Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Journal of Neurology

https://doi.org/10.1007/s00415-019-09489-5

REVIEW

A focus on secondary progressive multiple sclerosis (SPMS): challenges


in diagnosis and definition
Hernan Inojosa1 · Undine Proschmann1 · Katja Akgün1 · Tjalf Ziemssen1 

Received: 2 July 2019 / Revised: 24 July 2019 / Accepted: 26 July 2019


© Springer-Verlag GmbH Germany, part of Springer Nature 2019

Abstract
Secondary progressive multiple sclerosis (SPMS) is the second most common form of multiple sclerosis (MS). One in two
relapse remitting multiple sclerosis (RRMS) patients will develop SPMS within 15 years and up to two-thirds after 30 years,
leading to a progressive decrease of neurological function and limitation of daily activities. Nevertheless, the SPMS diagnosis
is often established retrospectively and delayed up to 3 years due to several patient- and clinician-related factors. Definitive
clinical diagnostic criteria are lacking and research is currently ongoing to identify imaging and biochemical biomarkers.
As new therapies are introduced, early SPMS diagnosis may represent a window of opportunity for intervention. New
approaches, endpoints or technologies could help physicians establishing a diagnosis. Here, we review SPMS in relation to
its diagnostic and definition challenges and current screening techniques and tools.

Keywords  SPMS · Diagnosis · Definition · Screening · Endpoints

Introduction of MS with a prevalence that varies geographically with up


to 57.8/100.000 in the UK, 37.1/100.000 in the USA and
Multiple sclerosis (MS) is a chronic inflammatory disease 33.3/100.000 in Germany [1].
of the central nervous system (CNS). The affected areas in Even though in the last decades, several disease-mod-
the CNS are multifocal and variable, so are natural history ifying therapies (DMT) have been developed for RRMS,
and individual clinical manifestation. Most of the patients for SPMS there are still few therapeutic approaches. Not
begin with a relapsing remitting phenotype of MS (RRMS) until March 2019, Siponimod was approved in the US as
characterized by episodic symptoms and signs (attacks or first treatment for SPMS [3]. Although the early diagnosis
relapses) with full or partial recovery and no disease pro- of SPMS or transition from RRMS can be difficult for the
gression between relapses. physician and confusing for the patients, it could open a win-
After some years, many of these patients present a grad- dow of opportunities for disease treatment. We review these
ual decline of the neurological function with and without challenges on SPMS and the current status on diagnosis and
relapses, which is characteristic for the evolution of sec- prognosis of this disease subtype.
ondary progressive disease (SPMS). SPMS is distinguished
from primary progressive MS (PPMS) by its distinct course,
which necessarily includes and follows an initial course of From RRMS to SPMS
RRMS [1]. It has been reported that 85–90% of patients
have RRMS at the time of diagnosis and up to eight in As commented above, up to 80% of patients could go from
every ten of them may advance to SPMS within 20 years RRMS to SPMS within 20 years [2]. However, epidemio-
[2]. Worldwide, SPMS is the second most common form logical data concerning secondary progression should be
carefully depicted considering the use of DMT, as it could
modify the natural history of the disease. Data based on
* Tjalf Ziemssen
Tjalf.Ziemssen@uniklinikum‑dresden.de cohort of patients who have not received DMTs suggest that
this conversion occurred in more than one-third of patients
1
Department of Neurology, Center of Clinical Neuroscience, within 8 years, one in two after 15 years and two in three
Carl Gustav Carus University Clinic, University Hospital after 30 years [4, 5]. Nevertheless, thanks to newer treatment
of Dresden, Fetscherstr. 74, 01307 Dresden, Germany

13
Vol.:(0123456789)
Journal of Neurology

Fig. 1  Illustration of the transition phase on the natural history of ease. As these mechanisms are exhausted, patients are more easily
MS. While patients are on the transition phase from RRMS to SPMS, aware of newer and more progressive symptoms. Similarly, approved
the brain reserve, cognitive reserve and other compensatory mecha- treatment options decrease as the disease progresses
nisms may cover early clinical manifestations of the progressive dis-

options the conversion rates may be much lower in future. role to these mechanisms [13, 14]. The patients may only
A study showed that after more than 16 years of follow-up, become aware of worsening when these compensatory
11.3% of a treated cohort transitioned to SPMS, numbers mechanisms are exhausted later after a transition phase to
much lower than the 36–50% expected considering previ- SPMS [15, 16]. These compensatory mechanisms can also
ous reports from the pre-treatment era [6]. Another study be an obstacle on the SPMS diagnosis in clinical practice
reported a longer time to progression in patients treated with [17] (Fig. 1).
first-generation DMTs compared with historical data from Recently, the concept of “silent progression” has been
patients of the same registry and after carefully adjusting to proposed by Cree et al. as an interesting term in the con-
different variables [7]. text of the subtle emergence of a neurodegenerative process
Pathophysiologically, the underlying neurodegenerative that cannot be early recognized on the evaluation of RRMS
processes at the CNS are already present in the asympto- patients [18]. On the same study, it was determined that
matic phase of MS. The main reason why people with MS do relapse activity was not associated with long-term disabil-
not notice they have SPMS in early stages of their disease is ity progression after 10-year follow-up even though it may
that the brain has the ability to compensate for neuronal loss contribute to relative brain volume loss. On the other hand,
[8]. The exact nature of these compensating mechanisms development of new lesions could increase the risk of long-
remains unknown, but some authors have postulated a larger term disability progression and further SPMS [18].
extra-region neuronal recruitment, increased homologous Women who have had a multifocal syndrome or were
area activation of additional brain areas and neuronal plas- older than 34 years at disease onset and/or have had treat-
ticity in form of local expansion (specially around lesions), ment failure were at increased risk of conversion to SPMS
which have been proven by neurophysiological and neuro- [4, 19]. The presence of motor symptoms at onset and male
imaging studies [9–12]. gender was strongly associated with shorter time to SPMS
The brain reserve (seen as maximal lifetime brain vol- development in untreated patients, likewise younger age at
ume), respectively, cognitive reserve could also have a pro- onset was associated with longer time to SPMS conversion
tective effect against progression and may play a supportive but also with younger age at progression [20].

13
Journal of Neurology

The effect of DMTs on conversion is still not universally patients and physicians do not detect new symptoms and
accepted. Although the use of DMTs in RRMS patients has limitations. The exact importance of this adjustment on the
shown to reduce the proportion of patients progressing to SPMS diagnosis is still not well described.
SPMS [6, 21–23], a long-term follow-up study reported sim- On the progressive phase, those MS symptoms that
ilar time to progression onset and clinical course in treated the patients had during the relapsing–remitting stage may
patients compared to natural history studies [4]. The use of become more difficult to cope with, having to experience
DMT may also slow brain volume loss preserving the brain additionally new complaints [16]. Patients with SPMS
reserve, which may have an impact on the onset of SPMS describe many physical and mental manifestations that
[13, 18]. can restrict their activity and worsen disability [16, 28].
Fatigue, as one of the most commonly reported symptoms
of MS (up to 75–90% at some point in time), plays an
The diagnostic challenge of SPMS—patients’ important role, and, together with the reduced mobility,
perspective can be one of the most difficult symptoms to deal with
[16, 37].
A key characteristic of MS has been the presence of multiple However, there is a day-to-day variation of neurological
lesions affecting diverse anatomical locations at different symptoms. MS symptoms variate with no clear replicable
time points. This phenomenon has been considered since pattern and related to stress. This variation could be chal-
its first formal description 150 years ago by Jean Martin lenging to understand by the patients and also an obstacle
Charcot until nowadays, being an important factor of the objectively detecting new symptoms or progression by the
current diagnostic criteria [24, 25]. physicians [38–40].
There is a great heterogeneity of clinical course, symp- The transition from RRMS to SPMS can be critical for
toms and response to therapy associated with different the patient at a psychosocial point of view, with uncer-
immunopathological patterns [14]. They can evolve differ- tainty and the feeling of being diagnosed again with a
ently between patients over time and involve all body sub- different disease, with fewer or no treatment options [41,
systems [16, 26, 27]. In addition, the progression of disease 42]. Communication skills of the treating neurologist with
can be differently modulated by genetic und epigenetic fac- concrete strategies should target this aspect of the disease.
tors varying from one patient to the other. The diagnostic process should be transparent to help the
Therefore, patients may notice subtle and different patients understand the meaning of this new phase [16].
changes that cause functional deficits and place further
burden on their daily living [28]. They may be aware of
an increase in the severity or intensity of their symptoms
and the appearance or worsening of some of MS-related Clinicians’ challenges
symptoms, such as spasticity and bladder or bowel prob-
lems [29–32]. Specific tasks such as sports activities or puz- For the clinicians, the diagnosis of the conversion from
zle solving may become more difficult in this early stage RRMS to SPMS is challenging (Fig.  2). There are no
of SPMS [30]. In RRMS patients who already experienced clear clinical, imaging, immunological or pathological
these symptoms for a while, these subtle changes may be criteria that define the beginning of SPMS or transition
difficult to detect and communicate. With the time, patients to the progressive phase of the disease [17, 43]. Clini-
accustom to their situation with impairments and disabili- cians frequently diagnose SPMS retrospectively, what
ties, reassessing their perceived limitations of everyday life means a delay of SPMS diagnosis by up to 3 years after a
[33, 34]. period of diagnostic uncertainty [44]. This steadily pro-
Compensatory processes at the CNS may delay the mani- gression of baseline symptoms is independent of relapses
festation of a progressive disease (Fig. 1). Every patient is or remission, which are much more easily perceived by
different and can have a different heritable neurological or the physician and patient as an intermittent neurological
brain reserve, modifiable cognitive reserve, or neuroplasti- deterioration [45]. It has been seen that up to two-thirds
city capacity [13]. of patients with insidious worsening of disability are still
The psychosocial adjustment or adaptation to the considered by clinicians to have RRMS [18]. Together
symptoms and disabilities secondary to MS is an impor- with this uncertainty recognizing the transition to SPMS
tant aspect of the therapeutic approach on patients with and lack of objective tests, the physicians tend to be more
MS, necessary to manage new disease demands [35]. This conservative than the patients regarding disease progres-
was better in those in remission than in those with SPMS sion to avoid the categorization on a final phase of the
[36]. On the other hand, this adaption on patients still with disease, sometimes also with an emotional component and
RRMS could also delay the diagnosis of SPMS, as the limited treatment options [8, 46].

13
Journal of Neurology

Fig. 2  Challenges and future  approaches on the definition of SPMS. adoption of a “Best definition”, development of screening tools and
The lack of standardization, diagnosis delay, difficult communication adoption of newer clinical endpoints and outcome measures may be a
by subtle symptomatic changes and low sensitivity of the EDSS as support for the physicians on the clinical practice
single diagnostic measure hinder a prompt diagnosis of SPMS. The

Defining SPMS It is based on frequent EDSS evaluations and sugges-


tive of SPMS with a worsening in the EDSS step (1.0-point
Several definitions of progressive disease and SPMS have with EDSS ≤ 5.5 or 0.5-point with EDSS ≥ 6.0), with a
been discussed, e.g. by G. Ebers on retrospective clinical minimum score of 4.0 and a pyramidal functional system
trial data or the Cambridge definition [45, 47]. Lublin et al. score of ≥ 2. This worsening should be confirmed at least
defined progressive disease as a steadily increasing, objec- for 3 months within the same functional system score (FS)
tively documented neurologic dysfunction or disability that leads to progression. This definition identified 18% of
without unequivocal recovery, admitting fluctuations and patients converting to SPMS compared with 14% who had
stability phases [17]. physician-diagnosed SPMS not using the definition, with a
Tightening up these definitions may facilitate earlier good sensitivity (89%) and specificity (86%). Importantly,
diagnosis and, as Lublin suggests, already existing datasets it also enabled a diagnosis 3 years earlier than physician-
might provide answers on this problem. diagnosed SPMS [43].
Some SPMS predictor tools have been developed with The required time to confirm symptomatic disability pro-
different approaches, parameters and output [43, 48, 49] gression has also been deeply discussed on different studies
(Table 1). On a large cohort study (MSBase) and based on as one of the most important factors concerning the stabil-
a retrospective analysis of data from 17.356 patients with ity of the detected progression. Lublin defines “confirmed
MS and after analysis of 576 possible SPMS definitions, a progression” as an increase in neurological dysfunction
‘best definition’ was developed that can be used in clinical throughout a defined time interval (for example, 3, 6 or
trials to enable comparability of future research studies [43]. 12 months) [17]. Similarly, Lorscheider et al. evaluated the

13
Journal of Neurology

Table 1  Overview of published SPMS predictor tools


msprediction.com SPMS nomogram Definitions of SPMS
MSARD (2014) [48] MSJ (2019) [49] Brain 2016 Lorscheider [43]

Study centre Gothenburg Karolinska MSBase


Source of data Of 306 untreated MS patients, 157 were diagnosed with Derivation cohort Swedish national registry SMSreg Analyzed 576 definitions of SPMS (based on EDSS, FS,
RRMS; 118/157 converted to SPMS; median observa- n = 8825 (of which 1488 converted to SPMS); patients confirmed progression and relapses)
tion time 11.5 years; Gothenburg Incidence Cohort included during 1940–2000, follow-up till 2016 N = 17,356 patients (June 2015) of which n = 2360 SPMS,
3 validation cohorts min follow-up 1 year
  British Columbia, n = 3967 (1980–2004), follow-up till
2008
  ACROSS and FREEDOMS
Approach used Onset of SPMS determined retrospectively based on Survival model containing selected variables based Comparing the different definitions to consensus diagno-
Lublin et al. [17]; continuous hazard functions/Poisson on literature with final selection based on backward sis of 3 neurologists
regression model selection model, Bayesian information criterion, and Sample of n = 200 pts of which n = 57 SPMS
prediction accuracy; SPMS based on Lublin et al. [17] Evaluation of long-term outcome—5 years
(retrospective in FREEDOMS)
Parameters Significant independent predictors: age, time since last Gender, age (year of birth), first recorded EDSS, age at Best definition: EDSS progression confirmed for
relapse; symptoms of last relapse and recovery disease onset (relapses and MRI not predictive) ≥ 3 months (1 point if EDSS < 6.0 or 0.5 if EDSS ≥ 6)
Not significant: gender, age at onset, number of relapses in the absence of a relapse, with minimum EDSS 4, and
and nature of first relapse pyramidal FS score 2
Output Estimates current risk of SPMS; expressed as a percent- Estimates risk of conversion to SPMS at 10, 15 and The data-derived algorithm was primarily designed for
age 20 years application to clinical trial data and to enable compara-
bility of future research studies

SPMS secondary progressive multiple sclerosis, EDSS expanded disease disability scale, FS functional system

13
Journal of Neurology

sensitivity and specificity of definitions considering 3, 6, tool, adding the T25FW and 9HPT to the regular neurologi-
12 and 24 months as confirmation period. The definitions cal examination [55]. A change of ≥ 20% for T25FW and
that led to the earliest diagnosis of SPMS were those with 9HPT, both parts of the Multiple Sclerosis Functional Com-
confirmation period of 6 months combined with a minimum posite Measure (MSFC), may be more sensitive on detecting
EDSS score of 3.0 or a confirmation period of 3 months with disease impact and progression and has been even demon-
a minimum EDSS score of 4.0. The definition with 3 months strated in cohorts of exclusively SPMS patients [54, 58–61].
as required confirmation was considered as the ideal with New outcome measures could be used as endpoints for
good accuracy and sensitivity despite lower specificity com- patients with RRMS, such as the confirmed disability pro-
pared to longer periods [43]. gression (CDP), that uses the parameter described also on
However, it has been reported that a short-term con- the EDSS-Plus; the progression independent of relapse
firmation period of just 3 or 6 months could overestimate activity (PIRA), helping to distinguish the progression of
the accumulation of disability by 30%, respectively, 26% disability; or the “no evidence of disease activity” (NEDA)
with low long-term progression stability and great risk of outcome [43, 62, 63]. On a similar order of ideas, the use
disability regression [50]. This was also considered on the of a cognition markers such as the Symbol Digit Modali-
MSBase definition: the psychometric characteristics of the ties Test (SDMT) combined with the EDSS could probably
EDSS (with lower variability at higher scores) and lower detect more progression events measuring different aspects
fluctuation after reaching certain level of disability allowed of disability [64].
a shorter confirmation period as long as a minimum disabil- These measures should be defined as a complement to
ity step and pyramidal FS where established. Added-on, the clinical history. An integration of new and old methods with
exclusion of progression events related to relapse activity the classical clinical history, in combination with several
had great influence on progression stability [43]. Therefore, patient outcome measures, could be key for the detection
using the EDSS as clinical outcome and with the definition of early progressive disease.
limits proposed, a 3-month confirmation period could be
enough at the definition of SPMS.
On the setting of clinical trials, the evaluation of events Screening tools for SPMS
unrelated to relapses is usually realized using the baseline
EDSS as a reference. A recent study by Kappos et al. has The relationship between physicians and patients is a chal-
shown that a roving EDSS reference, whereas the obtained lenging aspect. The physicians need excellent communica-
EDSS Step is compared with the last visit instead of the tive practices to detect subtle disease changes [65]. In addi-
baseline, could be more sensitive on this goal. With this sub- tion, physicians tend to avoid discussion of the emotional
tle change, 50% more overall worsening events and approxi- and social aspects of disease changes, wishing to avoid
mately 70% more progression events were detected than the alarming patients with news that they are entering the final
regular methods [51]. This could change the way clinical phase of MS disease [28, 44, 65]. Similarly, the lack of evi-
trials are designed and have a more realistic relationship dence-based treatments may obstruct the application of a
with the daily medical practice, where the EDSS is often “progressive” label.
compared with the last visit instead of the baseline. To help surmount such factors that contribute to diag-
In clinical practice, a detailed neurological examination nostic delay and under-diagnosis, screening tools, such as
leading to the Neurostatus-EDSS is not standard of care. the MS Progression Discussion Tool [66, 67], the MS pre-
That is why, instead of neurological examination and EDSS, diction score [48] or the SPMS nomogram [49], have been
a careful standardized neurological history and additional developed and are on their way into clinical practice. This
functional testing including specific functional systems could help clinicians quickly and more accurately to detect
as, e.g. cognition and vision can further help to assist with the early, and often subtle, signs of progression of SPMS in
an earlier diagnosis of SPMS. In addition, the EDSS as an a standardized manner.
assessment tool has been widely criticized due to its lack The MS Progression Discussion Tool, in example, inte-
of linearity, low responsiveness, sensitivity and inter-rater grates computer technologies in a digital tool based on a
reliability [52, 53]. A silent progression during RRMS phase set of weighted questions regarding relapses, symptoms
could not be detected due to EDSS limitations [18]. There- and impacts experienced by the patient in the last 6 months
fore, different outcome measures could be included on the (Fig.  3). It uses quantitative and qualitative methods as
diagnosis such as the Timed 25-Foot Walk (T25FW), the approach to evaluate subtle signs suggestive of progres-
9-Hole Peg Test (9HPT), low contrast letter acuity test and sion to differentiate RRMS from early SPMS. The variables
cognitive function testing [34, 54–57]. associated to the nature of the symptoms (e.g. intermittent
A modified version of the conventional EDSS called vs. persistent, stable vs. worsening) and the presence of
EDSS-Plus has been proposed as an interesting evaluation ambulatory, mobility and cognitive symptoms where most

13
Journal of Neurology

Fig. 3  MS Progression Discussion Tool [67]. Screening questions are impact of these symptoms. Symptoms evaluated involve following
addressed in three main areas considering the last 6 months: disease domains: visual, sensory, ambulatory, bladder and bowl, coordination
activity (also relapse and recovery, MRI), symptoms experienced and and balance, cognition, fatigue, speech and pain

significantly associated to early SPMS [66]. This tool could we summarize the current progress on the most important
differentiate between RRMS and SPMS with high sensitivity parameters and their possible future uses.
and specificity without need of an EDSS assessment.
Similarly, the SPMS nomogram emerges as the first vali- Imaging
dated graphical tool for representation of the patient’s risk
of developing SPMS within 10, 15 and 20 years. It uses data Imaging measures of progression are not established or
available on routine clinical setting, such as year of birth, standardized as phenotype descriptors for individual patients
sex, onset age, and first recorded EDSS to accurate predict although many markers are under consideration [71]. Newer
evolution [49]. non-conventional MRI technologies have been developed
improving the quality of the obtained data, with more tissue-
specific imaging tools and more sensitive measurements to
Biomarkers in SPMS detect grey matter lesions in vivo and progression in MS.
Some of the most used markers are the brain T2 hyperin-
Different contemporary biomarkers could have a clinical tense lesion volume, brain T1 hypointense volume, new and
utility to distinguish SPMS from RRMS, such as magnetic enlarging active T1 and T2 lesion number and global and
resonance imaging (MRI) data or protein and metabo- global/regional brain atrophy, the magnetization transfer
lite measurement of cerebrospinal fluid (CSF) or blood. ratio in brain lesions or the spinal cord cross-sectional area
Research is still ongoing to identify those that can be used [72–75]. Between them, the brain atrophy and spinal cord
for early identification of the transition to SPMS reflecting area may be the most promising biomarkers for monitoring
its immunopathological characteristics [68]. A combined neurodegeneration, particularly in patients with SPMS who
approach with different biomarkers has also been discussed are characterized by less inflammation. They could be a very
that could aid the diagnosis and track disease progression, promising tool for distinguishing disease- and treatment-
such as MRI variables, proteins and metabolites [69]. A related brain volume changes and also the transition from
composite marker that included cognitive evaluation and RRMS to SPMS [18, 71, 76–78].
MRI measures was also accurate in discriminating between Advanced MRI techniques are also on investigation and
RRMS and SPMS [70]. may provide higher pathological specificity and be more
Notwithstanding, there are still no defined biomark- closely associated with clinical correlates, such as the
ers that allow to anticipate the conversion to SPMS. Here, magnetization transfer ratio, diffusion tensor imaging and

13
Journal of Neurology

magnetic resonance spectroscopy or fractional anisotropy Prognosis in patients with SPMS


[76, 79].
The study of the retina through the Optical Coherence MS is variable from person to person, so predicting
Tomography (OCT) is also promising in MS progression patients’ prognoses is challenging. Generally, patients
detection and neuroprotective strategies. The measurement with SPMS have an unfavourable prognosis and can
of retinal nerve fibre layer (RNFL) thickness and macular expect a slow and unremitting worsening of their abil-
volume have been included in clinical trials on progressive ity to function. Data concerning time to reach determined
MS and data suggest that RNFL thinning may be greater in disability scores are variable [94]. Based on data from a
SPMS and PPMS patients compared to those with RRMS, cohort of more than 800 patients assessed over a period of
especially within quadrants of peripapillary retina [80]. 28 years, patients with SPMS progressed to EDSS of 6.0,
However, patients with progressive and non-progressive 8.0 and 10.0 within about 5, 15 and 30 years, respectively,
forms of MS have thinner RNFL compared to controls of SPMS onset [5]. Greater frequency of early relapse,
independent of history of optic neuritis [80, 81]. Thus, brainstem and cerebellar presentation, and disease onset
further data are needed to demonstrate how these measures at age ≤ 20  years were associated with faster progres-
could predict or detect disease progression. sion to EDSS 8.0 [5]. In another cohort of 212 patients
with RRMS followed up to 50 years, the median time to
EDSS 6.0 was 22 years and to EDSS 7.0 was 37 years
Molecular markers [95]. Fifty years after onset, 14% of patients remained
progressive, 22% were progressive but ambulatory, 16%
Alterations on selected biochemical markers on patients were non-ambulatory and 48% had died. It has also been
with SPMS compared with RRMS have been reported but reported that 25% of patients will reach a EDSS of 8.0
there is no clear standardized use of them at the moment. within 16 years [19].
High levels of 14-3-3 protein, tau, neurofilaments, chi- Most SPMS patients will find a continued accumulative
tinase 3-like 1 and cystatin C correlated with disability worsening of disability compared to the relapsing–remit-
progression in MS patients [76]. Up to 19 metabolites ting phase of the disease [31, 43]. Physical functioning
from the tryptophan and phenylalanine metabolisms show will generally be significantly impaired [31], with a great
unique alterations to the SPMS phenotype and significant group no longer being able to walk 100 m unaided [43].
correlations to EDSS, disease duration and MRI measures Those patients who are still able to work are more likely to
[82]. need time off work than those with RRMS [31].
Coping with a diagnosis of SPMS is emotionally dif-
ficult for patients, but the prognostic uncertainty and dif-
Neuronal destruction markers ficulties in communication can be equally challenging for
clinicians, particularly as patients’ views and opinions on
Current interesting biomarkers are the neurofilaments, espe- how much information they want varies widely [16, 96].
cially neurofilaments light chain (NfL). Nowadays, they Some patients experience unsatisfactory discussions with
can be even detectable at very low blood concentrations, clinicians, particularly at the time of diagnosis, which can
proportional to CSF levels. This is of great use in several have long-lasting effects and expectations on future dis-
neurological diseases, as well as in MS, being a sensitive cussions. Positive adjustment will help patients to cope
(although non-specific) and clinically meaningful biomarker emotionally and psychologically with SPMS. This is often
to monitor tissue damage on MS [83–87]. Regular moni- realized through self-management and adapting strategies,
toring of serum NfL could demonstrate future or present such as accepting the limitations placed on their daily liv-
disease activity even when other clinical parameters seemed ing by the progressive nature of the disease and staying
to be stable [88]. Blood NfL levels show a good correlation focused on the present and on positive aspects of their
with brain atrophy detected by MRI measures and are sen- lives in spite of such a debilitating disease [16, 96].
sitive to detect disease activity on progressive forms of the Patients with SPMS are likely to eventually require pal-
disease [89, 90]. NfL levels could even have a prognostic liative care. Support should be provided by a multidiscipli-
value of disease worsening, with a direct correlation between nary team considering their physical and emotional needs,
serum NfL percentile level and future brain and cervical spiritual and social care [32, 97, 98]. A palliative approach
spinal volume loss [91, 92]. Even the risk of conversion from offers patients and their families help with symptoms and
RRMS to SPMS has been associated with CSF-NfL levels support in anticipating and planning for decisions that are
[93]. However, the use of NfL is currently mostly restricted likely to arise as the disease progresses, with the overall
to sub-specialized centres and further data must be generated goal of providing the best possible quality of life [97].
to support its use on the daily practice.

13
Journal of Neurology

Conclusions 8. Davies F, Wood F, Brain KE et al (2016) The transition to sec-


ondary progressive multiple sclerosis: an exploratory qualita-
tive study of health professionals’ experiences. Int J MS Care
The diagnosis of SPMS is challenging for the patient 18(5):257–264. https​://doi.org/10.7224/1537-2073.2015-062
and physician and is frequently done retrospectively and 9. Rocca MA, Mezzapesa DM, Falini A et al (2003) Evidence
delayed. A definition based on the EDSS evaluation seems for axonal pathology and adaptive cortical reorganization in
patients at presentation with clinically isolated syndromes sug-
to have good psychometric characteristics, but the inte- gestive of multiple sclerosis. NeuroImage 18(4):847–855
gration of other clinical outcome measures such as the 10. Mainero C, Caramia F, Pozzilli C et al (2004) fMRI evidence
T25FW and 9HPT, as seen with newer definitions such as of brain reorganization during attention and memory tasks in
PIRA or CDP, could provide additional important informa- multiple sclerosis. NeuroImage 21(3):858–867. https​: //doi.
org/10.1016/j.neuro​image​.2003.10.004
tion. A complete neurological history as well as the use of 11. Tracy JI, Hampstead BM, Sathian K (2015) Cognitive plasticity
new technologies could clearly support SPMS diagnosis. in neurologic disorders. Oxford University Press, Oxford https​
The early diagnosis of SPMS, as soon as in the transition ://doi.org/10.1093/med/97801​99965​243.001.0001
phase could open a window of opportunities for the treat- 12. Lopez-Gongora M, Escartin A, Martinez-Horta S et  al
(2015) Neurophysiological evidence of compensatory brain
ment in this phase of MS disease. mechanisms in early-stage multiple sclerosis. PLoS ONE
10(8):e0136786. https​://doi.org/10.1371/journ​al.pone.01367​86
Compliance with ethical standards  13. Sumowski JF, Rocca MA, Leavitt VM et al (2013) Brain reserve
and cognitive reserve in multiple sclerosis: what you’ve got
Conflicts of interest  HI declares that he has no conflict of interest. UP and how you use it. Neurology 80(24):2186–2193. https​://doi.
received personal compensation from Bayer, Biogen and Roche for the org/10.1212/WNL.0b013​e3182​96e98​b
consulting service. KA received personal compensation from Novartis, 14. Ziemssen T, Kern R, Thomas K (2016) Multiple sclerosis:
Biogen Idec, Teva, Sanofi and Roche for the consulting service. TZ re- clinical profiling and data collection as prerequisite for per-
ceived personal compensation from Biogen, Bayer, Celgene, Novartis, sonalized medicine approach. BMC Neurol 16:124. https​://doi.
Roche, Sanofi, Teva for the consulting services. Ziemssen received ad- org/10.1186/s1288​3-016-0639-7
ditional financial support for the research activities from Bayer, BAT; 15. Dutta R, Trapp BD (2014) Relapsing and progressive forms of
Biogen, Novartis, Teva, and Sanofi. multiple sclerosis: insights from pathology. Curr Opin Neurol
27(3):271–278. https​://doi.org/10.1097/wco.00000​00000​00009​
4
16. Davies F, Edwards A, Brain K et al (2015) ’You are just left to
get on with it’: qualitative study of patient and carer experiences
References of the transition to secondary progressive multiple sclerosis. BMJ
Open 5(7):e007674. https:​ //doi.org/10.1136/bmjope​ n-2015-00767​
4
1. Khurana V SH, Medin J, Adlard N (2018) Estimated preva-
17. Lublin FD, Reingold SC, Cohen JA et al (2014) Defining the clini-
lence of diagnosed secondary progressive multiple sclerosis in
cal course of multiple sclerosis: the 2013 revisions. Neurology
the Americas and Europe: results from a systematic literature
83(3):278–286. https​://doi.org/10.1212/wnl.00000​00000​00056​0
search. In: Paper presented at the poster presented at the Ameri-
18. Cree BAC, Hollenbach JA, Bove R et al (2019) Silent progression
can Academy of Neurology Annual Meeting, 21–27 April 2018,
in disease activity-free relapsing multiple sclerosis. Ann Neurol
Los Angeles, CA, USA.
85(5):653–666. https​://doi.org/10.1002/ana.25463​
2. Multiple Sclerosis International Federation (MSIF). (2013)
19. Tremlett H, Zhao Y, Devonshire V (2009) Natural history com-
Atlas of MS 2013. https​: //www.msif.org/wp-conte​n t/uploa​
parisons of primary and secondary progressive multiple sclerosis
ds/2014/09/Atlas​-of-MS.pdf. Accessed 24 Apr 2019
reveals differences and similarities. J Neurol 256(3):374–381.
3. English C, Aloi JJ (2015) New FDA-approved disease-modify-
https​://doi.org/10.1007/s0041​5-009-0039-7
ing therapies for multiple sclerosis. Clin Ther 37(4):691–715.
20. Koch M, Kingwell E, Rieckmann P, Tremlett H (2010) The natu-
https​://doi.org/10.1016/j.clint​hera.2015.03.001
ral history of secondary progressive multiple sclerosis. J Neurol
4. Coret F, Perez-Miralles FC, Gascon F et al (2018) Onset of
Neurosurg Psychiatry 81(9):1039–1043. https​://doi.org/10.1136/
secondary progressive multiple sclerosis is not influenced
jnnp.2010.20817​3
by current relapsing multiple sclerosis therapies. Mult Scler
21. Jokubaitis VG, Spelman T, Kalincik T et al (2015) Predictors of
J Exp Transl Clin 4(2):2055217318783347. https ​ : //doi.
disability worsening in clinically isolated syndrome. Ann Clin
org/10.1177/20552​17318​78334​7
Transl Neurol 2(5):479–491. https​://doi.org/10.1002/acn3.187
5. Scalfari A, Neuhaus A, Daumer M et al (2014) Onset of sec-
22. Kavaliunas A, Manouchehrinia A, Stawiarz L et  al (2017)
ondary progressive phase and long-term evolution of multiple
Importance of early treatment initiation in the clinical course of
sclerosis. J Neurol Neurosurg Psychiatry 85(1):67–75. https​://
multiple sclerosis. Mult Scler (Houndmills, Basingstoke, Engl)
doi.org/10.1136/jnnp-2012-30433​3
23(9):1233–1240. https​://doi.org/10.1177/13524​58516​67503​9
6. Cree BA, Gourraud PA, Oksenberg JR et al (2016) Long-term
23. Brown JWL, Coles A, Horakova D et al (2019) Association of
evolution of multiple sclerosis disability in the treatment era.
initial disease-modifying therapy with later conversion to second-
Ann Neurol 80(4):499–510. https​://doi.org/10.1002/ana.24747​
ary progressive multiple sclerosis. JAMA 321(2):175–187. https​
7. Tedeholm H, Lycke J, Skoog B et al (2013) Time to second-
://doi.org/10.1001/jama.2018.20588​
ary progression in patients with multiple sclerosis who were
24. Charcot JM (1865) Sclérose des cordons latéraux de la moelle
treated with first generation immunomodulating drugs. Mult
épinière chez une femme hystérique atteinte de contractures des
Scler (Houndmills, Basingstoke, Engl) 19(6):765–774. https​://
4 membres. L’Union Médicale, Paris
doi.org/10.1177/13524​58512​46376​4
25. Thompson AJ, Banwell BL, Barkhof F et al (2018) Diagnosis
of multiple sclerosis: 2017 revisions of the McDonald criteria.

13
Journal of Neurology

Lancet Neurol 17(2):162–173. https​://doi.org/10.1016/s1474​ 42. Giovannetti AM, Giordano A, Pietrolongo E et al (2017) Man-
-4422(17)30470​-2 aging the transition (ManTra): a resource for persons with sec-
26. Lucchinetti C, Bruck W, Parisi J et al (2000) Heterogeneity of ondary progressive multiple sclerosis and their health profes-
multiple sclerosis lesions: implications for the pathogenesis of sionals: protocol for a mixed-methods study in Italy. BMJ Open
demyelination. Ann Neurol 47(6):707–717 7(8):e017254. https​://doi.org/10.1136/bmjop​en-2017-01725​4
27. Lassmann H, Bruck W, Lucchinetti C (2001) Heterogeneity of 43. Lorscheider J, Buzzard K, Jokubaitis V et al (2016) Defining
multiple sclerosis pathogenesis: implications for diagnosis and secondary progressive multiple sclerosis. Brain J Neurol 139(Pt
therapy. Trends Mol Med 7(3):115–121 9):2395–2405. https​://doi.org/10.1093/brain​/aww17​3
28. O’Loughlin E, Hourihan S, Chataway J et al (2017) The expe- 44. Katz Sand I, Krieger S, Farrell C, Miller AE (2014) Diagnos-
rience of transitioning from relapsing remitting to secondary tic uncertainty during the transition to secondary progressive
progressive multiple sclerosis: views of patients and health multiple sclerosis. Mult Scler (Houndmills, Basingstoke, Engl)
professionals. Disabil Rehabil 39(18):1821–1828. https​://doi. 20(12):1654–1657. https​://doi.org/10.1177/13524​58514​52151​
org/10.1080/09638​288.2016.12117​60 7
29. Ziemssen T (2011) Symptom management in patients with mul- 45. Kremenchutzky M, Rice GP, Baskerville J et al (2006) The natu-
tiple sclerosis. J Neurol Sci 311(Suppl 1):S48–52. https​://doi. ral history of multiple sclerosis: a geographically based study 9:
org/10.1016/s0022​-510x(11)70009​-0 observations on the progressive phase of the disease. Brain J Neu-
30. PatientsLikeMe. (2016) The voice of patientslikeme multiple rol 129(Pt 3):584–594. https​://doi.org/10.1093/brain​/awh72​1
sclerosis patients: a brief report on patient perceptions of impor- 46. Bamer AM, Cetin K, Amtmann D et al (2007) Comparing a self
tant treatment outcomes. https​://news.patie​ntsli​keme.com/sites​/ report questionnaire with physician assessment for determining
patie​ntsli​keme.newsh​q.busin​esswi​re.com/files​/doc_libra​r y/file/ multiple sclerosis clinical disease course: a validation study. Mult
The_Voice​_of_Patie​ntsLi​keMe_Multi​ple_Scler​osis_Patie​nts.pdf Scler (Houndmills, Basingstoke, Engl) 13(8):1033–1037. https​://
Accessed 06 April 2019 doi.org/10.1177/13524​58507​07762​4
31. Gross HJ, Watson C (2017) Characteristics, burden of illness, 47. Tuohy O, Costelloe L, Hill-Cawthorne G et al (2015) Alemtu-
and physical functioning of patients with relapsing-remitting zumab treatment of multiple sclerosis: long-term safety and effi-
and secondary progressive multiple sclerosis: a cross-sectional cacy. J Neurol Neurosurg Psychiatry 86(2):208–215. https​://doi.
US survey. Neuropsychiatr Dis Treat 13:1349–1357. https​://doi. org/10.1136/jnnp-2014-30772​1
org/10.2147/ndt.S1320​79 48. Skoog B, Tedeholm H, Runmarker B et al (2014) Continuous
32. Sihra N, Gibson S, Bradley L (2017) Meeting the clinical needs prediction of secondary progression in the individual course of
of patients with progressive multiple sclerosis. Clin Med (Lond, multiple sclerosis. Mult Scler Relat Disord 3(5):584–592. https​://
Engl) 17(3):286. https​://doi.org/10.7861/clinm​edici​ne.17-3-286 doi.org/10.1016/j.msard​.2014.04.004
33. Costelloe L, O’Rourke K, Kearney H et al (2007) The patient 49. Manouchehrinia A, Zhu F, Piani-Meier D et al (2019) Predicting
knows best: significant change in the physical component of the risk of secondary progression in multiple sclerosis: a nomogram.
Multiple Sclerosis Impact Scale (MSIS-29 physical). J Neurol Mult Scler (Houndmills, Basingstoke, Engl) 25(8):1102–1112.
Neurosurg Psychiatry 78(8):841–844. https​://doi.org/10.1136/ https​://doi.org/10.1177/13524​58518​78366​7
jnnp.2006.10575​9 50. Kalincik T, Cutter G, Spelman T et  al (2015) Defining reli-
34. Bosma LV, Sonder JM, Kragt JJ et al (2015) Detecting clinically- able disability outcomes in multiple sclerosis. Brain J Neurol
relevant changes in progressive multiple sclerosis. Mult Scler 138(11):3287–3298. https​://doi.org/10.1093/brain​/awv25​8
(Houndmills, Basingstoke, Engl) 21(2):171–179. https​://doi. 51. Kappos L, Butzkueven H, Wiendl H et al (2018) Greater sensi-
org/10.1177/13524​58514​54096​9 tivity to multiple sclerosis disability worsening and progression
35. Goretti B, Portaccio E, Zipoli V et al (2010) Coping strategies, events using a roving versus a fixed reference value in a prospec-
cognitive impairment, psychological variables and their relation- tive cohort study. Mult Scler (Houndmills, Basingstoke, Engl)
ship with quality of life in multiple sclerosis. Neurol Sci 31(Suppl 24(7):963–973. https​://doi.org/10.1177/13524​58517​70961​9
2):S227–230. https​://doi.org/10.1007/s1007​2-010-0372-8 52. Whitaker JN, McFarland HF, Rudge P, Reingold SC (1995) Out-
36. Sullivan CLWJ, Rabin BM et al (2004) Psychosocial adjustment comes assessment in multiple sclerosis clinical trials: a critical
to multiple sclerosis. Int J MS Care 6:98–105 analysis. Mult Scler (Houndmills, Basingstoke, Engl) 1(1):37–47.
37. Ayache SS, Chalah MA (2017) Fatigue in multiple sclerosis- https​://doi.org/10.1177/13524​58595​00100​107
Insights into evaluation and management. Neurophysiologie cli- 53. Meyer-Moock S, Feng YS, Maeurer M et al (2014) Systematic lit-
nique Clin Neurophysiol 47(2):139–171. https:​ //doi.org/10.1016/j. erature review and validity evaluation of the Expanded Disability
neucl​i.2017.02.004 Status Scale (EDSS) and the Multiple Sclerosis Functional Com-
38. Powell DJH, Liossi C, Schlotz W, Moss-Morris R (2017) Track- posite (MSFC) in patients with multiple sclerosis. BMC Neurol
ing daily fatigue fluctuations in multiple sclerosis: ecological 14:58. https​://doi.org/10.1186/1471-2377-14-58
momentary assessment provides unique insights. J Behav Med 54. Bosma L, Kragt JJ, Polman CH, Uitdehaag BM (2013) Walking
40(5):772–783. https​://doi.org/10.1007/s1086​5-017-9840-4 speed, rather than Expanded Disability Status Scale, relates to
39. Kasser SL, Goldstein A, Wood PK, Sibold J (2017) Symptom long-term patient-reported impact in progressive MS. Mult Scler
variability, affect and physical activity in ambulatory persons (Houndmills, Basingstoke, Engl) 19(3):326–333. https​://doi.
with multiple sclerosis: understanding patterns and time-bound org/10.1177/13524​58512​45434​6
relationships. Disabil Health J 10(2):207–213. https ​ : //doi. 55. Cadavid D, Jurgensen S, Lee S (2013) Impact of natalizumab on
org/10.1016/j.dhjo.2016.10.006 ambulatory improvement in secondary progressive and disabled
40. Kratz AL, Murphy SL, Braley TJ (2017) Ecological momentary relapsing-remitting multiple sclerosis. PLoS ONE 8(1):e53297.
assessment of pain, fatigue, depressive, and cognitive symptoms https​://doi.org/10.1371/journ​al.pone.00532​97
reveals significant daily variability in multiple sclerosis. Arch 56. Galetta KM, Balcer LJ (2013) Measures of visual pathway struc-
Phys Med Rehabil 98(11):2142–2150. https​://doi.org/10.1016/j. ture and function in MS: Clinical usefulness and role for MS trials.
apmr.2017.07.002 Mult Scler Relat Disord 2(3):172–182. https​://doi.org/10.1016/j.
41. Coetzee T, Zaratin P, Gleason TL (2015) Overcoming barriers in msard​.2012.12.004
progressive multiple sclerosis research. Lancet Neurol 14(2):132– 57. Motl RW, Cohen JA, Benedict R et al (2017) Validity of the timed
133. https​://doi.org/10.1016/S1474​-4422(14)70323​-0 25-foot walk as an ambulatory performance outcome measure for

13
Journal of Neurology

multiple sclerosis. Mult Scler (Houndmills, Basingstoke, Engl) 74. Naismith RT, Cross AH (2005) Multiple sclerosis and black holes:
23(5):704–710. https​://doi.org/10.1177/13524​58517​69082​3 connecting the pixels. Arch Neurol 62(11):1666–1668. https:​ //doi.
58. Goldman MD, Motl RW, Scagnelli J et  al (2013) Clinically org/10.1001/archn​eur.62.11.1666
meaningful performance benchmarks in MS: timed 25-foot walk 75. Newcombe J, Hawkins CP, Henderson CL et al (1991) Histopa-
and the real world. Neurology 81(21):1856–1863. https​://doi. thology of multiple sclerosis lesions detected by magnetic reso-
org/10.1212/01.wnl.00004​36065​.97642​.d2 nance imaging in unfixed postmortem central nervous system tis-
59. Kaufman M, Moyer D, Norton J (2000) The significant change for sue. Brain J Neurol 114(Pt 2):1013–1023. https:​ //doi.org/10.1093/
the Timed 25-foot Walk in the multiple sclerosis functional com- brain​/114.2.1013
posite. Mult Scler (Houndmills, Basingstoke, Engl) 6(4):286–290. 76. Gajofatto A, Calabrese M, Benedetti MD, Monaco S (2013)
https​://doi.org/10.1177/13524​58500​00600​411 Clinical, MRI, and CSF markers of disability progression
60. Kragt JJ, van der Linden FA, Nielsen JM et al (2006) Clinical in multiple sclerosis. Dis Mark 35(6):687–699. https​: //doi.
impact of 20% worsening on Timed 25-foot Walk and 9-hole Peg org/10.1155/2013/48495​9
Test in multiple sclerosis. Mult Scler (Houndmills, Basingstoke, 77. Inglese M, Oesingmann N, Casaccia P, Fleysher L (2011) Pro-
Engl) 12(5):594–598. https​://doi.org/10.1177/13524​58506​07076​ gressive multiple sclerosis and gray matter pathology: an MRI
8 perspective. Mt Sinai J Med NY 78(2):258–267. https​://doi.
61. Cadavid D, Cohen JA, Freedman MS et al (2017) The EDSS-Plus, org/10.1002/msj.20247​
an improved endpoint for disability progression in secondary pro- 78. Masek M, Vaneckova M, Krasensky J et al (2008) Secondary-
gressive multiple sclerosis. Mult Scler (Houndmills, Basingstoke, progressive form of multiple sclerosis: MRI changes versus clini-
Engl) 23(1):94–105. https​://doi.org/10.1177/13524​58516​63894​1 cal status. Neuro Endocrinol Lett 29(4):461–466
62. Kappos L WJ, Giovannoni G et al. (2017) Ocrelizumab reduces 79. Moccia M, de Stefano N, Barkhof F (2017) Imaging outcome
disability progression independent of relapse activity in patients measures for progressive multiple sclerosis trials. Multiple Scler
with relapsing multiple sclerosis. In: Paper presented at the The (Houndmills, Basingstoke, England) 23(12):1614–1626. https​://
7th Joint ECTRIMS–ACTRIMS Meeting. 25–28 October 2017, doi.org/10.1177/13524​58517​72945​6
Paris, France 80. Pulicken M, Gordon-Lipkin E, Balcer LJ et al (2007) Optical
63. Lu G, Beadnall HN, Barton J et  al (2018) The evolution of coherence tomography and disease subtype in multiple sclero-
“No Evidence of Disease Activity” in multiple sclerosis. Mult sis. Neurology 69(22):2085–2092. https​://doi.org/10.1212/01.
Scler Relat Disord 20:231–238. https​://doi.org/10.1016/j.msard​ wnl.00002​94876​.49861​.dc
.2017.12.016 81. Winges KM, Murchison CF, Bourdette DN, Spain RI (2019) Lon-
64. L Kappos (2019) A novel functional composite endpoint to char- gitudinal optical coherence tomography study of optic atrophy in
acterize disease progression in patients with secondary progres- secondary progressive multiple sclerosis: Results from a clini-
sive multiple sclerosis. In: Paper presented at the AAN 2019: Pre cal trial cohort. Multiple Scler (Houndmills, Basingstoke, Engl)
Congress Webcast. 25(1):55–62. https​://doi.org/10.1177/13524​58517​73913​6
65. Ha JF, Longnecker N (2010) Doctor-patient communication: a 82. Herman S, Akerfeldt T, Spjuth O et al (2019) Biochemical differ-
review. Ochsner J 10(1):38–43 ences in cerebrospinal fluid between secondary progressive and
66. Ziemssen T, Simsek D, Lahoz R, Verdun di Cantogno E relapsing(-)remitting multiple sclerosis. Cells 8(2):84. https​://doi.
(2015) Development of a screening tool to support identifica- org/10.3390/cells​80200​84
tion of patients with secondary progressive multiple sclerosis 83. Cai L, Huang J (2018) Neurofilament light chain as a biological
(Spms). Value Health 18(7):A763. https​://doi.org/10.1016/j. marker for multiple sclerosis: a meta-analysis study. Neuropsychi-
jval.2015.09.2497 atr Dis Treat 14:2241–2254. https​://doi.org/10.2147/ndt.S1732​80
67. https​://mspro​discu​ss.com/ 84. Disanto G, Barro C, Benkert P et al (2017) Serum Neurofilament
68. Iwanowski P, Losy J (2015) Immunological differences between light: A biomarker of neuronal damage in multiple sclerosis. Ann
classical phenothypes of multiple sclerosis. J Neurol Sci 349(1– Neurol 81(6):857–870. https​://doi.org/10.1002/ana.24954​
2):10–14. https​://doi.org/10.1016/j.jns.2014.12.035 85. Hakansson I, Tisell A, Cassel P et al (2017) Neurofilament light
69. Herman S, Khoonsari PE, Tolf A et al (2018) Integration of mag- chain in cerebrospinal fluid and prediction of disease activity in
netic resonance imaging and protein and metabolite CSF measure- clinically isolated syndrome and relapsing-remitting multiple
ments to enable early diagnosis of secondary progressive multiple sclerosis. Eur J Neurol 24(5):703–712. https​://doi.org/10.1111/
sclerosis. Theranostics 8(16):4477–4490. https:​ //doi.org/10.7150/ ene.13274​
thno.26249​ 86. Malmestrom C, Haghighi S, Rosengren L et al (2003) Neuro-
70. Kizlaitiene R, Kaubrys G, Giedraitiene N et al (2017) Composite filament light protein and glial fibrillary acidic protein as bio-
marker of cognitive dysfunction and brain atrophy is highly accu- logical markers in MS. Neurology 61(12):1720–1725. https​://doi.
rate in discriminating between relapsing-remitting and secondary org/10.1212/01.wnl.00000​98880​.19793​.b6
progressive multiple sclerosis. Med Sci Monit 23:588–597. https​ 87. Teunissen CE, Khalil M (2012) Neurofilaments as biomarkers in
://doi.org/10.12659​/msm.90323​4 multiple sclerosis. Multiple Scl (Houndmills, Basingstoke, Engl)
71. Barkhof F, Calabresi PA, Miller DH, Reingold SC (2009) Imaging 18(5):552–556. https​://doi.org/10.1177/13524​58512​44309​2
outcomes for neuroprotection and repair in multiple sclerosis tri- 88. Akgün K, Kretschmann N, Haase R et al (2019) Profiling indi-
als. Nat Rev Neurol 5(5):256–266. https​://doi.org/10.1038/nrneu​ vidual clinical responses by high-frequency serum neurofilament
rol.2009.41 assessment in MS. Neurol: Neuroimmunol Neuroinflammation
72. Filippi M, Agosta F (2007) Magnetization transfer MRI in mul- 6(3):e555. https​://doi.org/10.1212/NXI.00000​00000​00055​5
tiple sclerosis. J Neuroimaging 17(Suppl 1):22s–26s. https​://doi. 89. Silber E, Semra YK, Gregson NA, Sharief MK (2002) Patients
org/10.1111/j.1552-6569.2007.00132​.x with progressive multiple sclerosis have elevated antibodies to
73. Miller DH, Barkhof F, Frank JA et al (2002) Measurement of neurofilament subunit. Neurology 58(9):1372–1381. https​://doi.
atrophy in multiple sclerosis: pathological basis, methodological org/10.1212/wnl.58.9.1372
aspects and clinical relevance. Brain J Neurol 125(Pt 8):1676– 90. Gnanapavan S, Grant D, Morant S et al (2013) Biomarker report
1695. https​://doi.org/10.1093/brain​/awf17​7 from the phase II lamotrigine trial in secondary progressive MS-
neurofilament as a surrogate of disease progression. PLoS ONE
8(8):e70019. https​://doi.org/10.1371/journ​al.pone.00700​19

13
Journal of Neurology

91. Salzer J, Svenningsson A, Sundstrom P (2010) Neurofilament of predictors from onset. J Neurol 262(5):1148–1163. https​://doi.
light as a prognostic marker in multiple sclerosis. Multiple Scler org/10.1007/s0041​5-015-7674-y
(Houndmills, Basingstoke, Engl) 16(3):287–292. https​://doi. 96. Dennison L, McCloy Smith E, Bradbury K, Galea I (2016) How
org/10.1177/13524​58509​35972​5 do people with multiple sclerosis experience prognostic uncer-
92. Barro C, Benkert P, Disanto G et al (2018) Serum neurofilament tainty and prognosis communication? A qualitative study. PLoS
as a predictor of disease worsening and brain and spinal cord ONE 11(7):e0158982. https:​ //doi.org/10.1371/journa​ l.pone.01589​
atrophy in multiple sclerosis. Brain J Neurol 141(8):2382–2391. 82
https​://doi.org/10.1093/brain​/awy15​4 9 7. Oliver DJ, Borasio GD, Caraceni A et al (2016) A consensus
93. Bhan A, Jacobsen C, Myhr KM et  al (2018) Neurofilaments review on the development of palliative care for patients with
and 10-year follow-up in multiple sclerosis. Multiple Scler chronic and progressive neurological disease. Eur J Neurol
(Houndmills, Basingstoke, Engl) 24(10):1301–1307. https​://doi. 23(1):30–38. https​://doi.org/10.1111/ene.12889​
org/10.1177/13524​58518​78200​5 98. Kaur D, Kumar G, Billore N, Singh AK (2016) Defining the role
94. Vukusic S, Confavreux C (2003) Prognostic factors for progres- of physiotherapy in palliative care in multiple sclerosis. Indian
sion of disability in the secondary progressive phase of multiple J Palliat Care 22(2):176–179. https​://doi.org/10.4103/0973-
sclerosis. J Neurol Sci 206(2):135–137 1075.17959​9
95. Tedeholm H, Skoog B, Lisovskaja V et al (2015) The outcome
spectrum of multiple sclerosis: disability, mortality, and a cluster

13

You might also like