Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Reviews

Epidemiology and treatment of


multiple sclerosis in elderly populations
Caila B. Vaughn    1, Dejan Jakimovski   2, Katelyn S. Kavak1, Murali Ramanathan3,
Ralph H. B. Benedict1, Robert Zivadinov2,4 and Bianca Weinstock-​Guttman    1*
Abstract | The prevalence of multiple sclerosis (MS) and the age of affected patients are
increasing owing to increased longevity of the general population and the availability of effective
disease-​modifying therapies. However, ageing presents unique challenges in patients with MS
largely as a result of their increased frequency of age-​related and MS-​related comorbidities as
well as transition of the disease course from an inflammatory to a neurodegenerative phenotype.
Immunosenescence (the weakening of the immune system associated with natural ageing) might
be at least partly responsible for this transition, which further complicates disease management.
Currently approved therapies for MS are effective in preventing relapse but are not as effective
in preventing the accumulation of disability associated with ageing and disease progression.
Thus, ageing patients with MS represent a uniquely challenging population that is currently
underserved by existing therapeutic regimens. This Review focuses on the epidemiology of MS in
ageing patients. Unique considerations relevant to this population are discussed, including the
immunology and pathobiology of the complex relationship between ageing and MS, the safety
and efficacy of disease-​modifying therapies, when discontinuation of treatment might be
appropriate and the important role of approaches to support wellness and cognition.

Worldwide, the average age of individuals with multi- (termed very-​late-onset MS (VLOMS)). Therefore,
1
Jacobs Multiple Sclerosis ple sclerosis (MS) is increasing1. Advances in treating managing MS in elderly populations presents unique
Center for Treatment and vascular comorbidities, as well as the large armamentar- challenges related to age-​related and disease-​related
Research, Department of
Neurology, Jacobs School of
ium of effective disease-​modifying therapies, have con- comorbidities as well as the disease itself.
Medicine and Biomedical tributed to an increased likelihood of achieving stable This Review summarizes the epidemiology of MS
Sciences, State University of disease and to the increased longevity of patients with as it pertains to ageing individuals and discusses the
New York (SUNY), Buffalo, MS. However, all randomized controlled trials (RCTs) unique challenges facing elderly patients with MS. These
NY, USA.
performed to date to evaluate the safety and efficacy of challenges include the complexity of the relationship
2
Buffalo Neuroimaging disease-​modifying therapies have specifically excluded between ageing and MS, the lack of clarity regarding the
Analysis Center, Department
of Neurology, Jacobs School
elderly patients with MS. In addition, few studies have safety and efficacy of disease-​modifying therapies in
of Medicine and Biomedical evaluated the complex interaction of this chronic disease elderly individuals, controversy over when and whether
Sciences, State University of with the natural ageing process. discontinuation of treatment is appropriate and the
New York (SUNY), Buffalo, Some patients with MS present with a progressive importance of supporting wellness and cognition.
NY, USA.
course from disease onset, termed primary progressive
3
Department of MS (PPMS), but ~80% of patients with MS present with Epidemiology of MS in elderly patients
Pharmaceutical Sciences,
Jacobs School of Medicine
a relapsing–remitting course. Relapsing–remitting MS Incidence and prevalence. MS is a chronic demyelinat-
and Biomedical Sciences, (RRMS) is characterized by acute episodes of neurolog- ing condition of the CNS associated with good survival;
State University of New York ical impairment followed by a return to baseline func- these patients’ life expectancy is only slightly below that
(SUNY), Buffalo, NY, USA. tion. Eventually, recovery from these episodes becomes of the general population5–8. Contemporary estimates
4
Center for Biomedical incomplete and neurological disability accumulates, suggest an average reduction in lifespan of 6–10 years
Imaging at the Clinical which marks the transition to secondary progressive MS for patients with MS8,9. Therefore, the number of elderly
Translational Science Institute,
State University of New York
(SPMS). Most individuals are diagnosed with MS at age individuals with MS is increasing in conjunction with
(SUNY), Buffalo, NY, USA. 20–50 years2, and within this subgroup, >30% remain in ageing of the general population. In 2004, an analysis
*e-​mail: bw8@buffalo.edu the RRMS phase of the disease into old age3,4. However, a of data from a large MS registry in the United States
https://doi.org/10.1038/ few individuals are diagnosed with MS after age 50 years revealed that ~14% of patients with MS were ≥65 years
s41582-019-0183-3 (termed late-​onset MS (LOMS)) or even after age 60 years of age10.

NATure ReviewS | NeURology volume 15 | JUNE 2019 | 329


Reviews

Key points 50 years was once an exclusionary diagnostic criterion


for MS24–26. However, LOMS now accounts for 3.4−4.8%
• The prevalence of ageing individuals with multiple sclerosis (MS) is increasing and VLOMS for 0.5% of all MS diagnoses27–30. Finally, a
worldwide. report from Denmark published in 2018 stated that the
• Ageing people with MS present with unique challenges, including a high burden of incidence of MS specifically in individuals >50 years of
comorbidities and an altered immune system profile. age has increased substantially in both men and women31.
• Data on the safety and efficacy of current disease-​modifying therapy regimens in Additional studies are recommended to determine
elderly patients with MS are lacking, indicating the need for further studies in this whether the incidence of LOMS and VLOMS is increasing.
specific population. LOMS and VLOMS provide unique challenges for
• A substantial proportion of elderly patients with stable MS will need to consider the management of MS. Trials of all existing disease-​
whether to discontinue disease-​modifying therapy; data are currently insufficient to modifying therapies for MS specifically excluded age-
provide evidence-​based recommendations on this topic.
ing patients, and because a substantial portion of these
• Complementary lifestyle modifications that promote wellness and cognition can help individuals still have RRMS (which responds favourably
ageing patients with MS to manage their comorbidities and improve their quality of life.
to these treatments), information on the safety and effi-
cacy of these agents in this specific population is greatly
An overall increase in the prevalence of MS, regard- needed. In addition, older patients with MS are more likely
less of age, has been reported worldwide, including in than younger patients with MS to have comorbidities that
Canada11, Norway12, New Zealand13 and in commercial complicate their medical management32,33. The presence
insurance databases in the United States14. Longitudinal of comorbidities (particularly vascular, autoimmune or
data from large registries have also documented an musculoskeletal disease) at symptom onset has also been
increase in the prevalence of MS specifically in the age- associated with substantial delays in the diagnosis of MS34.
ing population in Canada1,11, Italy15 and Australia16,17.
Substantial published data supporting an increase specif- Mortality. Patients with MS show higher mortality than
ically in ageing individuals with MS come from Canada, the general population33,35–38. Although the accumulation
a nation with a high prevalence of MS1. The prevalence of progressive disability caused by the disease process itself
of MS in Manitoba, Canada, was estimated to be 226.7 is not always the immediate cause of death, MS is recorded
cases per 100,000 of the population in 2006, a statistically as an underlying cause of death for ~50% of patients with
significant increase from 32.6 per 100,000 in 1984 (ref.1). MS36. The results of a large study of individuals included
This study also documented a gradual upward trend in in the US Department of Defense administrative claims
the age of peak prevalence of MS, from 35–39 years in database showed that patients with MS have an increased
1984 to older age groups in subsequent years. The most all-​cause mortality ratio (2.9, 95% CI 2.7–3.2) compared
recent assessment (in 2004) found that the age of peak with the non-​MS population33. The largest increase in
prevalence of MS had risen to 55–59 years1. A 2015 mortality ratio (9.4, 95% CI 5.3–16.7) occurred in patients
report from Genoa, Italy, estimated the prevalence of MS with MS aged 50–59 years33. These findings were repli-
to be ~148.5 per 100,000 individuals and, importantly, cated in a similar study completed in Norway8. However,
that 18% of all patients with MS were >65 years of age15. as patients age, direct causes of death other than MS (such
In Australia, the prevalence of MS increased from <20 as cardiovascular disease, respiratory disease, infections
per 100,000 individuals to ~60 per 100,000 over a time and cancer) become increasingly prevalent39.
span of 45 years, and the most notable increase occurred Despite the documented increase in the prevalence
in individuals >60 years of age16. of MS, mortality in patients with MS seems to be either
Several reasons probably contribute to this increase stable or perhaps decreasing. A study of MS-​related
in the prevalence of MS. Chief among those reasons is mortality in Canada suggested that whereas overall
increased longevity, although changes in diagnostic cri- MS-​related mortality itself has remained fairly stable
teria and improvements in diagnostic tools, including over the past ~35 years, deaths attributed to MS have
the availability of advanced imaging modalities, are also steadily shifted to older age groups40. This change reflects
likely to be involved. An increase in the incidence of MS both the ageing of the population and the increased life
in central Finland was almost entirely attributable to the expectancy of patients with MS, which probably results
increased diagnostic use of MRI18. Similarly, a study con- from improved treatments for both MS and prevalent
ducted in the United States found that the incidence of comorbidities. A study that examined trends in mortal-
MS remained stable over a period of 15 years after adjust- ity for patients with MS in the United States found that
ment for age and sex19; comparable results were obtained MS-​specific mortality peaks at ~74 years of age and then
in a study conducted in Bavaria20. Conversely, studies declines substantially41. Individuals with MS are now
conducted in Italy21 and Australia22, as well as a large living longer; therefore, consideration of appropriate
meta-​regression analysis23, suggested that the overall inci- disease-​modifying therapies and other ways of main-
dence of MS is rising along with the increases in its prev- taining wellness and treating comorbidities in elderly
alence. Differences in methods of case ascertainment, individuals with MS are necessary.
emphasis on the use of MRI, increased awareness of the
disease and changes in diagnostic criteria might partially Immunopathology
explain the lack of consensus among these studies. A well-​functioning immune system seamlessly inte-
Regardless of whether the incidence of MS is truly ris- grates the innate response to an immediate threat with
ing, the diagnosis of incident MS after the age of 50 years the adaptive response necessary for future protection.
was formerly infrequent; indeed, symptom onset after age Dysregulation at many levels of the immune system is

330 | JUNE 2019 | volume 15 www.nature.com/nrneurol


Reviews

central to the development and progression of MS42. Inflammageing and immunosenescence. Ageing is
For example, autoreactive peripheral lymphocytes cross generally accompanied by immunosenescence54, a pro-
the degraded blood–brain barrier (BBB) into the CNS, found weakening of both the innate and adaptive com-
where they recognize myelin fragments and initiate an ponents of the immune system, which leads to ageing
immune response43. The resulting demyelination is the individuals having a reduced capacity for mounting an
most recognized hallmark of MS44. adequate immune response54. Along with natural age-
MS was originally thought to be primarily a dis- ing, patients with MS also experience an accumulation
ease of the adaptive immune system. Indeed, studies of iron within the CNS, resulting in oxidative stress55,56.
in the most appropriate animal model of MS, exper- As oligodendrocytes are damaged by the disease pro-
imental autoimmune encephalomyelitis, identified cess, iron is released and its extracellular accumula-
that cells of the adaptive immune system — primarily tion potentiates oxidative stress and results in further
T cells45 but also B cells46 — are major drivers of MS neurodegeneration55.
pathophysiology. The success of therapies directed Autophagy, the intrinsic mechanism that compen-
against T cell and B cell targets provides support for sates for the accumulation of non-​functional organelles
this hypothesis47. However, subsequent investigations and toxic products, supports cellular health when it
have shown that the innate immune system might also is functioning appropriately57. If this mechanism is
be involved in MS, particularly at progressive stages of overwhelmed, waste products accumulate, leading to
the disease48. cellular senescence and ultimately apoptosis, which
destroys damaged cells58. Evidence suggests that auto-
Lesion characteristics on imaging. Inflammation and phagy becomes less proficient in ageing cells57 (Fig. 1). In
T cell infiltration are present in active MS lesions dur- general, neurodegeneration is associated with decreased
ing the acute and relapsing phases of the disease and autophagy. However, patients with active, relapsing MS
can contribute to neurodegeneration, including axon show increased autophagy in peripheral immune cells
and cell loss47,49. A study that examined inflamma- compared with cells from healthy controls59. The para-
tion and neurodegeneration in the brains of patients dox observed in progressive MS is the presence of neu-
with MS found that T cell infiltration and substantial rodegeneration despite evidence of increased autophagy
inflammation were seen most frequently in active MS in the brain tissue59 (Fig. 2).
lesions50, which are associated with acute and relaps- In young patients, the robust repair capacity of the
ing disease. However, increasing numbers of T cells brain compensates for changes in the early (inflamma-
were also found in lesions associated with progressive tory) stage of MS, but in ageing individuals a decreased
disease, such as slowly expanding lesions 50. On the repair capacity and the neurodegenerative processes
contrary, few T cells were seen in inactive and remy- associated with ageing both lead to a decrease in com-
elinated (shadow) lesions. Although brain tissue from pensatory reserve60. Therefore, over time, axonal degen-
ageing individuals with long disease durations demon- eration, iron deposition and oxidative stress accumulate,
strated profound neurodegeneration, inflammation in eventually overwhelming the brain’s natural reserves and
remyelinated lesions had declined to levels compara- leading to disease progression61.
ble to those in the brains of age-​matched individuals Ageing is often accompanied by chronic low-​grade
without MS50. inflammation, a phenomenon known as inflammage-
Lesion characteristics vary dynamically according to ing62, probably as a result of chronic exposure to infectious
the patient’s age, disease duration and disease pheno- agents such as cytomegalovirus and Epstein–Barr virus.
type51. At any given time, various types of MS lesions Senescent immune cells accumulate in ageing individuals
can coexist. Lesions are histopathologically character- and create a pro-​inflammatory environment attributable
ized as active (highly inflammatory with a dense mac- to ongoing secretion of growth factors, pro-​inflammatory
rophage population), slowly expanding (with a central cytokines and autoreactive antibodies, among other fac-
astrocytic scar and activated microglia on the lesion tors62–64. These senescent cells are no longer able to con-
edge), inactive (containing demyelinated axons with tribute to tissue repair; instead, they recruit inflammatory
an astrocytic scar and no ongoing inflammatory activ- cells that further contribute to tissue damage65 (Fig. 1).
ity) or remyelinated (shadow lesions, which have an This chronic inflammation is believed to be associated
inflammation level comparable to that in age-​matched with the development of many age-​related diseases62.
controls)52. Advanced patient age and prolonged disease Thymic involution (the shrinking in size of the thy-
duration are associated with a reduced chance of detect- mus that occurs in ageing individuals) could also help
ing active lesions and an increased chance of detecting to explain this diminished immune response. Thymic
smouldering plaques51. Smouldering plaques, which are involution results in a decline in the production of
often also termed chronically active or slowly expanding naive T cells, which leads to reduced T cell activity in
plaques, demonstrate lesion-​specific rim activity asso- elderly individuals and an associated increase in the risk
ciated with iron-​laden macrophages and amplification of infection66,67. Another potential explanation for the
of the oxidative injury owing to ferritin accumulation53. reduced immune function observed in elderly patients
As the presence of smouldering lesions is particularly with MS is the reduction in neurotrophic factors68,69.
associated with the onset of progressive disease and This decreased neurotrophic support corresponds to a
with the accelerated accumulation of physical disability, decreased capacity for remyelination and to an overall
they have the potential to become an important imaging reduction in brain plasticity, which further compound
biomarker of progressive MS52. the effects of ageing and MS69.

NATure ReviewS | NeURology volume 15 | JUNE 2019 | 331


Reviews

SASP

Phagosome
Mitochondrion Lysosome

ROS +
Or
Apoptosis

ER

Nucleus

If the accumulation of damaged organelles


overburdens the autophagy process

Fig. 1 | Cellular processes involved in inflammageing and immunosenescence. Increased energy demands in ageing
immune cells lead to mitochondrial swelling and failure. The damaged mitochondria release large amounts of cytotoxic
reactive oxygen species (ROS), which damage other organelles as well as the cell’s own DNA , leading to functional
alterations. The damaged organelles are initially engulfed by autophagosomes, which merge with lysosomes to form
autophagolysosomes that digest and recycle their contents, a process termed autophagy. However, if autophagy becomes
overwhelmed, the cell either undergoes apoptosis or becomes senescent. Senescent cells exit the cell cycle and no longer
respond to pro-​apoptotic signals. Accordingly , these cells increase in size, do not proliferate and show increased
production of cytokines and other products, which can damage neighbouring cells. These features are termed the
senescence-​associated secretory phenotype (SASP). ER , endoplasmic reticulum.

The limited lesion-​repair capacity of elderly patients disease-​modifying therapies have proved effective for
with MS is partly related to a lack of remyelination, preventing relapse, many have not demonstrated a con-
which is a factor requiring consideration in the devel- vincing reduction in the accrual of disability in indi-
opment of new therapies56,70. In an autopsy series, the viduals with progressive disease42,73. As a result, many
brains of patients with progressive MS had a higher investigators have concluded that different mechanisms
lesion load and were also much less likely to have remy- underlie RRMS and progressive MS42.
elinated lesions than were those of patients with RRMS71.
The authors suggest that the remyelination deficit is due MRI findings in MS and ageing
to widespread axonal degradation, which leaves only few MRI-​derived evidence of MS pathology is an important
axons available for remyelination71. This study also con- tool not only in establishing the diagnosis but also in
firmed a previous report that documented the presence predicting and detecting disease progression and in eval-
of substantial inflammation in the brains of patients with uating responses to therapy74. The increasing utilization
MS50, even those with progressive MS71. Together with the of MRI has greatly contributed to understanding the
knowledge that BBB permeability is decreased in changes in MS pathology associated with increasing age
patients with progressive MS72, these findings suggest and with progressive MS. Here, we introduce some MRI
that disease activity remains compartmentalized behind features of MS that are particularly affected by ageing:
the BBB in progressive MS, which might present an lesion accumulation and detection; global, regional and
additional challenge for treatment. nuclei-​specific brain atrophy; spinal cord pathology;
Finally, the well-​established observation that immu- and the identification of comorbidities.
nomodulatory and anti-​inflammatory therapies are The aforementioned increase in iron concentration
most effective in the early stages of RRMS42,55 sug- at the rim of slowly expanding lesions provides the basis
gests that mechanisms involved in early disease stages for lesion visualization on gradient-​based susceptibility-​
might not be as relevant in progressive MS42. Although weighted imaging (SWI) and quantitative susceptibility

332 | JUNE 2019 | volume 15 www.nature.com/nrneurol


Reviews

Autoreactive T cell Fig. 2 | The influence of ageing on MS pathophysiology.


Young patients with relapsing–remitting multiple sclerosis
Natural killer cell (RRMS) have a highly active peripheral immune system,
which includes autoreactive T cells and B cells. These
B cell activated lymphoid cells are able to migrate through the
damaged and disrupted blood–brain barrier (BBB) and
Neutrophil 1
The MS brain enter the brain, where they cause inflammation. In ageing
patients with multiple sclerosis (MS), the overall level of
activation of the immune peripheral immune system is
considerably reduced compared with that in young
patients with RRMS, but ageing greatly increases BBB
permeability , resulting in increased cellular trafficking (1).
MS, along with other neurodegenerative diseases, shares
Proliferating the hallmark of an altered balance between M1-like
astrocytes (pro-inflammatory) and M2-like (anti-​inflammatory)
Cytokines
3 microglia, leading to an increased proportion of
M1-like microglia, which promote inflammatory
Neuron
neurodegeneration (2). The chronic inflammatory state
associated with ageing leads to proliferation of astrocytes
2 Myelin and astrocytosis (scar formation) and prevents effective
lesion repair, remyelination and debris clearance by
macrophages (which engulf myelin debris and present it to
B cells and T cells) (3). This persistent inflammatory activity
also leads to increased loss of neuronal synaptic processes.
Ageing neurons also exhibit decreased synaptic plasticity ,
which further exacerbates their synaptic dysfunction (4).
Damaged neuron
Increased accumulation of activated memory B cells
M2-like and plasma cells within the meninges and meningeal
M1-like microglia tertiary follicle-​like structures is found in ageing patients
microglia with progressive MS. These cells serve as a constant
Oligodendrocyte source of pro-​inflammatory signals (5). Activated and
Myelin debris compartmentalized T cells also maintain and self-​sustain
4
the chronic inflammatory state within the brain in ageing
patients with progressive MS (6).
Macrophage

T cell
receptor This measure explained a substantial proportion of the
6
variance in predictions of disability, even after con-
Activated B cell Activated trolling for brain atrophy and new or enlarging lesion
T cell activity79. Therefore, atrophied lesion volume has poten-
tial as a tool for assessing the severity of progressive MS
Plasma cell in the ageing population79.
Dendritic spine Lastly, the presence of a high burden of cortical
5
lesions (which occurs more frequently in progressive
MS than in RRMS or CIS) has been associated with a
high degree of physical and (especially) cognitive dis-
ability80,81. The visualization and quantification of cor-
tical lesions using either double-​inversion recovery or
gradient-​based sequences are useful to determine the
MS lesion burden82. Use of high-​field-strength MRI
mapping75,76. Although visualization of the paramagnetic scanners and novel sequences is likely to further con-
lesion rims can be readily performed by 7 T scanners, tribute to understanding the overall process of lesion
only in the past few years have these findings been repli- development83. Meningeal lymphoid follicle-​like struc-
cated using clinically available 3 T scanners77. MS-​related tures can be detected in 25–50% of patients with MS
processes occurring behind the restored BBB can also (reviewed elsewhere) 84, although their prevalence
be imaged using ultra-​small superparamagnetic iron is strongly influenced by the proportion of ageing
oxide particles taken up by circulating macrophages that patients and individuals with progressive MS in the
migrate to sites of inflammation78. study population81. These structures are thought to har-
Another potential imaging biomarker is atrophied bour autoreactive B cells, which could exacerbate local
lesion volume, which refers to the volume of periven- inflammation and cause cortical demyelination. These
tricular lesions (which are gradually replaced by cerebro- structures can be imaged as leptomeningeal contrast
spinal fluid (CSF) or other local atrophic changes)79. In enhancement foci. Multiple reports have corroborated
one study, the atrophied lesion volume on T2-weighted the finding of an increased prevalence of leptomenin-
MRI was higher in patients with progressive MS than in geal contrast-​enhancing structures in older patients with
those with RRMS or clinically isolated syndrome (CIS)79. progressive MS85,86.

NATure ReviewS | NeURology volume 15 | JUNE 2019 | 333


Reviews

Ageing patients with MS also have a substantial and those with PPMS. In addition, atrophy within the
increase in diffuse brain atrophy87. Over the past two cerebellar VI, crus I and VIIIa lobules contributes to spe-
decades, the rate of brain atrophy in patients with cific features of motor and cognitive impairment101,102.
MS has been repeatedly shown to predict short-​term, Although standardization and technical advances in
medium-​term and long-​term disability progression88. MRI hardware and software have made analyses of brain
Several longitudinal studies have attempted to identify atrophy clinically feasible at the group level, these neu-
a cut-​off value to define the threshold between ‘physio- rodegenerative outcomes are still not informative at an
logical’ and ‘pathological’ brain atrophy rates, which is individual patient level88.
proposed to be 0.4% per year89. However, brain volume A large subgroup of patients with suspected MS
in humans undergoes substantial variation throughout present with spinal demyelinating lesions and substan-
life90. The loss of brain volume is thought to be ~0.2% tial spinal cord atrophy, a pathology associated not only
per year from early adulthood to mid-​life, after which with increased risk of conversion to clinically definite
it gradually rises to ~0.5% per year at 60 years of age90. MS but also with long-​term disability progression103. In
Given that the loss of brain volume in healthy elderly contrast to their lack of cranial findings, patients with
individuals exceeds the supposedly pathological rate of PPMS might present with diffuse spinal cord changes,
0.5% per year, attempts to further refine these cut-​off which are associated with demyelination and axonal
values are futile90. loss104,105. Spinal cord atrophy can be demonstrated by
In addition to the effect of overall brain atrophy either measuring the cross-​sectional area at the level of
on the outcomes of patients with MS, accumulat- specific anatomical markers (such as specific cervical
ing evidence has pinpointed cortical atrophy as the vertebrae) or by measuring the cord volume between
main driver of physical and cognitive disability 91,92. two spinal levels. Patients with progressive MS have both
Compared with individuals who have CIS or RRMS, a higher cervical cord lesion load and a lower cervical
patients with SPMS demonstrate more atrophy in cross-​sectional cord area than those of patients with
grey matter but not in white matter92. Furthermore, RRMS, and both of these measures are independent
in longitudinal studies, patients with SPMS have up to contributors to increased levels of disability103. Patients
10-fold more grey matter atrophy than patients with with progressive MS also demonstrate marked atrophy
RRMS and up to 14-fold more grey matter atrophy of the spinal grey matter, which is strongly associated
than healthy age-​matched and sex-​matched controls91. with worse ambulation106. A longitudinal MRI study
Two independent studies employing component-​based demonstrated that spinal cord volume loss is largely
and event-​based analysis have found anatomically dis- independent of the cranial T2 lesion load and brain
tinct patterns of cortical atrophy in patients with MS. atrophy measures107. In this study, spinal cord atrophy
These patterns occur in a nonrandom manner that is was the only MRI-​derived metric associated with both
at least partially driven by disease duration93,94. As in physical disability level and disease progression107. In a
Alzheimer disease (AD), understanding the tempo- separate study, spinal cord atrophy was the only inde-
ral and spatial progression of grey matter atrophy in pendent MRI measure that could identify patients with
patients with MS could lead to improvements in clini- onset of progressive MS108.
cal trial design and the development of individualized In ageing patients with MS, the gradual development
treatment, tailored to age and disease duration, for of age-​associated immunosenescence precludes the use of
patients with MS95. inflammation-​based MRI outcome measures, which
Measures related to specific brain areas might be do not provide adequate or sufficient responsiveness to
more appropriate than total brain measures. Central intervention. New MRI measures that perform consist-
grey matter structures such as the thalamus are espe- ently across multiple MS phenotypes are greatly needed
cially vulnerable to MS pathology owing to their expo- and currently lacking. Use of global and regional meas-
sure to CSF-​borne neurotoxic factors96,97 and their role as ures (such as grey matter atrophy, thalamic volume loss
major sites of connection between afferent and efferent and expansion of the lateral ventricles) or composite
axons97,98. The results of a large, multicentre MRI study measures derived from several differentially affected
have corroborated the clinical importance of this vul- brain regions could ultimately serve as neurodegenerative
nerability to neurodegeneration in deep grey matter MRI end points in MS clinical trials.
structures99. Regardless of the clinical phenotype of MS,
deep grey matter had the fastest longitudinal atrophy Comorbidities
rate, and the loss of deep grey matter volume was the Ageing in patients with MS, as in the general population,
only measure of grey matter atrophy significantly asso- is accompanied by the development and accumulation of
ciated with disability accrual (P < 0.001)99. Lower deep comorbidities, which increase these individuals’ risk
grey matter volumes at baseline were strongly associated of both disability and death109. Moreover, some of the
with shorter times to progression of Expanded Disability comorbidities associated with ageing, such as cognitive
Status Scale (EDSS) scores99. Furthermore, another study decline, are also potential sequelae of MS. Identifying
showed that the thalamic atrophy rate remains consist- whether these features are due to MS or to normal ageing
ent throughout the entire MS disease course, making it can be a challenge.
a convenient MRI-​based primary end point100. The total Several studies have examined the incidence and
cerebellar volume and cerebellar cortex volume are other prevalence of comorbidities associated with MS10,32,33,110.
potential MRI-​based neurodegenerative outcome meas- All patients with MS are at an increased risk of devel-
ures that can be used in both ageing patients with MS oping mobility limitations, but this risk is particularly

334 | JUNE 2019 | volume 15 www.nature.com/nrneurol


Reviews

T1-MRI DWI apparent for ageing individuals10. Difficulty with walk-


a b ing is reported by >85% of patients with MS ≥65 years
of age, compared with 65% of those aged <65 years10.
Patients with MS aged ≥65 years were also much more
likely to report bowel and bladder difficulties than were
younger individuals with MS10. An analysis of the US
Department of Defense administrative claims database
found evidence of an increased risk of several catego-
ries of comorbidities in patients with MS33. Compared
with age-​matched non-​MS controls, patients with MS
had higher rates of malignancies, cardiovascular dis-
ease, psychiatric disorders, infections and other systemic
diseases, including diabetes mellitus33. Some of these
associations are discussed in more detail below.

Cardiovascular disease and diabetes mellitus. The


strongest associations relate to an increased risk of car-
FLAIR FLAIR
diovascular disease (event rate ratio 3.6, 95% CI 3.5–3.8)
d c and ischaemic stroke (event rate ratio 3.8, 95% CI 3.5–4.2)
in patients with MS versus the non-​MS population33.
Patients with MS who have cardiovascular disease also
seem to have a higher risk of death from this cause than
the general population6. Similar increases in the preva-
lence of cardiovascular comorbidities110, diabetes mel-
litus111 and other vascular comorbidities33,112 have also
been documented in other studies of patients with MS,
and several population-​based case–control and cohort
studies have documented an increased risk of hyperten-
sion112,113 and stroke109,114,115. However, an analysis of a
large administrative claims database in Canada found
that rates of hypertension, hyperlipidaemia and diabetes
mellitus were similar in patients with MS and the gen-
SWI SWI
eral population116, suggesting that additional studies are
needed to clarify these associations.
e f Regardless, ageing patients with MS can present with
a plethora of comorbidities associated with cerebrovas-
cular pathology, including small-​vessel disease, cerebral
hypoxia, iron accumulation and mitochondrial dysfunc-
tion. These cerebrovascular diseases are heterogeneous,
can coexist and look similar to MS pathology on imaging
studies117. These cerebrovascular features include but are
not limited to punctate or confluent white matter hyper-
intensities, cerebral microbleeds118, enlarged perivascu-
lar (Virchow–Robin) spaces, lacunae, small subcortical
or cortical microinfarcts and brain atrophy119 (Fig. 3). Of
note, the presence of white matter hyperintensities in the
juxta-​cortical region, spinal cord or optic nerve and
the involvement of U-​fibres are specific signs of MS pathol-
ogy117. Furthermore, the presence of a central vein is an
Fig. 3 | Cerebrovascular disease can mimic MS-​specific pathology on MRI. T1-weighted additional well-​known characteristic of MS lesions that
MRI (T1-MRI) (part a) and diffusion-​weighted imaging (DWI) (part b) scans from the same can be seen on T2*-weighted MRI as a thin hypointense
patient with multiple sclerosis (MS). A hypointense lesion on T1-MRI shows enhancement line that runs partially or entirely through the lesion120.
on DWI suggestive of a lacunar stroke within the left capsula interna (a, large black arrow; In the healthy ageing population, age-​associated cerebro-
b, white arrow). Lacunar stroke can resemble the appearance of an MS-​derived lesion. The vascular pathology has been associated with decreased
small arrow indicates an MS-​related T1-weighted hypointense lesion, also known as an MS total and regional cerebral volumes121,122. Multiple studies
black hole. Fluid-​attenuated inversion recovery (FL AIR) images from two different patients have shown that patients with MS and comorbid cardio­
with MS (parts c,d), showing (right) T2-hyperintense bands in the periventricular white
vascular disease have lower cross-​sectional (and accel-
matter and multiple small T2-hyperintense lesions outside the borders of the lateral
ventricles (white arrows), consistent with cerebral small-​vessel disease, and (left) symmetrical erated longitudinal loss of) global and regional brain
T2-hyperintense areas posterior to the lateral ventricles, suggestive of a microvascular volumes123,124. Successful differentiation of both the ori-
ischaemic process. Small hypointensities (white arrow) suggest cystic microinfarcts. Cerebral gin of white matter hyperintensities and age-​associated
microbleeds (white arrows) are present in susceptibility-​weighted imaging (SWI) scans from neuronal loss will help to guide clinical decision-​making
an ageing healthy individual (part e) and an ageing patient with MS (part f). in ageing patients with MS.

NATure ReviewS | NeURology volume 15 | JUNE 2019 | 335


Reviews

Neurological and psychiatric disorders. Some data risk of comorbidities, particularly as these patients age,
suggest that the prevalence of other neurological disor- and to ensure that their comorbidities are managed
ders, such as epilepsy, is also increased in patients with appropriately.
MS32. The concurrence of psychiatric disorders, includ-
ing depression, and MS is well established. In a meta-​ Safety and efficacy in elderly patients. Because clin-
analysis, the risk of depression in patients with MS was ical trials of existing disease-​modifying therapies for
estimated to be up to 23-fold higher than in the general MS have systematically excluded ageing patients, our
population32. However, long disease durations might knowledge of the safety and efficacy of these treatments
mitigate the strength of this association; older age125 in elderly populations is insufficient. In subgroup ana­
and longer time from disease onset126 have been asso- lyses, however, fingolimod was not more effective than
ciated with less-​severe depression in patients with MS, placebo in reducing the annualized relapse rate in
although other studies have reported little to no change patients >40 years of age149. Furthermore, when natali-
in patients’ depressive symptoms over time127,128. zumab was assessed for efficacy in patients with SPMS,
Little is understood about the association between the trial failed to meet its primary end point of reducing
MS and AD. Cognitive impairment affects individuals a multicomponent disability measure comprising the
with both diseases, although the clinical presentation of EDSS, Timed 25-Foot  Walk Test150 and the Nine-​Hole
cognitive impairment differs129. In MS, cognitive process- Peg Test151, although natalizumab did demonstrate effi-
ing speed is very commonly impaired, whereas in AD, cacy in reducing disease progression as assessed by the
episodic memory is typically most strongly affected129. Nine-​Hole Peg Test alone152.
One study conducted in individuals with LOMS
Disease-​modifying therapy examined the association between IFNβ treatment and
In late March 2019, two additional disease-​modifying disability progression in patients with RRMS153. The
therapies (cladribine and siponimod) were approved in results showed no evidence of a reduction in disability
the United States for the treatment of RRMS and active in this cohort of older (aged ≥50 years) patients with MS
SPMS130,131. It will be critical to see how the utilization of exposed to IFNβ compared with historical controls153.
these therapies affects ageing individuals with MS. Of The researchers acknowledged several potential reasons
the 15 previously FDA-​approved therapies for RRMS, for this negative finding. As previously described, ageing
14 remain on the market. By contrast, FDA approval patients with MS seem to have an inflammatory milieu
had previously been obtained for only one disease-​ different from that in their younger counterparts that
modifying therapy for PPMS (ocrelizumab) and only might not respond to immunomodulatory agents in the
one for SPMS (mitoxantrone)132. Therefore, few options same manner. Elderly patients with MS also often have
for disease-​modifying therapies were available in indi- comorbidities that can affect the efficacy of disease-​
viduals with progressive MS, who represent the majority modifying treatments. Finally, most studies are either
of patients with MS >65 years of age. The majority of too short or are not appropriately timed154 to capture
approved therapies for RRMS have failed to demonstrate either disease progression or disability accrual in elderly
efficacy in clinical trials as a treatment for progressive individuals, for whom MS is often not very active153.
MS. An apparent decrease in the effectiveness of disease-​ Taken together, these studies provide substantial evi-
modifying therapy in patients with progressive MS sug- dence suggesting that the role of the adaptive immune
gests that different pathobiological mechanisms might system in MS changes as individuals age.
come into play in the progressive stages of the disease. Studies of both approved155 and emerging147 disease-​
Notably, the clinical trials of disease-​modifying ther- modifying therapies have demonstrated slowing of
apies for RRMS were not designed to assess efficacy in disability progression in patients with progressive MS.
ageing patients. In fact, the pivotal clinical trials of the However, the participants in these trials were still fairly
most widely used disease-​modifying therapies specifi- young. In the ocrelizumab study in PPMS, the upper
cally excluded individuals aged >45 years (glatiramer age limit for eligibility was 55 years155, and for the
acetate133), aged >50 years (IFNβ1b134, natalizumab135 siponimod study in SPMS it was 60 years147. Cladribine
and alemtuzumab136,137) and aged >55 years (IFNβ1a138,139, is an oral medication that has now been approved by
dimethyl fumarate140,141, fingolimod142,143 and terifluno- the FDA131 for the treatment of RRMS, and has already
mide144,145). A currently recruiting phase III study of ocre- been approved in the European Union and several other
lizumab in patients with progressive MS (including some countries. In post hoc analyses of cladribine trial data
with SPMS) will have the highest upper age limit used presented at the 2018 annual meetings of the European
thus far in MS clinical trials — 65 years of age146 (Table 1). Committee for Treatment and Research in Multiple
Notably, clinical trials of siponimod, an oral selective Sclerosis (ECTRIMS) and the European Academy of
modulator of sphingosine 1-phosphate receptors 1 and 5 Neurology (EAN), the medication was similarly effective
(S1P1 and S1P5, respectively), demonstrated a significant in younger and older patients with RRMS — defined as
reduction in disability progression among patients with age above and below 45 years of age, respectively, in the
SPMS147. This therapy has now been approved by the CLARITY study152 and above and below 50 years of age,
FDA for the treatment of CIS, RRMS and active SPMS131. respectively, in the other study153.
In patients with MS, the presence of comorbidities is Real-​world data are needed from longitudinal MS
associated with a decreased likelihood of using disease-​ registries or prospective cohort studies to identify the
modifying therapy148. Therefore, it is important for clini- effects of disease-modifying therapies in elderly pati­ents
cians to counsel patients with MS about their increased with MS. RCTs also typically exclude elderly patients with

336 | JUNE 2019 | volume 15 www.nature.com/nrneurol


Reviews

MS who have comorbidities, for whom careful consid- to be most efficacious in young individuals with MS166,167.
eration of treatment options is very important. The fact The results of a large cohort study in patients with all
that elderly patients with progressive MS have a very phenotypes of MS (although ~78% had progressive MS)
low probability of relapse156 and decreased immune showed that each additional year of age was associated
activity51 should also be kept in mind. Therefore, studies with a 3% increase in the risk of neurological progression
designed to examine the efficacy of disease-modifying after autologous HSCT168. However, most studies of this
therapies in elderly patients with progressive MS should intervention in patients with MS have been conducted in
evaluate non-​traditional markers of disease progression individuals with median ages of ~25–40 years, and very
and activity157–159. few patients aged >50 years have been included167,168.
Given the increasing prevalence of elderly individuals
Treatment discontinuation. Controversy has flared over with MS and the lack of data on the safety and efficacy
whether ageing patients with MS can safely discontinue of current disease-​modifying therapies in the elderly MS
disease-​modifying therapies after a certain age or disease population, further studies specific to this subgroup are
duration160,161. Data from real-​world observational stud- both necessary and warranted. We are hopeful that the
ies in support of either continuation or discontinuation results of a large ongoing RCT that will assess therapy
are lacking, and experts consequently disagree on treat- discontinuation in adults ≥55 years of age will provide
ment recommendations for ageing patients with stable guidance on this very important issue169.
MS. Relapse frequency decreases substantially with age,
with the result that relapses are very infrequent among Additional considerations in elderly patients
patients with MS aged ≥60 years156,162,163. Some experts Wellness. A growing emphasis is being placed on life-
argue that an age ≥60 years represents an appropriate style strategies as a complement to traditional therapy
point to consider discontinuation of disease-​modifying in patients with MS. One of the most frequently targeted
therapies, especially those with unfavourable safety pro- lifestyle factors is smoking. In addition to potentially
files160. In an observational study of patients with MS aged increasing the risk of developing MS, the literature con-
≥60 years, ~30% chose to discontinue disease-​modify- sistently shows that smoking is associated with increased
ing therapies, most often at the recommendation of their disability and worse prognosis in patients with MS170.
provider163. In this population, only one relapse was doc- Conversion to SPMS is significantly accelerated in
umented, although ~10% of patients who discontinued patients who continue to smoke after diagnosis of MS
disease-​modifying therapy chose to reinitiate it during versus those who quit (by 4.7% for each year of continued
follow-​up163. However, in another observational study, smoking; P < 0.001)171. Other studies have documented
which had the advantage of a propensity-​matched com- an increased risk of attaining disability milestones (EDSS
parison group who chose to continue disease-​modifying scores 4.0 and 6.0) among smokers compared with either
therapies, the time to confirmed disability progression non-​smokers or former smokers172,173.
was significantly shorter in the patients who discon- Another highly studied lifestyle behaviour that
tinued disease-​modifying therapies than in those who has been evaluated in patients with MS is exercise.
continued to take them164. Routine exercise does seem to improve fatigue in these
In the absence of information from clinical trials patients170, but evidence is inconsistent with respect to
about the efficacy of disease-​modifying therapy in elderly whether exercise slows either disease progression or
patients with MS, investigators have attempted to use cognitive impairment174,175. Despite this inconsistency,
existing data to characterize the relationship between exercise is clearly important and recommended for
drug efficacy and age. One meta-​analysis determined that patients with MS by the US National Center on Health,
the efficacy of disease-​modifying therapy showed a strong Physical Activity and Disability, even if only for the
inverse association with increasing age165. This meta-​ beneficial effects of reducing obesity and preventing
analysis of 38 clinical trials, involving all FDA-​approved obesity-​related comorbidities as these patients age170.
disease-​modifying therapies, showed that age accounted Similarly inconsistent results have been documented
for almost 60% of the variation in slowing disability pro- for dietary interventions in patients with MS. Many dif-
gression for IFNβ clinical trials and >40% of the variation ferent types of diets have been evaluated in this popula-
in slowing disability progression for all disease-​modifying tion, including the Mediterranean diet, so-​called Paleo
therapies. In a linear regression model weighted for trial diets, low-​sodium and low-​fat diets and the Swank diet.
duration and sample size, adjustment for baseline EDSS To date, no substantial, consistent evidence favours one
scores did not significantly improve the fit of the model, specific diet over another. However, following a plant-​
which predicted that no disease-​modifying therapies were based, anti-​inflammatory diet is helpful for promoting
effective beyond the age of ~53 years. Moreover, high- overall health and might help to minimize comorbid
efficacy drugs were superior to low-efficacy drugs in terms conditions and potentially other symptoms associated
of slowing MS disability progression only in patients  with MS170. RCTs of dietary interventions in patients with
<40 years of age. MS are ongoing, and their results are expected to pro-
Also requiring consideration is the fact that a large vide additional evidence regarding this modifiable
proportion of elderly patients with MS still have RRMS, lifestyle behaviour176–178.
which is highly amenable to treatment. However, even Social support for ageing patients with MS is also
interventions that are highly effective against RRMS as extremely important. Qualitative studies have identified
well as progressive MS interventions, such as autologous social support and social engagement to be critical to
haematopoietic stem cell transplantation (HSCT), seem healthy ageing in patients with MS179,180. On the whole,

NATure ReviewS | NeURology volume 15 | JUNE 2019 | 337


Reviews

Table 1 | Pivotal clinical trials of disease-​modifying therapies for MS


Name and design Primary outcome Participants Upper Refs
age limit
Phase III double-​blind RCT Difference in relapse rate • 251 patients with RRMS 45 years 133

• 125 assigned to glatiramer acetate


• 126 assigned to placebo
PRISMS: phase III Number of relapses during the study • 502 patients with RRMSa 50 years 139

double-​blind RCT • 167 received 22 µg IFNβ1a


• 165 received 44 µg IFNβ1a
• 170 received placebo
Phase III double-​blind RCT Time to sustained disability progression • 301 patients with RRMS 55 years 138

(≥1.0 point on EDSS) • 158 assigned to IFNβ1a


• 143 assigned to placebo
Double-​blind RCT • Difference in relapse rate • 372 ambulatory patients with RRMS 50 years 134

• Proportion of relapse-​free patients • 125 assigned to 1.6 MIU IFNβ1b


• 124 assigned to 8.0 MIU IFNβ1b
• 123 assigned to placebo
MIMS: phase III Composite of five clinical measures: change • 188 patients with worsening RRMS or SPMSa 55 years 192

double-​blind RCT in EDSS score at 24 months; change in • 64 received 5 mg/m2 mitoxantrone


ambulation index at 24 months; number of • 60 received 12 mg/m2 mitoxantrone
relapses treated with corticosteroids; time • 64 received placebo
to first treated relapse; and change from
baseline in neurological status at 24 months
Phase III double-​blind RCT • Rate of clinical relapse at 1 year • 942 patients with RRMS 50 years 135

• Rate of sustained EDSS progression • 627 assigned to natalizumab


at 2 years • 315 assigned to placebo
FREEDOMS: phase III Difference in annualized relapse rate • 1,272 patients with RRMS 55 years 142

double-​blind RCT • 429 assigned to 1.25 mg fingolimod


• 425 assigned to 0.50 mg fingolimod
• 418 assigned to placebo
TRANSFORMS: phase III Annualized relapse rate • 1,153 patients with RRMSa 55 years 143

double-blind, active- • 369 received 1.25 mg fingolimod


comparator RCT • 398 received 0.50 mg fingolimod
• 386 received IFNβ1a
TEMSO: phase III Annualized relapse rate • 1,088 patients with RRMS 55 years 144

double-​blind RCT • 365 assigned to 7 mg teriflunomide


• 358 assigned to 14 mg teriflunomide
• 363 assigned to placebo
TOPIC: phase III Time to relapse (conversion to clinically • 614 patients with CIS 55 years 145

double-​blind RCT definite MS) • 203 assigned to oral 7 mg teriflunomide


• 214 assigned to oral 14 mg teriflunomide
• 197 assigned to placebo
TOWER: phase III Annualized relapse rate • 1,165 patients with RRMSa 55 years 193

double-​blind RCT • 407 received 7 mg teriflunomide


• 370 received 14 mg teriflunomide
• 388 received placebo
CONFIRM: phase III double-​ Annualized relapse rate over 2 years • 1,417 patients with RRMS 55 years 141

blind, active-​comparator RCT • 359 assigned to twice daily dimethyl fumarate


• 345 assigned to thrice daily dimethyl fumarate
• 350 assigned to glatiramer acetate
• 363 assigned to placebo
DEFINE: phase III Proportion of patients who had relapsed • 1,234 patients with RRMS 55 years 140

double-​blind RCT by 2 years • 410 assigned to twice daily dimethyl fumarate


• 416 assigned to thrice daily dimethyl fumarate
• 408 assigned to placebo
CAMMS223: phase II double-​ • Efficacy (time to sustained accumulation • 334 patients with RRMS 50 years 136

blind, active-​comparator RCT of disability) • 110 assigned to 24 mg alemtuzumab daily


• Relapse rate • 113 assigned to 12 mg alemtuzumab daily
• 111 assigned to subcutaneous IFNβ1a three
times weekly
CARE-​MS I: phase III double-​ • Relapse rate • 563 patients with RRMS 50 years 137

blind, active-​comparator RCT • Time to 6-month sustained disability • 376 assigned to alemtuzumab
accumulation • 187 assigned to IFNβ1a

338 | JUNE 2019 | volume 15 www.nature.com/nrneurol


Reviews

Table 1 (cont.) | Pivotal clinical trials of disease-modifying therapies for MS


Name and design Primary outcome Participants Upper Refs
age limit
OPERA I and OPERA II: two Annualized relapse rate • OPERA I: 821 patients with RRMS 55 years 194

identical phase III double-​ • OPERA II: 835 patients with RRMS
blind, active-​comparator RCTs • 600 mg ocrelizumab every 24 weeks versus
44 μg IFNβ1a three times weekly
ORATORIO: phase III, Percentage of patients with disability • 732 patients with PPMS 55 years 155

double-​blind RCT progression at 12 weeks • 488 assigned to ocrelizumab


• 244 assigned to placebo
CONSONANCEb: open-​label • Proportion of participants without • 600 patients with progressive MSc 65 years 146

efficacy and safety study evidence of progression • All patients assigned to 600 mg ocrelizumab
• Proportion of participants without every 24 weeks
evidence of progression for ≥24 weeks
and no active disease
Numbers of patients refer to the intention to treat (randomized) population, except where noted. CIS, clinically isolated syndrome; EDSS, Expanded Disability
Status Scale; MIU, million international units; MS, multiple sclerosis; PPMS, primary progressive MS; RCT, randomized controlled trial; RRMS, relapsing–remitting
MS; SPMS, secondary progressive MS. aPer-​protocol population. bCurrently enrolling patients. cIncludes both SPMS and PPMS.

lifestyle behaviours are an important complement to differ from their younger counterparts in that they have
therapy in this population and can have a strong effect more neurodegeneration and less-​effective neuronal
on quality of life as patients with MS age. repair processes. The specific pathobiology underlying
the weakening of repair processes in ageing patients
Cognition. Cognitive decline is a particular concern with MS is not well understood, but the reduced effi-
in ageing patients with MS as it can be a consequence cacy of disease-​modifying treatment in this population
of both the disease and the normal ageing process. clearly indicates that prevention of neurodegeneration
Cognitive impairment eventually develops in as many and the accumulation of disability are of critical impor-
as 65% of patients with MS181. Furthermore, cognitive tance. To that end, well-​designed studies are needed to
decline can become apparent early in the disease pro- understand the functioning of these repair processes
cess, potentially even before conversion from CIS to specifically in ageing individuals and to evaluate the
RRMS181. Interestingly, the rate of cognitive decline in efficacy and safety of treatments with neuroprotective
patients with MS does not accelerate with ageing182,183; and repair-​based mechanisms (including remyelination)
in fact, studies of cognitive decline in patients with MS in elderly patients with MS. For those with stable dis-
show that, after controlling for age, the frequency of ease on existing disease-​modifying therapy regimens,
MS-​associated cognitive impairment stays roughly the the timing and appropriateness of discontinuing this
same throughout life183. The trial investigators suggested therapy must also be specifically evaluated.
that rapid cognitive decline in ageing patients with MS Future studies are also needed to address the comor-
should prompt an evaluation for comorbid neurologi- bidities that affect elderly patients with MS. Studies that
cal disease183. Good evidence suggests that cognitive evaluated symptomatic therapies for improving cogni-
reserve moderates the rate of cognitive decline in MS, tion in elderly patients with MS have largely reported
as it does in AD184,185. Cognitive training has shown negative results. For example, donepezil (a centrally
success in improving cognitive functioning, including acting reversible acetylcholinesterase inhibitor used
motor tasks186, working memory187 and quality of life188, to treat cognitive dysfunction in patients with AD)
in patients with MS and should be encouraged for ageing was not effective in patients with MS189. Similarly, the
individuals with cognitive impairment. N-​methyl-d-​aspartate receptor inhibitor memantine
(which modestly improves cognition in patients with
Conclusions AD) did not improve cognitive function in patients
The data consistently show that patients with MS are with MS and had a particularly concerning safety pro-
living longer. However, ageing in this population comes file in this population190,191. Separately from medications,
with a multitude of concerns relating to the complex an emphasis should also be placed on promoting well-
interactions of the disease process with normal age- ness and improving quality of life for patients with MS,
ing, comorbidities and therapy (disease-​modifying including social support and cognitive training.
regimens and symptomatic treatments or therapies for
comorbidities). In addition, ageing patients with MS Published online 18 April 2019

1. Marrie, R. A., Yu, N., Blanchard, J., Leung, S. & 4. Scalfari, A., Neuhaus, A., Daumer, M., Muraro, P. A. & 6. Hirst, C., Swingler, R., Compston, D. A., Ben-​Shlomo, Y.
Elliott, L. The rising prevalence and changing age Ebers, G. C. Onset of secondary progressive phase and & Robertson, N. P. Survival and cause of death in
distribution of multiple sclerosis in Manitoba. long-​term evolution of multiple sclerosis. J. Neurol. multiple sclerosis: a prospective population-​based
Neurology 74, 465–471 (2010). Neurosurg. Psychiatry 85, 67–75 (2014). study. J. Neurol. Neurosurg. Psychiatry 79,
2. Finlayson, M. Concerns about the future among older 5. Koch-​Henriksen, N., Laursen, B., Stenager, E. & 1016–1021 (2008).
adults with multiple sclerosis. Am. J. Occup. Ther. 58, Magyari, M. Excess mortality among patients with 7. Hurwitz, B. J. Analysis of current multiple sclerosis
54–63 (2004). multiple sclerosis in Denmark has dropped registries. Neurology 76 (Suppl. 1), S7–S13 (2011).
3. Tutuncu, M. et al. Onset of progressive phase is an significantly over the past six decades: a population 8. Lunde, H. M. B., Assmus, J., Myhr, K. M., Bo, L. &
age-​dependent clinical milestone in multiple sclerosis. based study. J. Neurol. Neurosurg. Psychiatry 88, Grytten, N. Survival and cause of death in multiple
Mult. Scler. 19, 188–198 (2013). 626–631 (2017). sclerosis: a 60-year longitudinal population study.

NATure ReviewS | NeURology volume 15 | JUNE 2019 | 339


Reviews

J. Neurol. Neurosurg. Psychiatry 88, 621–625 33. Capkun, G. et al. Mortality and comorbidities in for autophagy and senescence cross-​talk. Cells 7,
(2017). patients with multiple sclerosis compared with a E162 (2018).
9. Kingwell, E. et al. Relative mortality and survival in population without multiple sclerosis: an observational 59. Alirezaei, M. et al. Elevated ATG5 expression in
multiple sclerosis: findings from British Columbia. study using the US Department of Defense autoimmune demyelination and multiple sclerosis.
J. Neurol. Neurosurg. Psychiatry 83, 61–66 (2012). administrative claims database. Mult. Scler. Relat. Autophagy 5, 152–158 (2009).
10. Minden, S. L., Frankel, D., Hadden, L. S., Srinath, K. P. Disord. 4, 546–554 (2015). 60. Peterson, J. W. & Trapp, B. D. Neuropathobiology
& Perloff, J. N. Disability in elderly people with 34. Marrie, R. A. et al. Comorbidity delays diagnosis and of multiple sclerosis. Neurol. Clin. 23, 107–129
multiple sclerosis: an analysis of baseline data from increases disability at diagnosis in MS. Neurology 72, (2005).
the Sonya Slifka Longitudinal Multiple Sclerosis Study. 117–124 (2009). 61. Dutta, R. & Trapp, B. D. Relapsing and progressive
Neurorehabilitation 19, 55–67 (2004). 35. Manouchehrinia, A., Tanasescu, R., Tench, C. R. & forms of multiple sclerosis: insights from pathology.
11. Rotstein, D. L. et al. Temporal trends in multiple Constantinescu, C. S. Mortality in multiple sclerosis: Curr. Opin. Neurol. 27, 271–278 (2014).
sclerosis prevalence and incidence in a large meta-​analysis of standardised mortality ratios. 62. Cevenini, E., Monti, D. & Franceschi, C. Inflamm-​
population. Neurology 90, e1435–e1441 (2018). J. Neurol. Neurosurg. Psychiatry 87, 324–331 ageing. Curr. Opin. Clin. Nutr. Metab. Care 16, 14–20
12. Grytten, N., Torkildsen, O. & Myhr, K. M. Time trends (2016). (2013).
in the incidence and prevalence of multiple sclerosis in 36. Scalfari, A. et al. Mortality in patients with multiple 63. Franceschi, C. et al. Inflamm-​aging. An evolutionary
Norway during eight decades. Acta Neurol. Scand. sclerosis. Neurology 81, 184–192 (2013). perspective on immunosenescence. Ann. NY Acad. Sci.
132, 29–36 (2015). 37. Kaufman, D. W. et al. Survival in commercially insured 908, 244–254 (2000).
13. Alla, S., Pearson, J., Debernard, L., Miller, D. & multiple sclerosis patients and comparator subjects in 64. Thewissen, M. et al. Analyses of immunosenescent
Mason, D. The increasing prevalence of multiple the U.S. Mult. Scler. Relat. Disord. 3, 364–371 markers in patients with autoimmune disease.
sclerosis in New Zealand. Neuroepidemiology 42, (2014). Clin. Immunol. 123, 209–218 (2007).
154–160 (2014). 38. Bronnum-​Hansen, H., Koch-​Henriksen, N. & 65. Childs, B. G. et al. Senescent cells: an emerging target
14. Dilokthornsakul, P. et al. Multiple sclerosis prevalence Stenager, E. Trends in survival and cause of death in for diseases of ageing. Nat. Rev. Drug Discov. 16,
in the United States commercially insured population. Danish patients with multiple sclerosis. Brain 127, 718–735 (2017).
Neurology 86, 1014–1021 (2016). 844–850 (2004). 66. Aw, D. & Palmer, D. B. The origin and implication of
15. Solaro, C. et al. The changing face of multiple 39. Ragonese, P., Aridon, P., Salemi, G., D’Amelio, M. & thymic involution. Aging Dis. 2, 437–443 (2011).
sclerosis: prevalence and incidence in an aging Savettieri, G. Mortality in multiple sclerosis: a review. 67. Haegert, D. G. Multiple sclerosis: a disorder of altered
population. Mult. Scler. 21, 1244–1250 (2015). Eur. J. Neurol. 15, 123–127 (2008). T cell homeostasis. Mult. Scler. Int. 2011, 461304
16. Barnett, M. H., Williams, D. B., Day, S., Macaskill, P. 40. Warren, S. A., Janzen, W., Warren, K. G., Svenson, L. W. (2011).
& McLeod, J. G. Progressive increase in incidence & Schopflocher, D. P. Multiple sclerosis mortality rates 68. Musella, A. et al. Interplay between age and
and prevalence of multiple sclerosis in Newcastle, in Canada, 1975–2009. Can. J. Neurol. Sci. 43, neuroinflammation in multiple sclerosis: effects on
Australia: a 35-year study. J. Neurol. Sci. 213, 134–141 (2016). motor and cognitive functions. Front. Aging Neurosci.
(1–6 (2003). 41. Amezcua, L., Rivas, E., Joseph, S., Zhang, J. & Liu, L. 10, 238 (2018).
17. Simpson, S. Jr. et al. Trends in the epidemiology of Multiple sclerosis mortality by race/ethnicity, age, sex, 69. Budni, J., Bellettini-​Santos, T., Mina, F., Garcez, M. L.
multiple sclerosis in Greater Hobart, Tasmania: and time period in the United States, 1999–2015. & Zugno, A. I. The involvement of BDNF, NGF and
1951 to 2009. J. Neurol. Neurosurg. Psychiatry 82, Neuroepidemiology 50, 35–40 (2018). GDNF in aging and Alzheimer’s disease. Aging Dis. 6,
180–187 (2011). 42. Hemmer, B., Kerschensteiner, M. & Korn, T. Role of the 331–341 (2015).
18. Sarasoja, T., Wikstrom, J., Paltamaa, J., Hakama, M. innate and adaptive immune responses in the course 70. Rist, J. M. & Franklin, R. J. Taking ageing into account
& Sumelahti, M. L. Occurrence of multiple sclerosis in of multiple sclerosis. Lancet Neurol. 14, 406–419 in remyelination-​based therapies for multiple sclerosis.
central Finland: a regional and temporal comparison (2015). J. Neurol. Sci. 274, 64–67 (2008).
during 30 years. Acta Neurol. Scand. 110, 331–336 43. Weissert, R. The immune pathogenesis of multiple 71. Luchetti, S. et al. Progressive multiple sclerosis
(2004). sclerosis. J. Neuroimmune Pharmacol. 8, 857–866 patients show substantial lesion activity that
19. Mayr, W. T. et al. Incidence and prevalence of multiple (2013). correlates with clinical disease severity and sex:
sclerosis in Olmsted County, Minnesota, 1985–2000. 44. Garg, N. & Smith, T. W. An update on a retrospective autopsy cohort analysis. Acta
Neurology 61, 1373–1377 (2003). immunopathogenesis, diagnosis, and treatment of Neuropathol. 135, 511–528 (2018).
20. Daltrozzo, T., Hapfelmeier, A., Donnachie, E., multiple sclerosis. Brain Behav. 5, e00362 (2015). 72. Correale, J., Gaitan, M. I., Ysrraelit, M. C. & Fiol, M. P.
Schneider, A. & Hemmer, B. A systematic assessment 45. Fletcher, J. M., Lalor, S. J., Sweeney, C. M., Tubridy, N. Progressive multiple sclerosis: from pathogenic
of prevalence, incidence and regional distribution of & Mills, K. H. G. T cells in multiple sclerosis and mechanisms to treatment. Brain 140, 527–546
multiple sclerosis in Bavaria from 2006 to 2015. experimental autoimmune encephalomyelitis. (2017).
Front. Neurol. 9, 871 (2018). Clin. Exp. Immunol. 162, 1–11 (2010). 73. Buck, D. & Hemmer, B. Treatment of multiple
21. Grassivaro, F. et al. Multiple sclerosis incidence and 46. ‘t Hart, B. A. et al. A B cell-​driven autoimmune sclerosis: current concepts and future perspectives.
prevalence trends in the province of Padua, northeast pathway leading to pathological hallmarks of J. Neurol. 258, 1747–1762 (2011).
Italy, 1965–2018. Neuroepidemiology 52, 41–46 progressive multiple sclerosis in the marmoset 74. Thompson, A. J. et al. Diagnosis of multiple sclerosis:
(2018). experimental autoimmune encephalomyelitis model. 2017 revisions of the McDonald criteria. Lancet
22. Ribbons, K., Lea, R., Tiedeman, C., Mackenzie, L. & Front. Immunol. 8, 804 (2017). Neurol. 17, 162–173 (2018).
Lechner-​Scott, J. Ongoing increase in incidence and 47. Reich, D. S., Lucchinetti, C. F. & Calabresi, P. A. 75. Hammond, K. E. et al. Quantitative in vivo magnetic
prevalence of multiple sclerosis in Newcastle, Multiple sclerosis. N. Engl. J. Med. 378, 169–180 resonance imaging of multiple sclerosis at 7 Tesla
Australia: a 50-year study. Mult. Scler. 23, (2018). with sensitivity to iron. Ann. Neurol 64, 707–713
(1063–1071 (2017). 48. Gandhi, R., Laroni, A. & Weiner, H. L. Role of the (2008).
23. Koch-​Henriksen, N. & Sorensen, P. S. innate immune system in the pathogenesis of multiple 76. Bagnato, F. et al. Tracking iron in multiple sclerosis:
The changing demographic pattern of multiple sclerosis. J. Neuroimmunol. 221, 7–14 (2010). a combined imaging and histopathological study at 7
sclerosis epidemiology. Lancet Neurol. 9, 520–532 49. Kasper, L. H. & Shoemaker, J. Multiple sclerosis Tesla. Brain 134, 3602–3615 (2011).
(2010). immunology: the healthy immune system versus the 77. Absinta, M. et al. Identification of chronic active
24. Bermel, R. A., Rae-​Grant, A. D. & Fox, R. J. Diagnosing MS immune system. Neurology 74 (Suppl. 1), S2–S8 multiple sclerosis lesions on 3 T MRI. AJNR Am.
multiple sclerosis at a later age: more than just (2010). J. Neuroradiol. 39, 1233–1238 (2018).
progressive myelopathy. Mult. Scler. 16, 1335–1340 50. Frischer, J. M. et al. The relation between 78. Vellinga, M. M. et al. Pluriformity of inflammation
(2010). inflammation and neurodegeneration in multiple in multiple sclerosis shown by ultra-​small iron
25. Poser, C. M. & Brinar, V. V. Diagnostic criteria for sclerosis brains. Brain 132, 1175–1189 (2009). oxide particle enhancement. Brain 131, 800–807
multiple sclerosis: an historical review. Clin. Neurol. 51. Frischer, J. M. et al. Clinical and pathological insights (2008).
Neurosurg. 106, 147–158 (2004). into the dynamic nature of the white matter multiple 79. Dwyer, M. G. et al. Atrophied brain lesion volume:
26. Gafson, A., Giovannoni, G. & Hawkes, C. H. sclerosis plaque. Ann. Neurol. 78, 710–721 (2015). a new imaging biomarker in multiple sclerosis.
The diagnostic criteria for multiple sclerosis: from 52. Lassmann, H., van Horssen, J. & Mahad, D. J. Neuroimaging 28, 490–495 (2018).
Charcot to McDonald. Mult. Scler. Relat. Disord. 1, Progressive multiple sclerosis: pathology and 80. Calabrese, M. et al. Cortical lesion load associates
9–14 (2012). pathogenesis. Nat. Rev. Neurol. 8, 647–656 (2012). with progression of disability in multiple sclerosis.
27. Polliack, M. L., Barak, Y. & Achiron, A. Late-​onset 53. Stephenson, E., Nathoo, N., Mahjoub, Y., Dunn, J. F. Brain 135, 2952–2961 (2012).
multiple sclerosis. J. Am. Geriatr. Soc. 49, 168–171 & Yong, V. W. Iron in multiple sclerosis: roles in 81. Calabrese, M. et al. Exploring the origins of grey
(2001). neurodegeneration and repair. Nat. Rev. Neurol. 10, matter damage in multiple sclerosis. Nat. Rev.
28. Delalande, S., De Seze, J., Ferriby, D., Stojkovic, T. & 459–468 (2014). Neurosci. 16, 147–158 (2015).
Vermersch, P. Late onset multiple sclerosis [French]. 54. Grebenciucova, E. & Berger, J. R. Immunosenescence: 82. Filippi, M. et al. Imaging cortical damage and
Rev. Neurol. (Paris) 158, 1082–1087 (2002). the role of aging in the predisposition to neuro-​ dysfunction in multiple sclerosis. JAMA Neurol. 70,
29. Tremlett, H. & Devonshire, V. Is late-​onset multiple infectious complications arising from the treatment of 556–564 (2013).
sclerosis associated with a worse outcome? Neurology multiple sclerosis. Curr. Neurol. Neurosci. Rep. 17, 61 83. Absinta, M., Sati, P. & Reich, D. S. Advanced MRI and
67, 954–959 (2006). (2017). staging of multiple sclerosis lesions. Nat. Rev. Neurol.
30. Hooge, J. P. & Redekop, W. K. Multiple sclerosis with 55. Lassmann, H. Pathology and disease mechanisms in 12, 358–368 (2016).
very late onset. Neurology 42, 1907–1910 (1992). different stages of multiple sclerosis. J. Neurol. Sci. 84. Zurawski, J., Lassmann, H. & Bakshi, R. Use of
31. Koch-​Henriksen, N., Thygesen, L. C., Stenager, E., 333, 1–4 (2013). magnetic resonance imaging to visualize
Laursen, B. & Magyari, M. Incidence of MS has 56. Sanai, S. A. et al. Aging and multiple sclerosis. leptomeningeal inflammation in patients with multiple
increased markedly over six decades in Denmark Mult. Scler. 22, 717–725 (2016). sclerosis: a review. JAMA Neurol. 74, 100–109 (2016).
particularly with late onset and in women. Neurology 57. Plaza-​Zabala, A., Sierra-​Torre, V. & Sierra, A. Autophagy 85. Zivadinov, R. et al. Leptomeningeal contrast
90, e1954–e1963 (2018). and microglia: novel partners in neurodegeneration and enhancement is associated with progression of cortical
32. Marrie, R. A. et al. A systematic review of the aging. Int. J. Mol. Sci. 18, E598 (2017). atrophy in MS: a retrospective, pilot, observational
incidence and prevalence of comorbidity in multiple 58. Aguilera, M. O., Delgui, L. R., Romano, P. S. & longitudinal study. Mult. Scler. 23, 1336–1345
sclerosis: overview. Mult. Scler. 21, 263–281 (2015). Colombo, M. I. Chronic infections: a possible scenario (2016).

340 | JUNE 2019 | volume 15 www.nature.com/nrneurol


Reviews

86. Harrison, D. M. et al. Leptomeningeal enhancement at 112. Kang, J. H., Chen, Y. H. & Lin, H. C. Comorbidities 135. Polman, C. H. et al. A randomized, placebo-​controlled
7 T in multiple sclerosis: frequency, morphology, and amongst patients with multiple sclerosis: a population-​ trial of natalizumab for relapsing multiple sclerosis.
relationship to cortical volume. J. Neuroimaging 27, based controlled study. Eur. J. Neurol. 17, N. Engl. J. Med. 354, 899–910 (2006).
461–468 (2017). 1215–1219 (2010). 136. CAMMS223 Trial Investigators et al. Alemtuzumab
87. Miller, D. H., Barkhof, F., Frank, J. A., Parker, G. J. & 113. Marrie, R. A. et al. Vascular comorbidity is associated versus interferon beta-1a in early multiple sclerosis.
Thompson, A. J. Measurement of atrophy in multiple with more rapid disability progression in multiple N. Engl. J. Med. 359, 1786–1801 (2008).
sclerosis: pathological basis, methodological aspects sclerosis. Neurology 74, 1041–1047 (2010). 137. Cohen, J. A. et al. Alemtuzumab versus interferon
and clinical relevance. Brain 125, 1676–1695 114. Christiansen, C. F. et al. Risk of arterial cardiovascular beta 1a as first-​line treatment for patients with
(2002). diseases in patients with multiple sclerosis: relapsing-​remitting multiple sclerosis: a randomised
88. Zivadinov, R. et al. Clinical relevance of brain atrophy a population-based cohort study. Neuroepidemiology controlled phase 3 trial. Lancet 380, 1819–1828
assessment in multiple sclerosis. Implications for its 35, 267–274 (2010). (2012).
use in a clinical routine. Expert Rev. Neurother. 16, 115. Jadidi, E., Mohammadi, M. & Moradi, T. High risk of 138. Jacobs, L. D. et al. Intramuscular interferon beta-1a
777–793 (2016). cardiovascular diseases after diagnosis of multiple for disease progression in relapsing multiple sclerosis.
89. De Stefano, N. et al. Establishing pathological cut-​offs sclerosis. Mult. Scler. 19, 1336–1340 (2013). The Multiple Sclerosis Collaborative Research Group
of brain atrophy rates in multiple sclerosis. J. Neurol. 116. Marrie, R. A. et al. Rising prevalence of vascular (MSCRG). Ann. Neurol. 39, 285–294 (1996).
Neurosurg. Psychiatry 87, 93–99 (2016). comorbidities in multiple sclerosis: validation of 139. PRISMS (Prevention of Relapses and Disability by
90. Hedman, A. M., van Haren, N. E., Schnack, H. G., administrative definitions for diabetes, hypertension, Interferon β-1a Subcutaneously in Multiple Sclerosis)
Kahn, R. S. & Hulshoff Pol, H. E. Human brain changes and hyperlipidemia. Mult. Scler. 18, 1310–1319 Study Group. Randomised double-​blind placebo-​
across the life span: a review of 56 longitudinal (2012). controlled study of interferon β-1a in relapsing/
magnetic resonance imaging studies. Hum. Brain 117. Geraldes, R. et al. The current role of MRI in remitting multiple sclerosis. Lancet 352, 1498–1504
Mapp. 33, 1987–2002 (2012). differentiating multiple sclerosis from its imaging (1998).
91. Fisher, E., Lee, J. C., Nakamura, K. & Rudick, R. A. mimics. Nat. Rev. Neurol. 14, 199–213 (2018). 140. Gold, R. et al. Placebo-​controlled phase 3 study of oral
Gray matter atrophy in multiple sclerosis: a 118. Zivadinov, R. et al. Cerebral microbleeds in multiple BG-12 for relapsing multiple sclerosis. N. Engl. J. Med.
longitudinal study. Ann. Neurol. 64, 255–265 sclerosis evaluated on susceptibility-​weighted images 367, 1098–1107 (2012).
(2008). and quantitative susceptibility maps: a case-​control 141. Fox, R. J. et al. Placebo-​controlled phase 3 study of
92. Fisniku, L. K. et al. Gray matter atrophy is related to study. Radiology 281, 884–895 (2016). oral BG-12 or glatiramer in multiple sclerosis. N. Engl.
long-​term disability in multiple sclerosis. Ann. Neurol. 119. Ter Telgte, A. et al. Cerebral small vessel disease: J. Med. 367, 1087–1097 (2012).
64, 247–254 (2008). from a focal to a global perspective. Nat. Rev. Neurol. 142. Kappos, L. et al. A placebo-​controlled trial of oral
93. Steenwijk, M. D. et al. Cortical atrophy patterns in 14, 387–398 (2018). fingolimod in relapsing multiple sclerosis. N. Engl. J.
multiple sclerosis are non-​random and clinically 120. Sati, P. et al. The central vein sign and its clinical Med. 362, 387–401 (2010).
relevant. Brain 139, 115–126 (2016). evaluation for the diagnosis of multiple sclerosis: 143. Cohen, J. A. et al. Oral fingolimod or intramuscular
94. Eshaghi, A. et al. Progression of regional grey matter a consensus statement from the North American interferon for relapsing multiple sclerosis. N. Engl. J.
atrophy in multiple sclerosis. Brain 141, 1665–1677 Imaging in Multiple Sclerosis Cooperative. Nat. Rev. Med. 362, 402–415 (2010).
(2018). Neurol. 12, 714–722 (2016). 144. O’Connor, P. et al. Randomized trial of oral
95. Stankoff, B. & Louapre, C. Can we use regional grey 121. Moon, S. Y. et al. Prospective associations between teriflunomide for relapsing multiple sclerosis. N. Engl.
matter atrophy sequence to stage neurodegeneration white matter hyperintensities and lower extremity J. Med. 365, 1293–1303 (2011).
in multiple sclerosis? Brain 141, 1580–1583 (2018). function. Neurology 90, e1291–e1297 (2018). 145. Miller, A. E. et al. Oral teriflunomide for patients with
96. Cristofanilli, M. et al. Progressive multiple sclerosis 122. Srinivasa, R. N. et al. Cardiovascular risk factors a first clinical episode suggestive of multiple sclerosis
cerebrospinal fluid induces inflammatory associated with smaller brain volumes in regions (TOPIC): a randomised, double-​blind, placebo-​
demyelination, axonal loss, and astrogliosis in mice. identified as early predictors of cognitive decline. controlled, phase 3 trial. Lancet Neurol. 13, 977–986
Exp. Neurol. 261, 620–632 (2014). Radiology 278, 198–204 (2016). (2014).
97. Vidaurre, O. G. et al. Cerebrospinal fluid ceramides 123. Kappus, N. et al. Cardiovascular risk factors are 146. US National Library of Medicine. ClinicalTrials.gov
from patients with multiple sclerosis impair neuronal associated with increased lesion burden and brain https://clinicaltrials.gov/ct2/show/NCT03523858
bioenergetics. Brain 137, 2271–2286 (2014). atrophy in multiple sclerosis. J. Neurol. Neurosurg. (2019).
98. Haider, L. et al. The topograpy of demyelination and Psychiatry 87, 181–187 (2016). 147. Kappos, L. et al. Siponimod versus placebo in
neurodegeneration in the multiple sclerosis brain. 124. Jakimovski, D. et al. Hypertension and heart disease secondary progressive multiple sclerosis (EXPAND):
Brain 139, 807–815 (2016). are associated with development of brain atrophy in a double-​blind, randomised, phase 3 study. Lancet
99. Eshaghi, A. et al. Deep grey matter volume loss drives multiple sclerosis: a 5-year longitudinal study. Eur. J. 391, 1263–1273 (2018).
disability worsening in multiple sclerosis. Ann. Neurol. Neurol. 26, 87 (2019). 148. Zhang, T. et al. Examining the effects of comorbidities
83, 210–222 (2018). 125. Kneebone, I. I., Dunmore, E. C. & Evans, E. Symptoms on disease-​modifying therapy use in multiple sclerosis.
100. Azevedo, C. J. et al. Thalamic atrophy in MS: an MRI of depression in older adults with multiple sclerosis Neurology 86, 1287–1295 (2016).
marker of neurodegeneration throughout disease. (MS): comparison with a matched sample of younger 149. Devonshire, V. et al. Relapse and disability outcomes
Ann. Neurol. 83, 223–234 (2018). adults. Aging Ment. Health 7, 182–185 (2003). in patients with multiple sclerosis treated with
101. Cocozza, S. et al. Cerebellar lobule atrophy and 126. Chwastiak, L. et al. Depressive symptoms and severity fingolimod: subgroup analyses of the double-​blind,
disability in progressive MS. J. Neurol. Neurosurg. of illness in multiple sclerosis: epidemiologic study randomised, placebo-​controlled FREEDOMS study.
Psychiatry 88, 1065–1072 (2017). of a large community sample. Am. J. Psychiatry 159, Lancet Neurol. 11, 420–428 (2012).
102. Moroso, A. et al. Posterior lobules of the cerebellum 1862–1868 (2002). 150. Motl, R. W. et al. Validity of the timed 25-foot walk as
and information processing speed at various stages of 127. Patten, S. B., Metz, L. M. & Reimer, M. A. an ambulatory performance outcome measure for
multiple sclerosis. J. Neurol. Neurosurg. Psychiatry Biopsychosocial correlates of lifetime major depression multiple sclerosis. Mult. Scler. 23, 704–710 (2017).
88, 146–151 (2016). in a multiple sclerosis population. Mult. Scler. 6, 151. Hervault, M., Balto, J. M., Hubbard, E. A. & Motl, R. W.
103. Kearney, H., Miller, D. H. & Ciccarelli, O. Spinal cord 115–120 (2000). Reliability, precision, and clinically important change
MRI in multiple sclerosis — diagnostic, prognostic and 128. Beal, C. C., Stuifbergen, A. K. & Brown, A. of the Nine-​Hole Peg Test in individuals with multiple
clinical value. Nat. Rev. Neurol. 11, 327–338 (2015). Depression in multiple sclerosis: a longitudinal sclerosis. Int. J. Rehabil. Res. 40, 91–93 (2017).
104. Abdel-​Aziz, K. et al. Evidence for early analysis. Arch. Psychiatr. Nurs. 21, 181–191 (2007). 152. Kapoor, R. et al. Effect of natalizumab on disease
neurodegeneration in the cervical cord of patients with 129. Roy, S. et al. Preliminary investigation of cognitive progression in secondary progressive multiple
primary progressive multiple sclerosis. Brain 138, function in aged multiple sclerosis patients: challenges sclerosis (ASCEND): a phase 3, randomised,
1568–1582 (2015). in detecting comorbid Alzheimer’s disease. Mult. Scler. double-​blind, placebo-​controlled trial with an
105. Tsagkas, C. et al. Preferential spinal cord volume loss Relat. Disord. 22, 52–56 (2018). open-​label extension. Lancet Neurol. 17, 405–415
in primary progressive multiple sclerosis. Mult. Scler. 130. U.S. Food and Drug Administration. FDA News Release: (2018).
https://doi.org/10.1177/1352458518775006 FDA approves new oral treatment for multiple sclerosis. 153. Shirani, A. et al. Multiple sclerosis in older adults:
(2018). FDA https://www.fda.gov/NewsEvents/Newsroom/ the clinical profile and impact of interferon beta
106. Schlaeger, R. et al. Association between thoracic spinal PressAnnouncements/ucm634837.htm (2019). treatment. Biomed. Res. Int. 2015, 451912 (2015).
cord gray matter atrophy and disability in multiple 131. U.S. Food and Drug Administration. FDA News Release: 154. Giovannoni, G. et al. Is multiple sclerosis a length-​
sclerosis. JAMA Neurol. 72, 897–904 (2015). FDA approves new oral drug to treat multiple sclerosis. dependent central axonopathy? The case for
107. Tsagkas, C. et al. Spinal cord volume loss: a marker of FDA https://www.fda.gov/NewsEvents/Newsroom/ therapeutic lag and the asynchronous progressive MS
disease progression in multiple sclerosis. Neurology PressAnnouncements/ucm634469.htm (2019). hypotheses. Mult. Scler. Relat. Disord. 12, 70–78
91, e349–e358 (2018). 132. National Multiple Sclerosis Society. Disease-​modifying (2017).
108. Zeydan, B. et al. Cervical spinal cord atrophy: an early therapies for MS. NationalMSSociety https://www. 155. Montalban, X. et al. Ocrelizumab versus placebo in
marker of progressive MS onset. Neurol. Neuroimmunol. nationalmssociety.org/NationalMSSociety/media/ primary progressive multiple sclerosis. N. Engl. J.
Neuroinflamm. 5, e435 (2018). MSNationalFiles/Brochures/Brochure-​MS-Disease-​ Med. 376, 209–220 (2017).
109. Murtonen, A., Kurki, S., Hanninen, K., Modifying-Medications.pdf (2018). 156. Paz Soldan, M. M. et al. Relapses and disability
Soilu-​Hanninen, M. & Sumelahti, M. L. Common 133. Johnson, K. P. et al. Copolymer 1 reduces relapse rate accumulation in progressive multiple sclerosis.
comorbidities and survival in MS: risk for stroke, type 1 and improves disability in relapsing-​remitting multiple Neurology 84, 81–88 (2015).
diabetes and infections. Mult. Scler. Relat. Disord. 19, sclerosis: results of a phase III multicenter, double-​ 157. Wolinsky, J. S. et al. Evaluation of no evidence of
109–114 (2018). blind placebo-​controlled trial. The Copolymer 1 progression or active disease (NEPAD) in patients
110. Tettey, P., Simpson, S. Jr., Taylor, B. V. & Multiple Sclerosis Study Group. Neurology 45, with primary progressive multiple sclerosis in the
van der Mei, I. A. Vascular comorbidities in the onset 1268–1276 (1995). ORATORIO trial. Ann. Neurol. 84, 527–536 (2018).
and progression of multiple sclerosis. J. Neurol. Sci. 134. IFNβ Multiple Sclerosis Study Group. Interferon beta-​ 158. Fox, E. J. et al. Ocrelizumab reduces progression of
347, 23–33 (2014). lb is effective in relapsing-​remitting multiple sclerosis. upper extremity impairment in patients with primary
111. Hussein, W. I. & Reddy, S. S. Prevalence of diabetes in I. Clinical results of a multicenter, randomized, double-​ progressive multiple sclerosis: findings from the
patients with multiple sclerosis. Diabetes Care 29, blind, placebo-​controlled trial. 1993 [classical article]. phase III randomized ORATORIO trial. Mult. Scler. 24,
1984–1985 (2006). Neurology 57 (Suppl. 5), S3–S9 (2001). 1862–1870 (2018).

NATure ReviewS | NeURology volume 15 | JUNE 2019 | 341


Reviews

159. Manouchehrinia, A. et al. Age related multiple viscoelasticity in multiple sclerosis: a novel application multiple sclerosis: a randomized, placebo-​controlled
sclerosis severity score: disability ranked by age. of magnetic resonance elastography. Neuroradiology study. J. Neurol. Sci. 363, 69–76 (2016).
Mult. Scler. 23, 1938–1946 (2017). 59, 61–67 (2017). 192. Hartung, H. P. et al. Mitoxantrone in progressive
160. Kister, I. Disease-​modifying therapies can be safely 176. US National Library of Medicine. ClinicalTrials.gov multiple sclerosis: a placebo-​controlled, double-​blind,
discontinued in an individual with stable relapsing-​ https://clinicaltrials.gov/ct2/show/NCT02282878 randomised, multicentre trial. Lancet 360,
remitting MS — YES. Mult. Scler. 23, 1188–1190 (2017). 2018–2025 (2002).
(2017). 177. US National Library of Medicine. ClinicalTrials.gov 193. Confavreux, C. et al. Oral teriflunomide for
161. Tobin, W. O. & Weinshenker, B. G. Disease-​modifying https://clinicaltrials.gov/ct2/show/NCT03718247 patients with relapsing multiple sclerosis (TOWER):
therapies can be safely discontinued in an individual (2018). a randomised, double-​blind, placebo-​controlled,
with stable relapsing-​remitting MS — NO. Mult. Scler. 178. US National Library of Medicine. ClinicalTrials.gov phase 3 trial. Lancet Neurol. 13, 247–256 (2014).
23, 1190–1192 (2017). https://clinicaltrials.gov/ct2/show/NCT03808545 194. Hauser, S. L. et al. Ocrelizumab versus interferon
162. Tremlett, H., Zhao, Y., Joseph, J. & Devonshire, V. (2019). beta-1a in relapsing multiple sclerosis. N. Engl.
Relapses in multiple sclerosis are age- and time-​ 179. Ploughman, M. et al. Factors influencing healthy aging J. Med. 376, 221–234 (2017).
dependent. J. Neurol. Neurosurg. Psychiatry 79, with multiple sclerosis: a qualitative study. Disabil.
1368–1374 (2008). Rehabil. 34, 26–33 (2012). Acknowledgements
163. Hua, L. H., Fan, T. H., Conway, D., Thompson, N. & 180. Harrison, T., Blozis, S. & Stuifbergen, A. Longitudinal The authors’ research is supported in part by grants from the
Kinzy, T. G. Discontinuation of disease-modifying predictors of attitudes toward aging among women National Multiple Sclerosis Society (HC 1411–02004) and
therapy in patients with multiple sclerosis over with multiple sclerosis. Psychol. Aging 23, 823–832 Biogen Idec (US-​M SG-15-10855) to B.W.-G. and from
age 60. Mult. Scler. https://doi.org/10.1177/ (2008). Advancing Research in Multiple Sclerosis (ARMS) to the
1352458518765656 (2018). 181. Julian, L. J. Cognitive functioning in multiple sclerosis. Jacobs Multiple Sclerosis Center for Treatment and Research.
164. Kister, I. et al. Discontinuing disease-​modifying Neurol. Clin. 29, 507–525 (2011).
therapy in MS after a prolonged relapse-​free period: 182. Bodling, A. M., Denney, D. R. & Lynch, S. G. Cognitive Author contributions
a propensity score-​matched study. J. Neurol. aging in patients with multiple sclerosis: a cross-​ C.B.V., D.J., K.S.K., R.Z. and B.W.-G. were involved in all
Neurosurg. Psychiatry 87, 1133–1137 (2016). sectional analysis of speeded processing. Arch. Clin. aspects of article preparation. M.R. and R.H.B.B. contributed
165. Weideman, A. M., Tapia-​Maltos, M. A., Johnson, K., Neuropsychol. 24, 761–767 (2009). to discussions of the article content, writing and review or
Greenwood, M. & Bielekova, B. Meta-​analysis of the 183. Roy, S. et al. Differential effects of aging on motor and editing of the manuscript before submission.
age-​dependent efficacy of multiple sclerosis cognitive functioning in multiple sclerosis. Mult. Scler.
treatments. Front. Neurol. 8, 577 (2017). 23, 1385–1393 (2017). Competing interests
166. Sormani, M. P. et al. Autologous hematopoietic stem 184. Sumowski, J. F., Chiaravalloti, N. & DeLuca, J. C.B.V. declares that she has received consultancy fees from
cell transplantation in multiple sclerosis: a meta-​ Cognitive reserve protects against cognitive Merck/EMD Serono. M.R. declares that he has received
analysis. Neurology 88, 2115–2122 (2017). dysfunction in multiple sclerosis. J. Clin. Exp. research funding from the US National Institute of
167. Muraro, P. A. et al. Autologous haematopoietic stem Neuropsychol. 31, 913–926 (2009). Neurological Disorders and Stroke and the US National
cell transplantation for treatment of multiple sclerosis. 185. Benedict, R. H., Morrow, S. A., Weinstock Guttman, B., Science Foundation. R.H.B.B. declares that he has received
Nat. Rev. Neurol. 13, 391–405 (2017). Cookfair, D. & Schretlen, D. J. Cognitive reserve research support from Accorda, Biogen, Genzyme,
168. Muraro, P. A. et al. Long-​term outcomes after moderates decline in information processing speed in Mallinckrodt and Novartis; consultancy fees from Biogen,
autologous hematopoietic stem cell transplantation multiple sclerosis patients. J. Int. Neuropsychol. Soc. Genentech, Genzyme, Novartis, Roche, Sanofi and Teva; and
for multiple sclerosis. JAMA Neurol. 74, 459–469 16, 829–835 (2010). compensation for activities relating to continuing medical
(2017). 186. Charvet, L. E., Shaw, M. T., Haider, L., Melville, P. & education from EMD Serono. R.Z. declares that he has
169. US National Library of Medicine. ClinicalTrials.gov Krupp, L. B. Remotely-​delivered cognitive remediation received speakers’ and consultancy fees from Celgene, Claret
https://clinicaltrials.gov/ct2/show/NCT03073603 in multiple sclerosis (MS): protocol and results from a Medical, EMD Serono, Genzyme-​Sanofi, IMS Health, Novartis
(2019). pilot study. Mult. Scler. J. Exp. Transl Clin. 1, and Roche-​Genentech and financial research support from
170. Moss, B. P., Rensel, M. R. & Hersh, C. M. Wellness and 2055217315609629 (2015). Claret Medical, IMS Health, Intekrin, Genzyme-​Sanofi and
the role of comorbidities in multiple sclerosis. 187. Perez-​Martin, M. Y., Gonzalez-​Platas, M., Eguia-​Del Novartis. B.W.-G. declares that she has received fees for con-
Neurotherapeutics 14, 999–1017 (2017). Rio, P., Croissier-​Elias, C. & Jimenez Sosa, A. Efficacy sultancy, acting as a speaker and serving on the scientific
171. Ramanujam, R. et al. Effect of smoking cessation on of a short cognitive training program in patients with advisory boards of Biogen Idec, EMD Serono, Genzyme-​
multiple sclerosis prognosis. JAMA Neurol. 72, multiple sclerosis. Neuropsychiatr. Dis. Treat. 13, Sanofi, Novartis, Questcor and Teva Neuroscience, and finan-
1117–1123 (2015). 245–252 (2017). cial research support from Aspreva, Biogen Idec, EMD
172. D’Hooghe, M. B., Haentjens, P., Nagels, G. & 188. Grasso, M. G. et al. Evaluation of the impact of Serono, Genzyme, the Immune Tolerance Network Clinical
De Keyser, J. Alcohol, coffee, fish, smoking and disease cognitive training on quality of life in patients Trials Group, the National Multiple Sclerosis Society, the NIH
progression in multiple sclerosis. Eur. J. Neurol. 19, with multiple sclerosis. Eur. Neurol. 78, 111–117 (not related to the present work), Novartis and Teva
616–624 (2012). (2017). Neuroscience. D.J. and K.S.K. declare no competing
173. Manouchehrinia, A. et al. Tobacco smoking and 189. O’Carroll, C. B. et al. Is donepezil effective for interests.
disability progression in multiple sclerosis: United multiple sclerosis-​related cognitive dysfunction?
Kingdom cohort study. Brain 136, 2298–2304 (2013). A critically appraised topic. Neurologist 18, 51–54 Publisher’s note
174. Sandroff, B. M., Motl, R. W., Scudder, M. R. & (2012). Springer Nature remains neutral with regard to jurisdictional
DeLuca, J. Systematic, evidence-​based review of 190. Villoslada, P., Arrondo, G., Sepulcre, J., Alegre, M. & claims in published maps and institutional affiliations.
exercise, physical activity, and physical fitness effects Artieda, J. Memantine induces reversible neurologic
on cognition in persons with multiple sclerosis. impairment in patients with MS. Neurology 72, Reviewer information
Neuropsychol. Rev. 26, 271–294 (2016). 1630–1633 (2009). Nature Reviews Neurology thanks M. Magyari and other
175. Sandroff, B. M., Johnson, C. L. & Motl, R. W. 191. Peyro Saint Paul, L. et al. Efficacy and safety profile of anonymous reviewer(s) for their contribution to the peer
Exercise training effects on memory and hippocampal memantine in patients with cognitive impairment in review of this work.

342 | JUNE 2019 | volume 15 www.nature.com/nrneurol

You might also like