relapsing multiple sclerosis. Ann Neurol tung HP, CARE-MS I investigators et al (2012) Alemtuzumab
39:285–294. doi:10.1002/ana.410390304 versus interferon beta 1a as first-line treatment for patients
157. Ebers GC, PRISMS (Prevention of Relapses and Disability by with relapsing-remitting multiple sclerosis: a randomised
Interferon beta-1a Subcutaneously in Multiple Sclerosis) Study controlled phase 3 trial. Lancet 380:1819–1828. doi:10.1016/
Group (1998) Randomised double-blind placebo-controlled S0140-6736(12)61769-3
study of interferon ß-1a in relapsing/remitting multiple sclerosis. 171. Polman CH, O’Connor PW, Havrdova E, Hutchinson M, Kap-
Lancet 352:1498–1504. doi:10.1016/S0140-6736(98)03334-0 pos L, Miller DH, AFFIRM Investigators et al (2006) A ran-
158. European Medicines Agency (2015) Rebif 22 micrograms solu- domized, placebo-controlled trial of natalizumab for relapsing
tion [summary of product characteristics]. http://www.ema. multiple sclerosis. N Engl J Med 354:899–910. doi:10.1056/
europa.eu/docs/en_GB/document_library/EPAR_-_Product_ NEJMoa044397
Information/human/000136/WC500048681.pdf. Accessed 20 172. Hartung HP, Gonsette R, Konig N, Kwiecinski H, Guseo A,
July 2017 Morrissey SP, Mitoxantrone in Multiple Sclerosis Study Group
159. European Medicines Agency (2014) Plegridy 63 micrograms (MIMS) (2002) Mitoxantrone in progressive multiple sclerosis: a
and 94 micrograms solution [summary of product characteris- placebo-controlled, double-blind, randomised, multicentre trial.
tics]. http://www.ema.europa.eu/docs/en_GB/document_library/ Lancet 360:2018–2025. doi:10.1016/S0140-6736(02)12023-X
EPAR_-_Product_Information/human/002827/WC500170302. 173. Martinelli Boneschi F, Vacchi L, Rovaris M, Capra R, Comi G
pdf. Accessed 20 July 2017 (2013) Mitoxantrone for multiple sclerosis. Cochrane Database
160. Johnson KP, Brooks BR, Cohen JA, Ford CC, Goldstein J, Lisak Syst Rev 5:CD002127. doi:10.1002/14651858.CD002127.pub3
RP, Copolymer 1 Multiple Sclerosis Study Group et al (1995) 174. Amato MP, Portaccio E, Goretti B, Zipoli V, Battaglini M,
Copolymer 1 reduces relapse rate and improves disability in Bartolozzi ML et al (2007) Association of neocortical volume
relapsing-remitting multiple sclerosis: results of a phase III changes with cognitive deterioration in relapsing-remitting
multicenter, double-blind placebo-controlled trial. Neurology multiple sclerosis. Arch Neurol 64:1157–1161. doi:10.1001/
45:1268–1276. doi:10.1212/WNL.45.7.1268 archneur.64.8.1157
161. Fox RJ, Miller DH, Phillips JT, Hutchinson M, Havrdova E, 175. Popescu V, Agosta F, Hulst HE, Sluimer IC, Knol DL, Sormani
Kita M, CONFIRM Study Investigators et al (2012) Placebo- MP, MAGNIMS Study Group et al (2013) Brain atrophy and
controlled phase 3 study of oral BG-12 or glatiramer in mul- lesion load predict long term disability in multiple sclerosis.
tiple sclerosis. N Engl J Med 367:1087–1097. doi:10.1056/ J Neurol Neurosurg Psychiatry 84:1082–1091. doi:10.1136/
NEJMoa1206328 jnnp-2012-304094
162. Viglietta V, Miller D, Bar-Or A, Phillips JT, Arnold DL, Selmaj 176. Fisher E, Rudick RA, Cutter G, Baier M, Miller D, Weinstock-
K et al (2015) Efficacy of delayed-release dimethyl fumarate in Guttman B et al (2000) Relationship between brain atrophy and
relapsing-remitting multiple sclerosis: integrated analysis of the disability: an 8-year follow-up study of multiple sclerosis patients.
phase 3 trials. Ann Clin Transl Neurol 2:103–118. doi:10.1002/ Mult Scler 6:373–377. doi:10.1177/135245850000600602
acn3.148 177. Jacobsen C, Hagemeier J, Myhr KM, Nyland H, Lode K,
163. O’Connor P, Wolinsky JS, Confavreux C, Comi G, Kappos L, Bergsland N et al (2014) Brain atrophy and disability progres-
Olsson TP, TEMSO Trial Group et al (2011) Randomized trial sion in multiple sclerosis patients: a 10-year follow-up study.
of oral teriflunomide for relapsing multiple sclerosis. N Engl J J Neurol Neurosurg Psychiatry 85:1109–1115. doi:10.1136/
Med 365:1293–1303. doi:10.1056/NEJMoa1014656 jnnp-2013-306906
164. Confavreux C, O’Connor P, Comi G, Freedman MS, Miller AE, 178. Lavorgna L, Bonavita S, Ippolito D, Lanzillo R, Salemi G, Patti F
Olsson TP, TOWER Trial Group et al (2014) Oral teriflunomide et al (2014) Clinical and magnetic resonance imaging predictors
for patients with relapsing multiple sclerosis (TOWER): a ran- of disease progression in multiple sclerosis: a nine-year follow-up
domised, double-blind, placebo-controlled, phase 3 trial. Lancet study. Mult Scler 20:220–226. doi:10.1177/1352458513494958
Neurol 13:247–256. doi:10.1016/S1474-4422(13)70308-9 179. Mowry EM, Beheshtian A, Waubant E, Goodin DS, Cree BA,
165. Kappos L, Radue EW, O’Connor P, Polman C, Hohlfeld R, Qualley P et al (2009) Quality of life in multiple sclerosis is asso-
Calabresi P, FREEDOMS Study Group et al (2010) A placebo- ciated with lesion burden and brain volume measures. Neurology
controlled trial of oral fingolimod in relapsing multiple sclerosis. 72:1760–1765. doi:10.1212/WNL.0b013e3181a609f8
N Engl J Med 362:387–401. doi:10.1056/NEJMoa0909494 180. Miller JR (2004) The importance of early diagnosis of multiple
166. Calabresi PA, Radue EW, Goodin D, Jeffery D, Rammohan sclerosis. J Manag Care Pharm 10(3 suppl B):S4–S11
KW, Reder AT et al (2014) Safety and efficacy of fingoli- 181. Rotstein DL, Healy BC, Malik MT, Chitnis T, Weiner HL (2015)
mod in patients with relapsing-remitting multiple sclerosis Evaluation of no evidence of disease activity in a 7-year lon-
(FREEDOMS II): a double-blind, randomised, placebo-con- gitudinal multiple sclerosis cohort. JAMA Neurol 72:152–158.
trolled, phase 3 trial. Lancet Neurol 13:545–556. doi:10.1016/ doi:10.1001/jamaneurol.2014.3537
S1474-4422(14)70049-3 182. Langer-Gould A, Popat RA, Huang SM, Cobb K, Fontoura P,
167. Cohen JA, Barkhof F, Comi G, Hartung HP, Khatri BO, Montal- Gould MK, Nelson LM (2006) Clinical and demographic pre-
ban X, TRANSFORMS Study Group et al (2010) Oral fingoli- dictors of long-term disability in patients with relapsing-remit-
mod or intramuscular interferon for relapsing multiple sclerosis. ting multiple sclerosis: a systematic review. Arch Neurol 63:
N Engl J Med 362:402–415. doi:10.1056/NEJMoa0907839 1686–1691. doi:10.1001/archneur.63.12.1686
13
2374 J Neurol (2017) 264:2351–2374
183. Runmarker B, Andersen O (1993) Prognostic factors in a multiple 189. Cossburn M, Pace AA, Jones J, Ali R, Ingram G, Baker K
sclerosis incidence cohort with twenty-five years of follow-up. et al (2011) Autoimmune disease after alemtuzumab treatment
Brain 116(Pt 1):117–134. doi:10.1093/brain/116.1.117 for multiple sclerosis in a multicenter cohort. Neurology 77:
184. Salzer J, Svenningsson A, Sundstrom P (2010) Neurofilament 573–579. doi:10.1212/WNL.0b013e318228bec5
light as a prognostic marker in multiple sclerosis. Mult Scler 190. Daniels GH, Vladic A, Brinar V, Zavalishin I, Valente W,
16:287–292. doi:10.1177/1352458509359725 Oyuela P et al (2014) Alemtuzumab-related thyroid dysfunc-
185. Sola P, Mandrioli J, Simone AM, Ferraro D, Bedin R, Annecca tion in a phase 2 trial of patients with relapsing-remitting mul-
R et al (2011) Primary progressive versus relapsing-onset tiple sclerosis. J Clin Endocrinol Metab 99:80–89. doi:10.1210/
multiple sclerosis: presence and prognostic value of cer- jc.2013-2201
ebrospinal fluid oligoclonal IgM. Mult Scler 17:303–311. 191. Penkert H, Delbridge C, Wantia N, Wiestler B, Korn T (2016)
doi:10.1177/1352458510386996 Fulminant central nervous system nocardiosis in a patient treated
186. Tintore M, Rovira A, Rio J, Otero-Romero S, Arrambide G, Tur with alemtuzumab for relapsing-remitting multiple sclerosis.
C et al (2015) Defining high, medium and low impact prognostic JAMA Neurol 73:757–759. doi:10.1001/jamaneurol.2016.0146
factors for developing multiple sclerosis. Brain 138:1863–1874. 192. Senior P, Arnold D, Cohen J, Fox E, Hartung H-P, Havrdova E
doi:10.1093/brain/awv105 et al (2016) Incidence and timing of thyroid adverse events in
187. Theodore Phillips J, Erwin AA, Agrella S, Kremenchutzky M, patients with RRMS treated with alemtuzumab through 5 years
Kramer JF, Darkes MJ et al (2015) Consensus management of of the CARE-MS studies. Neurology 86(16 suppl):P2.086
gastrointestinal events associated with delayed-release dimethyl 193. Hillen ME, Cook SD, Samanta A, Grant E, Quinless JR, Rajas-
fumarate: a Delphi study. Neurol Ther 4:137–146. doi:10.1007/ ingham JK (2015) Fatal acute liver failure with hepatitis B virus
s40120-015-0037-x infection during natalizumab treatment in multiple sclerosis.
188. European Medicines Agency (2016) Scientific conclusions and Neurol Neuroimmunol Neuroinflamm 2:e72. doi:10.1212/
grounds for the variation to the terms of the marketing authorisa- NXI.0000000000000072
tion. http://www.ema.europa.eu/docs/en_GB/document_library/ 194. Shenoy ES, Mylonakis E, Hurtado RM, Venna N (2011)
EPAR_-_Scientific_Conclusion/human/003718/WC500209905. Natalizumab and HSV meningitis. J Neurovirol 17:288–290.
pdf. Accessed 14 July 2016 doi:10.1007/s13365-011-0027-4
13