Next Article in Journal
Olfactory Stimulation Effect of Aldehydes, Nonanal, and Decanal on the Human Electroencephalographic Activity, According to Nostril Variation
Previous Article in Journal
A Mini-Review: Clinical Development and Potential of Aptamers for Thrombotic Events Treatment and Monitoring
Previous Article in Special Issue
Neuromyelitis Optica Spectrum Disorder and Anti-MOG Syndromes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Diagnosis and Management of Progressive Multiple Sclerosis

Mellen Center for Multiple Sclerosis, Neurological Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
*
Author to whom correspondence should be addressed.
Biomedicines 2019, 7(3), 56; https://doi.org/10.3390/biomedicines7030056
Submission received: 8 July 2019 / Revised: 23 July 2019 / Accepted: 26 July 2019 / Published: 29 July 2019
(This article belongs to the Special Issue Multiple Sclerosis: Diagnosis and Treatment)

Abstract

:
Multiple sclerosis is a chronic autoimmune disease of the central nervous system that results in varying degrees of disability. Progressive multiple sclerosis, characterized by a steady increase in neurological disability independently of relapses, can occur from onset (primary progressive) or after a relapsing–remitting course (secondary progressive). As opposed to active inflammation seen in the relapsing–remitting phases of the disease, the gradual worsening of disability in progressive multiple sclerosis results from complex immune mechanisms and neurodegeneration. A few anti-inflammatory disease-modifying therapies with a modest but significant effect on measures of disease progression have been approved for the treatment of progressive multiple sclerosis. The treatment effect of anti-inflammatory agents is particularly observed in the subgroup of patients with younger age and evidence of disease activity. For this reason, a significant effort is underway to develop molecules with the potential to induce myelin repair or halt the degenerative process. Appropriate trial methodology and the development of clinically meaningful disability outcome measures along with imaging and biological biomarkers of progression have a significant impact on the ability to measure the efficacy of potential medications that may reverse disease progression. In this issue, we will review current evidence on the physiopathology, diagnosis, measurement of disability, and treatment of progressive multiple sclerosis.

Graphical Abstract

1. Introduction

Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system that affects over 2.3 million people globally, with an estimated prevalence of approximately 310 per 100,000 population in the United States [1,2]. Most patients (~90%) have relapsing–remitting disease at onset, which typically is followed by a secondary progressive course, while a minority of patients have a primary progressive course from onset (~10%). Relapsing–remitting MS (RRMS) is characterized by frequent formation of inflammatory lesions in the brain and spinal cord. Approved disease-modifying therapies (DMTs) target the inflammatory component of the disease, and strong evidence support their effectiveness in RRMS. However, trials evaluating their efficacy in slowing disease progression have shown mixed results, or have shown only modest effects in slowing progression. The goal of this review is to provide a comprehensive overview on the current knowledge of the pathogenesis, diagnosis, and treatment of progressive MS, as well as future directions in the field.

2. Pathogenesis

The pathogenesis of MS is incompletely elucidated. This is particularly the case for progressive MS, for which various and sometimes conflicting data have been proposed to explain the underlying pathogenic process of progression [3]. In RRMS, actively demyelinating plaques are the most prominent lesion type, and are characterized by inflammatory demyelination and axonal transection within the lesions [4,5,6]. However, active lesions are rare in progressive MS, and axonal transection is not seen as frequently within inactive lesions compared to highly inflammatory recently developed lesions [3,4,5]. Whole brain atrophy, smoldering and enlarging lesions, cortical demyelination (specifically subpial lesions), and diffuse axonal injury and microglial activation in normal appearing grey and white matter are prominent in patients with progressive MS compared to patients with early RRMS [6,7,8,9,10]. Disability in progressive MS is thought to be related to secondary neurodegeneration of chronically demyelinating axons, which is thought to be driven by a series of factors, including: (1) inflammation and lesion accumulation, with subsequent retrograde and anterograde degeneration, (2) mitochondrial damage and subsequently virtual hypoxia and oxidative stress, (3) iron accumulation in myelin sheath and oligodendrocytes with subsequent amplification of oxidative stress, (4) lymphoid follicle-like structures that might contribute to sustaining cortical pathology, and [5] age-related neurodegeneration and reduced neuronal reserve (loss of the ability to compensate for axonal loss) [3,11,12,13,14,15]. A recent paper by Brown et al. [16], among others, showed that the early use of DMTs, specifically highly-effective DMTs, decreases the odds of conversion to secondary progressive MS (SPMS), which supports the role of early disease activity in the development of long-term disability progression [16,17,18,19,20,21]. The role of age-related mechanisms is supported by the fact that children with MS rarely present with progressive disease and have a longer time to reach secondary progression and disability milestones compared to adult-onset MS, and that certain disability milestones are acquired at certain ages independently of the duration of the disease [22,23]. However, for primary progressive MS (PPMS), the time course of irreversible damage is not clearly affected by the presence or absence of superimposed relapses [24]. Recent observations support a change in the natural history of MS with earlier use of highly-effective DMTs; however, this effect seems to be more clearly evidenced in RRMS [25]. Despite the wide variability of clinical and radiological presentations and the inherent pathological differences between RRMS, SPMS, and PPMS, the consensus is that PPMS is biologically part of the MS spectrum [13].

3. Diagnostic Criteria and Disease Course Definitions

The diagnosis of progressive MS is based on patient-reported clinical history, and should be confirmed based on objective physical examination findings. Based on the 2017 McDonald diagnostic criteria, PPMS can be diagnosed in patients with a 1-year history of disability progression, which can be retrospectively or prospectively determined, independent of clinical relapses, plus two of the following criteria: (1) One or more T2 lesions characteristic of MS in one or more typical brain regions (periventricular, cortical or juxtacortical, infratentorial); (2) two or more T2 lesions in the spinal cord, and (3) the presence of CSF-specific oligoclonal bands. Unlike the 2010 McDonald criteria, both symptomatic and asymptomatic MRI lesions are taken into account [26,27]. The panel also recommends specifying a provisional disease course at the time of diagnosis, and whether disease activity and/or progression are present or not based on the previous year’s history, which can then be revisited based on periodic re-evaluation [27]. Providing a clinical definition of disease progression, however, is somewhat harder. Progression is characterized by a steady increase in neurological disability occurring independently of relapses [27,28]. Symptoms can fluctuate (i.e., pseudo relapses), and bona fide superimposed relapses might occur. Detailed history taking is key in differentiating events suggestive of disease activity from worsening of previously experienced symptoms in the context of fatigue, heat, or stress. PPMS is defined by a progressive course from onset and SPMS by a progressive course following an initial relapsing–remitting course. The 2013 revisions of MS clinical course definitions aimed at standardizing the terminology across clinicians and researchers, for prognostication, design of clinical trials, and treatment decisions purposes [28]. These definitions included the presence or absence of clinical or radiological activity, and the presence or absence of disability progression into the phenotypic description of the disease. Figure 1 illustrates the currently used description of different progressive MS phenotypes. The distinction between “active” and “inactive” progressive disease, whether primary or secondary, has important therapeutic implications. This will be discussed in the treatment section of this manuscript.

4. Disability Outcome Measures

Disability progression in MS affects multiple functional domains, occurs insidiously over time, and can be difficult to quantify in an objective, comprehensive, and reproducible manner. Reliable detection of clinical and sub-clinical progression is key to interpreting treatment efficacy in trials and in clinical practice of DMT and repair promoting strategies in progressive MS. The expanded disability status scale (EDSS) is the most commonly used clinical outcome measure in trials for quantification of physical disability in MS. However, the EDSS does not comprehensively reflect disability status, and is particularly restricted in assessing cognitive and upper extremity functions [29]. Other limitations of the EDSS include poor intra and inter-rater variability especially for lower scores. The test also shows little sensitivity to detect change, especially in patients with scores of 6.0 or more, and the EDSS is difficult to administer in routine care [30,31,32]. Since it is an ordinal scale, changes in EDSS scores are not equivalent across the range of the scale. Finally, the most commonly used outcome measure in progressive MS trials is the 3 or 6-month confirmed disability progression, which might be insufficient to predict long-term disability worsening [33]. Evaluating multiple functional domains improves the likelihood of observing a change in patients with MS. The multiple sclerosis functional composite (MSFC) [34,35] was developed as a quantitative clinical measure of neurologic disability to overcome some of the shortcomings of the EDSS. Cognitive function was evaluated originally using the paced auditory serial addition test (PASAT), but more recently using the symbol–digit modalities test (SDMT), and upper extremity function is evaluated using the 9-hole peg test (9HPT) [36,37]. Walking speed is usually evaluated using the timed-25-foot walk test (T25FW). The MSFC has proven to be more sensitive to change than the EDSS, and correlates with subsequent changes in EDSS [34] T1 and T2 lesion load on brain MRI [38], and patient reported physical and emotional functioning and quality of life [39]. In the interferon beta (IFN-β)-1a SPMS trial, there was some benefit of treatment on the MSFC z-scores but not on the EDSS [40]. This benefit was mainly driven by two of the components of the MSFC, the 9HPT and the PASAT, which further illustrates the importance of a comprehensive neurological evaluation to assess disability progression. The low contrast letter acuity testing using a low-contrast Sloan letter chart was later proposed as an additional component of the MSFC to capture visual dysfunction with a high sensitivity, and also correlates with other components of the MSFC and the EDSS [37,41,42]. In addition, internal consistency (reliability) is higher for the MSFC (Cronbach’s alpha coefficient 0.93−0.96) than for the EDSS (Cronbach’s alpha 0.7) [43,44]. Although the MSFC has been used as an outcome measure in clinical trials [40,45,46], administration in routine clinical practice is time consuming, and requires personnel to administer the test properly [47]. Recently, there has been increasing interest in developing tools to facilitate and standardize testing in MS patients. For example, a technology-enabled version of the MSFC has been developed and was incorporated into routine clinical practice. This tool, called the multiple sclerosis performance test (MSPT), comprises a battery of quantitative neuroperformance assessments administered using a suite of iPad® applications modeled after the MSFC approach, and has allowed the gathering of large-scale comprehensive and standardized measures of disability from routine care [48,49]. A limitation of these outcome measures is the uncertainty of what constitutes a clinically meaningful change, although a threshold of ± 20% change in T25FW [50,51] ±15–20% in the 9HPT [52,53] ±10% (or 4 points) in the SDMT [53,54] and ±7 letters in the low contrast letter acuity test [55] have been suggested as clinically meaningful in MS.

5. Measuring Disease Progression

Numerous imaging biomarkers that have been proposed for the monitoring of progressive disease in MS trials, whole brain atrophy being the most widely used. Monitoring of T1 hypointensity evolution over time on conventional imaging has also been suggested. Other more advanced MRI techniques that can reflect axonal loss in progressive MS include thalamic volume, spinal cord atrophy, hippocampal volume, gray matter fraction, cortical lesion quantification, and sodium imaging, among others [56,57]. Several additional advanced MRI measures have been used as exploratory outcomes [58] as well as neurophysiological measures, such as evoked potentials [59]. Optical coherence tomography (OCT) is a non-invasive tool that allows the measurements of retinal nerve fiber layer thickness, ganglion cell/inner plexiform layer thickness, and macular volume. Since these measurements correlate with whole brain and gray matter atrophy and physical disability, OCT can serve as an outcome measure of axonal loss in phase 2 proof of concept clinical trials of progressive MS [60]. Other imaging and non-imaging techniques are used in remyelination trials as biomarkers of myelin repair, and include magnetization transfer ratio, diffusion-weighted imaging, myelin water imaging, and visual evoked potentials [56].
There has been increasing interest in the use of serum neurofilament light chain (NfL) as a biomarker in MS over the past few years [61]. Serum NfL levels correlate with CSF NfL levels and reflect disease activity and response to therapy [61]. In progressive MS, NfL levels appear to be associated with superimposed clinical or radiological activity, as well as T1-hypointense lesion volume [62]. Recent data suggests that CSF NfL correlates with disease activity rather than progression, irrespective of the disease subtype, and does not reflect disease severity [63,64], whereas previous data report a strong correlation between NfL levels and future worsening on the EDSS and brain and cervical spinal volume loss [65,66]. Serum NfL levels can be easily obtained, and are being further investigated and used as an outcome measure in newer progressive MS trials.

6. Treatment

6.1. Anti-Inflammatory Disease-Modifying Therapies

6.1.1. Approved Therapies

Ocrelizumab (Ocrevus®) a humanized monoclonal antibody targeting the CD20 antigen on B-cells. Ocrelizumab exerts its anti-inflammatory effects by causing rapid and profound depletion of B cells. Ocrelizumab has been studied in patients with RRMS in two phase 3 double-blind, placebo-controlled randomized clinical trial (OPERA I and II) [67] and in patients with PPMS (ORATORIO trial) [68], but not in patients with SPMS. Participants with PPMS were required to have positive oligoclonal bands to enroll in the study. In the ORATORIO trial ocrelizumab significantly reduced the risk of 24-week confirmed disability progression compared to placebo (29.6% with ocrelizumab versus 35.7% with placebo). Treatment with ocrelizumab in PPMS also decreased worsening on the timed-25-foot walk, T2 lesion volume, and brain atrophy at 120 weeks compared to placebo. In a pre-specified subgroup analysis, the magnitude of the effect of ocrelizumab was larger in patients with baseline enhancing lesions and younger patients; however, older patients without enhancing lesions at baseline also derived benefit across primary and secondary endpoints [68]. The ENCORE study evaluated the effect of ocrelizumab on upper limb function in the ORATORIO cohort, and also showed positive results (reduction in time—to 12- and 24-week confirmed ≥15% increase on 9HPT by 37% (hazard ratio 0.627; p = 0.001) and 39% (HR = 0.607; p = 0.002) for both-hands) [69]. Ocrelizumab was approved by the FDA and the EMA to treat PPMS in 2017. Ongoing trials aim at evaluating the effect of ocrelizumab on hand function in patients with more advanced disability (ORATORIO-HAND, NCT03562975), and in a broader range of patients (with PPMS and SPMS, up to 65 years old) (CONSONNANCE, NCT03523858). It is important to note that previous trials using the chimeric monoclonal antibody rituximab, which has a similar mechanism of action as ocrelizumab, yielded results in one pivotal trial that shaped the inclusion criteria for the ORATORIO study. In the OLYMPUS trial of rituximab, the primary endpoint was negative but sub-group analysis found that younger patients with clinical or radiological evidence of disease activity did derive treatment benefit. The subgroup of patients who were older and did not have gadolinium-enhancing lesions had faster disability progression than those on placebo [70]. This indicates a potential harm of treating with a B-cell therapy in this population. Another recent observational study using retrospective data from three European centers and propensity score matching, analyzed the effect of rituximab treatment on disability progression in patients with SPMS. In this study, patients with SPMS treated with rituximab had significantly delayed confirmed progression compared with matched untreated controls at up to 10 years [71]. The difference in effects seen between ocrelizumab and rituximab are most likely related to trial design; however, biological differences may exist as well, given that ocrelizumab appears to promote cell death via greater antibody-dependent cellular cytotoxicity (ADCC) activity and less complement-dependent cytotoxicity (CDC) activity compared to rituximab, and has a more favorable antigenic profile compared to rituximab [72,73,74].
Siponimod (Mayzent®) is a selective sphingosine-1-phosphate receptor 1 and 5 modulator, which inhibits the egress of lymphocytes from lymph nodes, thus decreasing their entry into the CNS. In addition to its anti-inflammatory effects, siponimod has been found to have putative neuroprotective and repair properties in preclinical studies. It was recently approved for the treatment of SPMS based on the results of the EXPAND trial [75]. Compared to the placebo group, a significant reduction in time to 3 and 6-month confirmed disability progression (relative risk reductions of 21% and 26%, respectively) was observed in the siponimod group, and this trend was consistent in subgroup analysis with respect to pre-treatment relapse activity, disease progression rate, and disease severity. Significant reduction in markers of disease activity were also observed in the siponimod group, including annualized relapse rate, time to relapse, and gadolinium-enhancing lesions, and new/enlarging T2 lesions. Brain volume change from baseline was lower in the siponimod group at both months 12 and 24 compared to placebo [75]. Pre-clinical data also suggest that sphingosine-1-phosphate modulators cross the blood–brain barrier, and have the potential to improve morphological markers of remyelination [76]. In addition, modulation of sphingosine-1-phosphate receptor 5 has been shown to promote remyelination in vitro [76]. Interestingly, fingolimod, a sphingosine-1-phosphate receptor 1 to 5 modulator, failed to show significant reduction in confirmed disability worsening in patients with PPMS in the INFORMS trial [77]. The fact that other DMTs with a similar mechanism of action to siponimod and ocrelizumab failed to show benefit in PMS can be due to different patient population and sub-optimal trial designs, but also illustrates the lack of efficacy of anti-inflammatory strategies in the prevention of disability worsening in inactive PMS, and the need to develop molecules with a potential effect on neurodegeneration.
Mitoxantrone is a DNA intercalating agent that interferes with the replication and proliferation of B and T lymphocytes. Its use is nowadays limited due to the well-known serious adverse events (including cardio-toxicity, leukemia, amenorrhea, infections, alopecia, leucopenia, anemia, and hepatotoxicity) [78], and the availability of safer DMTs. The mitoxantrone in progressive multiple sclerosis (MIMS) trial was a double-blind, multicenter, phase 3 trial that randomized patients with worsening RRMS or SPMS to placebo or low (5 mg/m2) or high (12 mg/m2) dose mitoxantrone for 2 years [79]. About half of the participants had SPMS, with or without clinical activity in the year prior to enrollment. The primary outcome was a combination of five clinical measures: change from baseline EDSS at 24 months, change from baseline ambulation index at 24 months, number of relapses treated with corticosteroids, time to first treated relapse, and change from baseline standardized neurological status at 24 months [79]. In the cohort as a whole, a beneficial effect on the primary outcome clinical composite measure was observed for the mitoxantrone, with comparable treatment effects in patients with and without relapses in the year prior to enrollment. A few years later, the MIMS trial group analyzed the effect of low and high-dose mitoxantrone on measures of radiological activity in a subgroup of patients with worsening RRMS or SPMS, and surprisingly did not show a consistent effect of mitoxantrone on the presence of gadolinium-enhancing lesions for up to 24 months compared to placebo [80]. Mitoxantrone is approved by the FDA for the treatment of RRMS, SPMS, and what was previously referred to as “progressive relapsing MS”. There is no evidence that supports a benefit of mitoxantrone in PPMS without clinical or radiological activity [81,82]. Single-nucleotide polymorphism in the ATP-binding cassette transporter genes may serve as pharmacogenetic markers associated with clinical response to mitoxantrone in RRMS and SPMS [83]; however, this association was not observed in patients with PPMS [81].
Cladibine produces rapid and long-lasting reductions in T-lymphocytes and rapid but transient reduction in B-lymphocytes, by disrupting cellular metabolism, inhibiting DNA synthesis and repair, and subsequent apoptosis of affected cells. The oral formulation of cladribine (Mavenclad®) has recently received FDA approval for the treatment of active RRMS and SPMS based on the results of the CLARITY and ORACLE MS trials, and post-hoc analysis of the ONWARD trial [84,85,86,87]. Intravenous formulations of cladribine have been mainly studied in progressive MS. An initial small trial (24 matched patients with clinically definite progressive MS as defined at the time of the study, baseline EDSS of 4.7, randomized to receive IV cladribine or placebo) showed a significant benefit of cladribine on EDSS worsening, with some patients even experiencing improvement on EDSS at month 12 [88]. There was also a positive effect on T2 lesion volume in this study. Another larger trial was then conducted in light of these results. Patients with SPMS or PPMS and a median baseline EDSS score of 6.0 were randomly assigned to receive either placebo or cladribine 0.07 mg/kg/day for 5 consecutive days every 4 weeks for 2 or 6 cycles, followed by placebo, for 8 cycles [89]. No benefit of cladribine on the primary outcome (mean change in EDSS at month 12) was observed compared to placebo. As expected, there was a significant effect of cladribine on gadolinium-enhancing lesions and T2 lesion accumulation, and a somewhat marginal benefit in a subgroup analysis of SPMS patients. Similarly, in another study, cladribine did not have a beneficial effect on whole brain volumes compared to placebo [90].
In the ONWARD trial, the effect of oral cladribine as an add-on to IFN-β in patients with active RRMS or SPMS was studied [87]. As expected, cladribine + IFN-β was superior to placebo + IFN-β in reducing annualized relapse rate and gadolinium-enhancing lesions. However, the confirmed EDSS progression over 96 weeks was similar between the two groups. In a post-hoc analysis of subgroups in the intention-to-treat population, cladribine + IFN-β was superior to placebo + IFN-β in reduction of annualized relapse rate (relative risk ratio of 0.11, 95% CI 0.01–0.94) in patients with active SPMS. Hence, oral cladribine was approved by the FDA for the treatment of active SPMS, but not for progressive MS without evidence of clinical or radiological activity.

6.1.2. Therapies with Negative or Weak Effect in Progressive MS

Studies of other anti-inflammatory DMTs in progressive MS have yielded deceiving results. Interferon-beta (IFN-β) has complex immunomodulatory effects (downregulation of pro-inflammatory and upregulation of anti-inflammatory cytokines). There is no robust evidence of the beneficial effect of IFN-β on progression, although early treatment in RRMS decreases conversion to SPMS, which is attributable to its anti-inflammatory properties [16,17,18,19]. In SPMS, IFN-β significantly delayed time to confirmed disability progression compared to those receiving placebo in the European SPMS IFN-β-1b trial [91]. However, discrepant results were observed in the North American SPMS IFN-β-1b trial, which did not show a difference in time to 6-month sustained EDSS progression compared to placebo [92]. A post hoc pooled analysis of the clinical trial data of both groups was performed to better clarify this discrepancy, and showed that patients in the European studies who benefited from treatment were significantly younger (41 vs. 46.9 years, p < 0.001), with shorter disease duration (13.1 vs. 14.7 years, p < 0.001), and more active disease (number of relapses in the last 2 years 1.7 vs. 0.8, contrast-enhancing lesions 2.6 vs. 1.5, p < 0.001) [93]. The lack of benefit of IFN-β-1b in patients with SPMS with less active disease was confirmed in other SPMS trials as well [94,95].
IFN-β trials in PPMS have also been largely mixed, with negative results on primary (confirmed disability progression) and most secondary endpoints [96,97,98], although significant differences in MSFC scores, MRI T2 lesion volume, and MRI T1 lesion volume after 2 years of treatment favoring IFN-β-1a was observed in one trial [98].
Glatiramer acetate (GA), a synthetic polypeptide with a complex and incompletely understood immunomodulatory mechanism of action, was studied in patients with PPMS in the PROMiSe trial, a multicenter, placebo-controlled, double-blind randomized clinical trial comparing GA to placebo over a 3-year period [99]. This trial was successful in including a large majority of patients without signs of disease activity. GA decreased markers of radiological activity (gadolinium enhancing lesions and accumulation of T2 lesions), and had some benefit on disability progression in males, but there was no effect on the primary outcome and the study was terminated early [99,100].
As discussed earlier in this manuscript, the INFORMS trial failed to show a positive effect of fingolimod (Gilenya®) on reduction in confirmed disability worsening in patients with PPMS [77]. The primary endpoint was defined by a composite of outcomes including the EDSS, T25FW, and 9HPT. There was no benefit of fingolimod on the disability composite (HR = 0.95, 95%FW CI 0.80–1.10, p = 0.544). Fingolimod has not been studied in SPMS.
Natalizumab (Tysabri®) is a monoclonal antibody that exerts its potent anti-inflammatory effect by inhibiting the alpha-4 integrin and subsequently preventing the migration of T-lymphocytes across the blood–brain barrier. The ASCEND trial evaluated the effect of natalizumab on a composite score including the EDSS, T25FW, and 9HPT in patients with SPMS [101]. No benefit was observed on the composite primary outcome, and individually on the EDSS and 9HPT, although in a post hoc analysis, there was a 44% reduction in hand function progression measured by the 9HPT (OR 0.56, 95% CI 0.40–0.80, p = 0.001).
Rituximab is a chimeric monoclonal antibody targeting the CD20 antigen on pre-B-cells and mature B-cells that has been used in many autoimmune neurological disorders of the central and peripheral nervous systems for decades [102]. Similarly to ocrelizumab, it causes rapid and profound depletion of B-cells via antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) mechanisms leading to B-cell death [73,74]. Rituximab has been used off-label to treat progressive MS in certain countries, and there has been long-standing evidence of its efficacy to control inflammatory disease activity in observational studies [103]. Furthermore, as discussed above, a recent study also suggests that rituximab significantly delayed confirmed progression in SPMS [71]. The OLYMPUS trial evaluated the effect of rituximab on disability progression in patients with PPMS [70]. This was a phase 2/3 multicenter, placebo-controlled trial involving 439 patients with PPMS for 96 weeks. There were no differences in the primary endpoint in the overall cohort (time to 12-week confirmed disability progression using the EDSS at 96 weeks). There was a significant effect on T2 lesion volume, which was lower with rituximab. An important point of this trial is the result of the subgroup analysis, which showed a significant difference on the primary endpoint in the subgroup of patients who were <51 years old and who had baseline enhancing lesions (hazard ratio 0.52 (p = 0.010) and 0.41 (p = 0.007), respectively), whereas rituximab-treated patients who were older than 51 years and had no enhancing lesions at baseline had non-significant but worse outcomes than the placebo group (hazard ratio 1.27 (p = 0.425)). This highlights the predominantly anti-inflammatory effect of B-cell therapies in MS.
There has been evidence of the presence of lymphoid follicle-like structures in the cerebral meninges that are typically adjacent to large subpial lesions, and associated with more severe cortical pathology and accelerated disability progression in patients with SPMS [14,15]. Based on these observations, the eventual role of these lymphoid follicles in sustaining cortical injury and accelerating clinical worsening was hypothesized, and the effect of intrathecal rituximab was evaluated since IV rituximab does not cross the blood–brain barrier [104,105,106,107]. The RIVITALISE trial, a randomized, double-blind trial study of intravenous and intrathecal rituximab in patients with SPMS, showed that intrathecal rituximab transiently decreased the B cell counts in the CSF and did not induce consistent effects on CSF biomarkers [104].
A very recent trial evaluated the safety and efficacy of intrathecal rituximab in eight patients with progressive MS who had focal leptomeningeal contrast-enhancement on contrast-enhanced T2-FLAIR [105]. Transient reduction in CSF B cells and biomarkers (reduction in chemokine ligand 13 (CXCL-13) levels with an increase in B cell-activating factor belonging to the TNF family (BAFF) levels), along with profound peripheral B cell depletion was observed; however, the number of leptomeningeal lesions did not change.
Other immunomodulating and immunosuppressive therapies have been investigated in PMS. Examples include azathioprine [108,109,110], cyclophosphamide [111,112,113,114], intravenous immunoglobulins [110,115], methotrexate [116,117,118], cyclosporine [109,119], mycophenolate mofetil [120], laquinimod [121], and MBP8298 [122]. Results from these trials have been largely negative, with no or modest benefit on markers of disability progression.

6.2. Remyelination and Neuroprotection in Progressive MS

While some agents have shown to be beneficial on slowing disability progression, no molecules have shown to have an effect on halting progression or reversing neurological damage in well-powered clinical trials. There are two therapeutic approaches that are thought to be promising to achieve the latter: remyelination and neuroprotection. Remyelinating agents can theoretically repair damage and neuroprotective agents can theoretically prevent axonal loss. High-throughput methods have generated many promising remyelinating molecules to test in pre-clinical studies, to be followed by phase 1, 2, and 3 trials. Unfortunately, studies have been mostly negative or have shown only modest benefits on measures of brain atrophy for which clinical significance still needs to be better elucidated. Table 1 provides an overview of positive trials of molecules with a putative remyelinating and neuroprotective effect. Some agents such as biotin and mesenchymal stem cells may have both remyelinating and neuroprotective effects; these medications are discussed under neuroprotection.
In MS, progression secondary to neurodegeneration is thought to be secondary to chronic demyelination [3]. While neuronal cell bodies and axons have a low potential of regeneration, myelin may have a potential for repair. Remyelination can be seen in shadow plaques, which supports this potential repair property likely mediated by oligodendrocytes [3,6]. A few compounds promoting endogenous oligodendrocyte progenitor cell differentiation have been found in pre-clinical work to be promising remyelinating agents [156]. Out of thousands of compounds identified through high-volume screening of existing drugs [124], opicinumab [128,129,130], and clemastine [125] have been studied in phase 2 trials. Clemastine demonstrated improvement in P100 latency on visual evoked potential in patients with chronic optic neuropathy compared to placebo [125]. Opicinumab showed a similar effect in patients with acute optic neuritis [128,129] (Table 1). Other potentially effective molecules are currently being evaluated, including domperidone (in SPMS, NCT02308137), quetiapine (in RRMS and progressive MS, NCT02087631), liothyronine (in RRMS and progressive MS, NCT02760056), among others. Miconazole and clobetazol have recently been identified as agents with a potential to produce mature oligodendrocytes from progenitor cells [157].
Most of the trials of molecules with a potential for axonal repair have been deceiving. The SPRINT-MS study results were promising; however, phase 3 trials are needed to confirm this result before considering ibudilast as a therapy for progressive MS [132]. Similarly, lipoic acid and simvastatin [134] appear to be potentially beneficial in slowing brain atrophy, and further studies are needed to confirm the results of the phase 2 trials. In a randomized placebo-controlled phase 2 trial, phenytoin was found to have neuroprotective effects after acute optic neuritis compared to placebo [139]. The MS-SMART trial (NCT01910259) failed to show a benefit of amiloride, fluoxetine, or riluzole on brain atrophy in progressive MS (results presented at ECTRIMS 2018). High-dose biotin [153,154,155] and mesenchymal stem cells appear to have a dual effect on neuroprotection and remyelination, and have been studied in multiple small trials and open-label studies in progressive MS, with promising results (Table 1). It is important to note that treatment with autologous hematopoietic stem cell transplantation is reserved for patients with treatment-refractory highly active MS, specifically those who have poor prognostic factors of future disability, including ongoing clinical or radiological activity despite treatment with potent DMT [158], and is out of the scope of this review. Other compounds that are being investigated in phase 2 or 3 trials, include idebenone (NCT01854359), masitinib (NCT01433497), hormone-based therapies (ACTH (NCT01950234)), and erythropoietin (NCT01144117), lithium (NCT01259388), and T-cell receptor vaccines (NCT02057159), among others.

6.3. Symptomatic Management

Optimal symptom management is essential to improve quality of life of patients and to complement the beneficial effect of long-term maintenance therapies in MS. The most commonly encountered symptoms in MS include fatigue, spasticity-related symptoms, neuropathic pain, urinary dysfunction, sleep disturbances, and mood changes. A high number of patients complain of more than one symptom, many of which may be interrelated. For example, poor sleep and depression worsens diurnal fatigue, which can hence not solely be attributed to the disease. Routine evaluations should include screening for persistent symptoms, preferably using validated scales. A general rule in our experience is to start by treating the most disabling or consequential symptom, titrate medications up slowly, use molecules that have a potential to address more than one symptom, and avoid polypharmacy. Combining pharmacological and non-pharmacological approaches to address specific symptoms is important. For example, gait difficulties can be addressed with physical therapy (with a focus on improving ataxia or muscle strength depending on presentation), spasticity management (stretching, anti-spasticity medications, botulinum toxin injections, baclofen pump), and fatigue with dalfampridine. A significant emphasis should also be placed on physical, occupational, and speech therapy. Evaluation and treatment by a multi-disciplinary team is key to provide optimal care across the range of dysfunction in progressive MS. General wellness measures and management of comorbidities should always be discussed with patients, most importantly hyperlipidemia, hypertension, and diabetes control, consuming a healthy diet, weight loss, smoking cessation, vitamin D supplementation, osteoporosis management, and emotional wellness [159].

7. Challenges in Progressive MS Treatment and Research

There are many unmet needs in the field of progressive MS. First, our experience with anti-inflammatory medications like ocrelizumab and siponimod has shown positive but modest results. These medications work on the inflammatory component of the disease, and their potential mechanism of action on the neurodegenerative aspect of MS is probably minimal, if any. As discussed above, the rituximab/ocrelizumab and fingolimod/siponimod experiences in progressive MS provide evidence that therapeutic approaches for progressive MS should probably focus on a different pathophysiological aspect of the disease. Second, trial methodology has significant implications for the effect of agents in progressive MS. Study population selection and amount of disease activity (pre-trial and in-trial annualized relapse rates, presence or absence of baseline gadolinium enhancing lesions) in enrolled subjects are key in driving these efficacy differences between therapies. The definition of progressive MS also varies between trials, and often but not always, a minimum confirmed EDSS step of three at trial entry is required in progressive MS trials, which makes the study populations somewhat heterogeneous and trial results inconsistent and difficult to compare across studies [57].
Third, the selection of appropriate clinical outcome measures plays an important role in capturing treatment effects in progressive MS trials, as discussed earlier in this manuscript [160]. Non-ambulatory patients are typically excluded from progressive MS trials, and the benefit of different therapies on functional domains other than gait function could yield more promising results, specifically in this patient population. For example, studying hand and cognitive function or using composite outcomes like the MSFC rather than relying solely on ambulation as a primary endpoint could inform treatment effects in a more sensitive way. Hand function might be more amenable to treatment compared to lower extremity function in patients with more advanced disability. In the ASCEND trial for instance, natalizumab was associated with a 44% reduction in the relative risk of confirmed upper limb disability progression measured by the 9HPT (adjusted OR 0·56 (95% CI 0·40–0·80); p = 0·001), whereas no benefit was observed on other measures of disability like the EDSS and the T25FW [101]. In a pre-specified baseline subgroup analysis of patients with EDSS ≥6.0 and age >45 years from the OROTARIO trial, ocrelizumab also reduced disability progression as measured by the 12-week confirmed 9HPT in older non-ambulatory patients [161]. Moreover, despite the high prevalence of cognitive dysfunction in progressive MS, there are methodological gaps in outcome measures of cognition in MS, and cognitive function is not adequately and comprehensively evaluated in trials [57,162]. The use of composite outcome measures has been used increasingly in progressive MS trials, and appear to have higher sensitivity to changes. For example, the primary endpoint of the INFORMS trial was a novel composite outcome measure that was defined as a 3-month confirmed change from baseline of the EDSS, the T25FW, or the 9HPT, and although the trial was negative, this outcome measure detected changes with excellent sensitivity in this population [77] Trials evaluating therapies with a remyelinating or repair potential are expected to have a modest clinical effect and can particularly benefit from using composite measures to enhance detection of changes. This approach was used in the opicinimab (SYNERGY) [129] and ibudilast (SPRINT-MS) [132] trials. MRI outcome measures also have some limitations. For example, measures of whole brain atrophy are mostly used as primary endpoints, and have the issue of being variable, which is a concern for interpretability. Other measures like cortical atrophy or magnetization transfer ratio might be more useful depending on the population and mechanism of action of the investigational product.
Finally, the efficacy of molecules with a potential for remyelination and/or neuroprotection needs to be confirmed in large trials with clinically meaningful outcome measures, which will require time and resources. Ultimately, for FDA approval, an agent that can demonstrate clinically meaningful changes in disability measures in large multicenter phase 3 trials is needed. To be able to achieve that, research on the optimal outcome markers is needed. Moreover, many of these molecules are old drugs that do not generate interest in the pharmaceutical industry.
Overall, optimal and novel trial methodology and development of sensitive and clinically meaningful outcome measures and biomarkers are needed in the near-future. The International Progressive MS Alliance and the UK Expert Consortium for Progression in MS Clinical Trials have a mission to expedite development of therapies for progressive forms of MS, which includes the development of optimal trial designs and more responsive outcome measures.

8. Conclusions

A significant amount of effort is delivered to improve knowledge in the field of progressive MS. Future trials will incorporate lessons from previous trials, and hopefully, therapies that halt or even stop neurodegeneration in MS will be available in the future.

Author Contributions

G.M. drafted the manuscript. D.O. reviewed and edited the manuscript for scientific content.

Funding

This research received no external funding.

Conflicts of Interest

Gabrielle Macaron is currently supported by the National Multiple Sclerosis Society Institutional Clinician Training Award ICT 0002. Gabrielle Macaron received fellowship funding from the Biogen Fellowship Grant 6873-P-FEL. She also served in an advisory board for Genentech/Roche. Daniel Ontaneda received research support from the National Multiple Sclerosis Society, National Institutes of Health, Patient Centered Research Institute, Race to Erase MS Foundation, Genentech, and Genzyme. He also received consulting fees from Biogen Idec, Genentech/Roche, Genzyme, Novartis, and Merck.

References

  1. Wallin, M.T.; Culpepper, W.J.; Campbell, J.D.; Nelson, M.L.; Langer-Gould, A.; Marrie, R.A.; Cutter, G.R.; Kaye, W.E.; Wagner, L.; Tremlett, H.; et al. The prevalence of MS in the United States: A population-based estimate using health claims data. Neurology 2019, 92, e1019–e1024. [Google Scholar] [CrossRef] [PubMed]
  2. Browne, P.; Chandraratna, D.; Angood, C.; Tremlett, H.; Baker, C.; Taylor, B.V.; Thompson, A.J. Atlas of Multiple Sclerosis 2013: A growing global problem with widespread inequity. Neurology 2014, 83, 1022–1024. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Mahad, D.H.; Trapp, B.D.; Lassmann, H. Pathological mechanisms in progressive multiple sclerosis. Lancet Neurol. 2015, 14, 183–193. [Google Scholar] [CrossRef]
  4. Trapp, B.D.; Peterson, J.; Ransohoff, R.M.; Rudick, R.; Mörk, S.; Bö, L. Axonal Transection in the Lesions of Multiple Sclerosis. N. Engl. J. Med. 1998, 338, 278–285. [Google Scholar] [CrossRef] [PubMed]
  5. Kornek, B.; Storch, M.K.; Weissert, R.; Wallstroem, W.; Stefferl, A.; Olsson, T.; Linington, C.; Schmidbauer, M.; Lassmann, H. Multiple sclerosis and chronic autoimmune encephalomyelitis: A comparative quantitative study of axonal injury in active, inactive, and remyelinated lesions. Am. J. Pathol. 2000, 157, 267–276. [Google Scholar] [CrossRef]
  6. Frischer, J.M.; Weigand, S.D.; Guo, Y.; Kale, N.; Parisi, J.E.; Pirko, I.; Mandrekar, J.; Bramow, S.; Metz, I.; Bruck, W.; et al. Clinical and Pathological Insights into the Dynamic Nature of the White Matter Multiple Sclerosis Plaque. Ann. Neurol. 2015, 78, 710–721. [Google Scholar] [CrossRef]
  7. De Stefano, N.; Giorgio, A.; Battaglini, M.; Rovaris, M.; Sormani, M.P.; Barkhof, F.; Korteweg, T.; Enzinger, C.; Fazekas, F.; Calabrese, M.; et al. Assessing brain atrophy rates in a large population of untreated multiple sclerosis subtypes. Neurology 2010, 74, 1868–1876. [Google Scholar] [CrossRef] [PubMed]
  8. Lucchinetti, C.F.; Rauschka, H.; Bergmann, M.; Schmidbauer, M.; Kutzelnigg, A.; Parisi, J.E.; Stadelmann, C.; Brück, W.; Lassmann, H. Cortical demyelination and diffuse white matter injury in multiple sclerosis. Brain 2005, 128, 2705–2712. [Google Scholar]
  9. Amato, M.P.; Portaccio, E.; Goretti, B.; Zipoli, V.; Battaglini, M.; Bartolozzi, M.L.; Stromillo, M.L.; Guidi, L.; Siracusa, G.; Sorbi, S.; et al. Association of Neocortical Volume Changes with Cognitive Deterioration in Relapsing-Remitting Multiple Sclerosis. Arch. Neurol. 2007, 64, 1157–1161. [Google Scholar] [CrossRef]
  10. Calabrese, M.; Agosta, F.; Rinaldi, F.; Mattisi, I.; Grossi, P.; Favaretto, A.; Atzori, M.; Bernardi, V.; Barachino, L.; Rinaldi, L.; et al. Cortical Lesions and Atrophy Associated with Cognitive Impairment in Relapsing-Remitting Multiple Sclerosis. Arch. Neurol. 2009, 66, 1144–1150. [Google Scholar] [CrossRef]
  11. Mahad, D.J.; Ziabreva, I.; Campbell, G.; Lax, N.; White, K.; Hanson, P.S.; Lassmann, H.; Turnbull, D.M. Mitochondrial changes within axons in multiple sclerosis. Brain 2009, 132, 1161–1174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Trapp, B.D.; Stys, P.K. Virtual hypoxia and chronic necrosis of demyelinated axons in multiple sclerosis. Lancet Neurol. 2009, 8, 280–291. [Google Scholar] [CrossRef]
  13. Ontaneda, D.; Thompson, A.J.; Fox, R.J.; ACohen, J. Progressive multiple sclerosis: Prospects for disease therapy, repair, and restoration of function. Lancet 2017, 389, 1357–1366. [Google Scholar] [CrossRef]
  14. Howell, O.W.; Reeves, C.A.; Nicholas, R.; Carassiti, D.; Radotra, B.; Gentleman, S.M.; Serafini, B.; Aloisi, F.; Roncaroli, F.; Magliozzi, R.; et al. Meningeal inflammation is widespread and linked to cortical pathology in multiple sclerosis. Brain 2011, 134, 2755–2771. [Google Scholar] [CrossRef] [PubMed]
  15. Magliozzi, R.; Howell, O.; Vora, A.; Serafini, B.; Nicholas, R.; Puopolo, M.; Reynolds, R.; Aloisi, F. Meningeal B-cell follicles in secondary progressive multiple sclerosis associate with early onset of disease and severe cortical pathology. Brain 2007, 130, 1089–1104. [Google Scholar] [CrossRef]
  16. Brown, J.W.L.; Coles, A.; Horakova, D.; Havrdova, E.; Izquierdo, G.; Prat, A.; Girard, M.; Duquette, P.; Trojano, M.; Lugaresi, A.; et al. Association of Initial Disease-Modifying Therapy with Later Conversion to Secondary Progressive Multiple Sclerosis. JAMA 2019, 321, 175–187. [Google Scholar] [CrossRef] [PubMed]
  17. Bergamaschi, R.; Quaglini, S.; Tavazzi, E.; Amato, M.P.; Paolicelli, D.; Zipoli, V.; Romani, A.; Tortorella, C.; Portaccio, E.; D’Onghia, M.; et al. Immunomodulatory therapies delay disease progression in multiple sclerosis. Mult. Scler. J. 2016, 22, 1732–1740. [Google Scholar] [CrossRef]
  18. Trojano, M.; Pellegrini, F.; Paolicelli, D.; Fuiani, A.; Zimatore, G.B.; Tortorella, C.; Simone, I.L.; Patti, F.; Ghezzi, A.; Zipoli, V.; et al. Real-life impact of early interferon beta therapy in relapsing multiple sclerosis. Ann. Neurol. 2009, 66, 513–520. [Google Scholar] [CrossRef]
  19. Trojano, M.; Pellegrini, F.; Fuiani, A.; Paolicelli, D.; Zipoli, V.; Zimatore, G.B.; Monte, E.D.; Portaccio, C.; Lepore, V.; Livrea, P.; et al. New natural history of interferon-beta-treated relapsing multiple sclerosis. Ann. Neurol. 2007, 61, 300–306. [Google Scholar] [CrossRef]
  20. Haider, L.; Simeonidou, C.; Steinberger, G.; Hametner, S.; Grigoriadis, N.; Deretzi, G.; Kovacs, G.G.; Kutzelnigg, A.; Lassmann, H.; Frischer, J.M. Multiple sclerosis deep grey matter: The relation between demyelination, neurodegeneration, inflammation and iron. J. Neurol. Neurosurg. Psychiatry 2014, 85, 1386–1395. [Google Scholar] [CrossRef]
  21. Hametner, S.; Wimmer, I.; Haider, L.; Pfeifenbring, S.; Brück, W.; Lassmann, H. Iron and neurodegeneration in the multiple sclerosis brain. Ann. Neurol. 2013, 74, 848–861. [Google Scholar] [CrossRef]
  22. Harding, K.E.; Liang, K.; Cossburn, M.D.; Ingram, G.; Hirst, C.; Pickersgill, T.; Te Water, N.; Johann, W.; Ben-Shlomo, M.; Robertson, Y.; et al. Long-term outcome of paediatric-onset multiple sclerosis: A population-based study. J Neurol. Neurosurg. Psychiatry 2013, 84, 141–147. [Google Scholar] [CrossRef]
  23. Leray, E.; Yaouanq, J.; Le Page, E.; Coustans, M.; Laplaud, D.; Oger, J.; Edan, G. Evidence for a two-stage disability progression in multiple sclerosis. Brain 2010, 133, 1900–1913. [Google Scholar] [CrossRef] [Green Version]
  24. Confavreux, C.; Moreau, T.; Vukusic, S.; Adeleine, P. Relapses and Progression of Disability in Multiple Sclerosis. N. Engl. J. Med. 2000, 343, 1430–1438. [Google Scholar] [CrossRef]
  25. Beiki, O.; Frumento, P.; Bottai, M.; Manouchehrinia, A.; Hillert, J. Changes in the Risk of Reaching Multiple Sclerosis Disability Milestones in Recent Decades: A Nationwide Population-Based Cohort Study in Sweden. JAMA Neurol. 2019, 1–7. [Google Scholar] [CrossRef]
  26. Polman, C.H.; Reingold, S.C.; Banwell, B.; Clanet, M.; Cohen, J.A.; Filippi, M.; Fujihara, K.; Havrdova, E.K.; Hutchinson, M.; Kappos, L.; et al. Diagnostic criteria for multiple sclerosis: 2010 Revisions to the McDonald criteria. Ann. Neurol. 2011, 69, 292–302. [Google Scholar] [CrossRef] [Green Version]
  27. Thompson, A.J.; Banwell, B.L.; Barkhof, F.; Carroll, W.; Coetzee, T.; Comi, G.; Correale, J.; Fazekas, F.; Filippi, M.; Freedman, M.; et al. Diagnosis of multiple sclerosis: 2017 revisions of the McDonald criteria. Lancet Neurol. 2018, 17, 162–173. [Google Scholar] [CrossRef]
  28. Lublin, F.; Stephen, C.; Reingold, P.; Cohen, J.A.; Cutter, G.; Sørensen, P.S.; Thompson, A.J.; Wolinsky, J.S.; Balcer, L.J.; Banwell, B.; et al. Defining the clinical course of multiple sclerosis: The 2013 revisions. Neurology 2014, 83, 278–286. [Google Scholar] [CrossRef] [Green Version]
  29. Whitaker, J.N.; McFarland, H.F.; Rudge, P.; Reingold, R.S. Outcomes assessment in multiple sclerosis trials. Mult. Scler. 1995, 1, 37–47. [Google Scholar] [CrossRef]
  30. Noseworthy, J.H.; Vandervoort, M.K.; Wong, C.J.; Ebers, G.C. Interrater variability with the Expanded Disability Status Scale (EDSS) and Functional Systems (FS) in a multiple sclerosis clinical trial. Neurology 1990, 40, 971. [Google Scholar] [CrossRef]
  31. Goodkin, D.E.; Cookfair, D.; Wende, K.; Bourdette, D.; Pullicino, P.; Scherokman, B.; Whitham, R. Inter- and intrarater scoring agreement using grades 1.0 to 3.5 of the Kurtzke Expanded Disability Status Scale (EDSS). Neurology 1992, 42, 859. [Google Scholar] [CrossRef]
  32. Kragt, J.J.; Nielsen, J.M.; Van Der Linden, F.A.; Uitdehaag, B.M.; Polman, C.H. How similar are commonly combined criteria for EDSS progression in multiple sclerosis? Mult. Scler. J. 2006, 12, 782–786. [Google Scholar] [CrossRef]
  33. Ontaneda, D.; ACohen, J.; Amato, M.P. Clinical outcome measures for progressive MS trials. Mult. Scler. J. 2017, 23, 1627–1635. [Google Scholar] [CrossRef] [Green Version]
  34. Cutter, G.R.; Baier, M.L.; Rudick, R.A.; Cookfair, D.L.; Fischer, J.S.; Petkau, J.; Syndulko, K.; Weinshenker, B.G.; Antel, J.P.; Confavreux, C.; et al. Development of a multiple sclerosis functional composite as a clinical trial outcome measure. Brain 1999, 122, 871–882. [Google Scholar] [CrossRef]
  35. Fischer, J.; Rudick, R.; Cutter, G.; Reingold, S. The Multiple Sclerosis Functional Composite measure (MSFC): An integrated approach to MS clinical outcome assessment. Mult. Scler. J. 1999, 5, 244–250. [Google Scholar] [CrossRef]
  36. Cohen, J.A.; Cutter, G.R.; Fischer, J.S.; Goodman, A.D.; Heidenreich, F.R.; Jak, A.J.; Kniker, J.E.; Kooijmans, M.F.; Lull, J.M.; Sandrock, A.W.; et al. Use of the Multiple Sclerosis Functional Composite as an Outcome Measure in a Phase 3 Clinical Trial. Arch. Neurol. 2001, 58, 961–967. [Google Scholar] [CrossRef]
  37. Polman, C.H.; Rudick, R.A. The Multiple Sclerosis Functional Composite: A clinically meaningful measure of disability. Neurology 2010, 74, 8–15. [Google Scholar] [CrossRef]
  38. Kalkers, N.; Bergers, L.; De Groot, V.; Lazeron, R.; Van Walderveen, M.; Uitdehaag, B.; Polman, C.; Barkhof, F. Concurrent validity of the MS Functional Composite using MRI as a biological disease marker. Neurology 2001, 56, 215–219. [Google Scholar] [CrossRef]
  39. Miller, D.M.; Rudick, R.A.; Cutter, G.; Baier, M.; Fischer, J.S. Clinical Significance of the Multiple Sclerosis Functional Composite. Arch. Neurol. 2000, 57, 1319–1324. [Google Scholar] [CrossRef] [Green Version]
  40. Cohen, J.A.; Cutter, G.R.; Fischer, J.S.; Goodman, A.; Heidenreich, F.R.; Kooijmans, M.F.; Sandrock, A.W.; Rudick, R.A.; Simon, J.H.; Simonian, N.A.; et al. Benefit of interferon -1a on MSFC progression in secondary progressive multiple sclerosis. Neurology 2002, 59, 679–687. [Google Scholar] [CrossRef]
  41. Baier, M.L.; Cutter, G.R.; Rudick, R.A.; Miller, D.; Cohen, J.A.; Weinstock-Guttman, B.; Mass, M.; Balcer, L.J. Low-contrast letter acuity testing captures visual dysfunction in patients with multiple sclerosis. Neurology 2005, 64, 992–995. [Google Scholar] [CrossRef]
  42. Balcer, L.J.; Baier, M.L.; Cohen, J.A.; Kooijmans, M.F.; Sandrock, A.W.; Nano-Schiavi, M.L.; Pfohl, D.C.; Mills, M.; Bowen, J.; Ford, C.; et al. Contrast letter acuity as a visual component for the Multiple Sclerosis Functional Composite. Neurology 2003, 61, 1367–1373. [Google Scholar] [CrossRef]
  43. Freeman, J.; Hobart, J.; Thompson, A. Kurtzke scales revisited: The application of psychometric methods to clinical intuition. Brain 2000, 123, 1027–1040. [Google Scholar]
  44. Rasova, K.; Martinkova, P.; Vyskotova, J.; Sedova, M. Assessment set for evaluation of clinical outcomes in multiple sclerosis: Psychometric properties. Patient Relat. Outcome Meas. 2012, 3, 59–70. [Google Scholar] [CrossRef]
  45. ACohen, J.; Coles, A.J.; Arnold, D.L.; Confavreux, C.; Fox, E.J.; Hartung, H.-P.; Havrdova, E.K.; Selmaj, K.W.; Weiner, H.L.; Fisher, E.; et al. Alemtuzumab versus interferon beta 1a as first-line treatment for patients with relapsing-remitting multiple sclerosis: A randomised controlled phase 3 trial. Lancet 2012, 380, 1819–1828. [Google Scholar]
  46. Polman, C.; Hohlfeld, R.; Agoropoulou, C.; Leyk, M.; Zhang-Auberson, L.; Burtin, P.; Kappos, L.; Radue, E.-W.; O’Connor, P.; Calabresi, P.; et al. A Placebo-Controlled Trial of Oral Fingolimod in Relapsing Multiple Sclerosis. N. Engl. J. Med. 2010, 362, 387–401. [Google Scholar] [Green Version]
  47. Meyer-Moock, S.; Feng, Y.-S.; Maeurer, M.; Dippel, F.-W.; Kohlmann, T. Systematic literature review and validity evaluation of the Expanded Disability Status Scale (EDSS) and the Multiple Sclerosis Functional Composite (MSFC) in patients with multiple sclerosis. BMC Neurol. 2014, 14, 58. [Google Scholar] [CrossRef]
  48. Rudick, R.A.; Miller, D.; Bethoux, F.; Rao, S.M.; Lee, J.-C.; Stough, D.; Reece, C.; Schindler, D.; Mamone, B.; Alberts, J. The Multiple Sclerosis Performance Test (MSPT): An iPad-Based Disability Assessment Tool. J. Vis. Exp. 2014, e51318. [Google Scholar] [CrossRef]
  49. Macaron, G.; Moss, B.P.; Li, H.; Baldassari, L.; Rao, S.; Schindler, D.; Alberts, J.; Weber, M.; Ayers, M.; Bethoux, F.; et al. Technology-enabled assessments to enhance multiple sclerosis clinical care and research. Neurol. Clin. Pract. 2019, in press. [Google Scholar]
  50. Motl, R.W.; A Cohen, J.; Benedict, R.; Phillips, G.; LaRocca, N.; Hudson, L.D.; Rudick, R. Multiple Sclerosis Outcome Assessments Consortium Validity of the timed 25-foot walk as an ambulatory performance outcome measure for multiple sclerosis. Mult. Scler. J. 2017, 23, 704–710. [Google Scholar] [CrossRef]
  51. Feng, J.; Qu, J.; Felix, C.; McGinley, M.; Nakamura, K.; Macaron, G.; Moss, B.; Li, H.; Jones, S.; Rao, S.; et al. Quantitative MRI and patient-reported outcomes validate clinically meaningful changes on tests of ambulation and hand function. ACTRIMS 2019, P050. [Google Scholar]
  52. Feys, P.; Lamers, I.; Francis, G.; Benedict, R.; Phillips, G.; LaRocca, N.; Hudson, L.D.; Rudick, R. Multiple Sclerosis Outcome Assessments Consortium the Nine-Hole Peg Test as a manual dexterity performance measure for multiple sclerosis. Mult. Scler. J. 2017, 23, 711–720. [Google Scholar] [CrossRef]
  53. Benedict, R.H.; DeLuca, J.; Phillips, G.; LaRocca, N.; Hudson, L.D.; Rudick, R. Multiple Sclerosis Outcome Assessments Consortium Validity of the Symbol Digit Modalities Test as a cognition performance outcome measure for multiple sclerosis. Mult. Scler. J. 2017, 23, 721–733. [Google Scholar] [CrossRef]
  54. Macaron, G.; Moss, B.; Baldassari, L.E.; Conway, D.; McGinley, M.; Alshehri, E.; Feng, J.; Bermel, R.; Boissy, A.; Cohen, J.A.; et al. Cross-sectional predictive value of clinically meaningful change in processing speed on self-reported cognition and MRI metrics. ACTRIMS 2019, P034. [Google Scholar]
  55. Balcer, L.J.; Raynowska, J.; Nolan, R.; Galetta, S.L.; Kapoor, R.; Benedict, R.; Phillips, G.; LaRocca, N.; Hudson, L.; Rudick, R.; et al. Validity of low-contrast letter acuity as a visual performance outcome measure for multiple sclerosis. Mult. Scler. J. 2017, 23, 734–747. [Google Scholar] [CrossRef]
  56. Oh, J.; Ontaneda, D.; Azevedo, C.; Klawiter, E.; Absinta, M.; Arnold, D.; Bakshi, R.; Calabresi, P.; Crainiceanu, C.; Dewey, B.; et al. Imaging outcome measures of neuroprotection repair in MS: A consensus statement from NAIMS. Neurology 2019, 92, 519–533. [Google Scholar] [CrossRef]
  57. Ontaneda, D.; Fox, R.J.; Chataway, J. Clinical trials in progressive multiple sclerosis: Lessons learned and future perspectives. Lancet Neurol. 2015, 14, 208–223. [Google Scholar] [CrossRef]
  58. Mahajan, K.R.; Ontaneda, D. The Role of Advanced Magnetic Resonance Imaging Techniques in Multiple Sclerosis Clinical Trials. Neurotherapeutics 2017, 14, 905–923. [Google Scholar] [CrossRef]
  59. Hardmeier, M.; Leocani, L.; Fuhr, P. A new role for evoked potentials in MS? Repurposing evoked potentials as biomarkers for clinical trials in MS. Mult. Scler. J. 2017, 23, 1309–1319. [Google Scholar] [CrossRef]
  60. Saidha, S.; Al-Louzi, O.; Ratchford, J.N.; Bhargava, P.; Oh, J.; Newsome, S.D.; Prince, J.L.; Pham, D.; Roy, S.; Van Zijl, P.; et al. Optical Coherence Tomography Reflects Brain Atrophy in Multiple Sclerosis: A Four-Year Study. Ann. Neurol. 2015, 78, 801–813. [Google Scholar] [CrossRef]
  61. Novakova, L.; Zetterberg, H.; Sundström, P.; Axelsson, M.; Khademi, M.; Gunnarsson, M.; Malmeström, C.; Svenningsson, A.; Olsson, T.; Piehl, F.; et al. Monitoring disease activity in multiple sclerosis using serum neurofilament light protein. Neurology 2017, 89, 2230–2237. [Google Scholar] [CrossRef] [Green Version]
  62. Damasceno, A.; Dias-Carneiro, R.P.C.; Moraes, A.S.; Boldrinib, V.O.; Quintilianob, R.P.S.; de Paula Galdino da Silvab, V.A.; Fariasb, A.S.; Brandãob, C.O.; Damascenoa, B.P.; dos Santos, L.M.B.; et al. Clinical MRI correlates of CSF neurofilament light chain levels in relapsing progressive multiple sclerosis. Mult. Scler. Relat. Disord. 2019, 30, 149–153. [Google Scholar] [CrossRef]
  63. Martin, S.J.; McGlasson, S.; Hunt, D.; Overell, J. Cerebrospinal fluid neurofilament light chain in multiple sclerosis and its subtypes: A meta-analysis of case-control studies. J. Neurol. Neurosurg. Psychiatry 2019. [Google Scholar] [CrossRef]
  64. Pawlitzki, M.; Schreiber, S.; Bittner, D.; Kreipe, J.; Leypoldt, F.; Rupprecht, K.; Carare, R.O.; Meuth, S.G.; Vielhaber, S.; Körtvélyessy, P. CSF Neurofilament Light Chain Levels in Primary Progressive MS: Signs of Axonal Neurodegeneration. Front. Neurol. 2018, 9, 9. [Google Scholar] [CrossRef]
  65. Barro, C.; Naegelin, Y.; Schädelin, S.; Giardiello, A.; Zecca, C.; Blennow, K.; Zetterberg, H.; Leppert, D.; Gobbi, C.; Kuhle, J.; et al. Serum Neurofilament light: A biomarker of neuronal damage in multiple sclerosis. Ann. Neurol. 2017, 81, 857–870. [Google Scholar]
  66. Barro, C.; Benkert, P.; Disanto, G.; Tsagkas, C.; Amann, M.; Naegelin, Y.; Leppert, D.; Gobbi, C.; Granziera, C.; Yaldizli, O.; et al. Serum neurofilament as a predictor of disease worsening and brain and spinal cord atrophy in multiple sclerosis. Brain 2018, 141, 2382–2391. [Google Scholar] [CrossRef]
  67. Hauser, S.L.; Bar-Or, A.; Comi, G.; Giovannoni, G.; Hartung, H.-P.; Hemmer, B.; Lublin, F.; Montalban, X.; Rammohan, K.W.; Selmaj, K.; et al. Ocrelizumab versus Interferon Beta-1a in Relapsing Multiple Sclerosis. N. Engl. J. Med. 2017, 376, 221–234. [Google Scholar] [CrossRef]
  68. Montalban, X.; Hauser, S.L.; Kappos, L.; Arnold, D.L.; Bar-Or, A.; Comi, G.; De Seze, J.; Giovannoni, G.; Hartung, H.-P.; Hemmer, B.; et al. Ocrelizumab versus Placebo in Primary Progressive Multiple Sclerosis. N. Engl. J. Med. 2017, 376, 209–220. [Google Scholar] [CrossRef]
  69. Fox, E.; Markowitz, C.; Applebee, A.; Montalban, X.; Wolinsky, J.S.; Belachew, S.; Damian, F.; Han, J.; Musch, B.; Giovannoni, G. Effect of ocrelizumab on upper limb function in patients with primary progressive multiple sclerosis (PPMS) in the oratorio study (ENCORE). J. Neurol. Neurosurg. Psychiatry 2018, 89, A14. [Google Scholar] [CrossRef]
  70. Hawker, K.; O’Connor, P.; Freedman, M.S.; Calabresi, P.A.; Antel, J.; Simon, J.; Hauser, S.; Waubant, E.; Vollmer, T.; Panitch, H.; et al. Rituximab in patients with primary progressive multiple sclerosis: Results of a randomized double-blind placebo-controlled multicenter trial. Ann. Neurol. 2009, 66, 460–471. [Google Scholar] [CrossRef]
  71. Naegelin, Y.; Naegelin, P.; Von Felten, S.; Lorscheider, J.; Sonder, J.; Uitdehaag, B.M.J.; Scotti, B.; Zecca, C.; Gobbi, C.; Kappos, L.; et al. Association of Rituximab Treatment with Disability Progression Among Patients With Secondary Progressive Multiple Sclerosis. JAMA Neurol. 2019, 76, 274. [Google Scholar] [CrossRef]
  72. Gelfand, J.M.; Cree, B.A.C.; Hauser, S.L. Ocrelizumab and Other CD20+ B-Cell-Depleting Therapies in Multiple Sclerosis. Neurotherapeutics 2017, 14, 835–841. [Google Scholar] [CrossRef] [Green Version]
  73. Klein, C.; Lammens, A.; Schäfer, W.; Georges, G.; Schwaiger, M.; Mössner, E.; Hopfner, K.-P.; Umana, P.; Niederfellner, G. Epitope interactions of monoclonal antibodies targeting CD20 and their relationship to functional properties. mAbs 2013, 5, 22–33. [Google Scholar] [CrossRef] [Green Version]
  74. Feng, J.J.; Ontaneda, D. Treating primary-progressive multiple sclerosis: Potential of ocrelizumab and review of B-cell therapies. Degener. Neurol. Neuromuscul. Dis. 2017, 7, 31–45. [Google Scholar] [CrossRef]
  75. Kappos, L.; Bar-Or, A.; Cree, B.A.C.; Fox, R.J.; Giovannoni, G.; Gold, R.; Vermersch, P.; Arnold, D.L.; Arnould, S.; Scherz, T.; et al. Siponimod versus placebo in secondary progressive multiple sclerosis (EXPAND): A double-blind, randomised, phase 3 study. Lancet 2018, 391, 1263–1273. [Google Scholar] [CrossRef]
  76. Jackson, S.J.; Giovannoni, G.; Baker, D. Fingolimod modulates microglial activation to augment markers of remyelination. J. Neuroinflamm. 2011, 8, 76. [Google Scholar] [CrossRef]
  77. Lublin, F.; Miller, D.H.; Freedman, M.S.; Cree, B.A.C.; Wolinsky, J.S.; Weiner, H.; Lubetzki, C.; Hartung, H.-P.; Montalban, X.; Uitdehaag, B.M.J.; et al. Oral fingolimod in primary progressive multiple sclerosis (INFORMS): A phase 3, randomised, double-blind, placebo-controlled trial. Lancet 2016, 387, 1075–1084. [Google Scholar] [CrossRef]
  78. Martinelli Boneschi, F.; Vacchi, L.; Rovaris, M.; Capra, R.; Comi, G. Mitoxantrone for multiple sclerosis. Cochrane Database Syst. Rev. 2013, 5, CD002127. [Google Scholar] [CrossRef]
  79. Hartung, H.P.; Gonsette, R.; König, N.; Kwiecinski, H.; Guseo, A.; Morrissey, S.; Krapf, S.; Zwingers, T. Mitoxantrone in Multiple Sclerosis Study Group. Mitoxantrone in progressive multiple sclerosis: A placebo-controlled, double-blind, randomised, multicentre trial. Lancet 2002, 28, 2018–2025. [Google Scholar] [CrossRef]
  80. Krapf, H.; Morrissey, S.P.; Zenker, O.; Zwingers, T.; Gonsette, R.; Hartung, H.-P. Effect of mitoxantrone on MRI in progressive MS: Results of the MIMS trial. Neurology 2005, 65, 690–695. [Google Scholar] [CrossRef]
  81. Grey Née Cotte, S.; Salmen Née Stroet, A.; Von Ahsen, N.; Starck, M.; Winkelmann, A.; Zettl, U.K.; Comabella, M.; Montalban, X.; Zipp, F.; Fleischer, V.; et al. Lack of efficacy of mitoxantrone in primary progressive Multiple Sclerosis irrespective of pharmacogenetic factors: A multi-center, retrospective analysis. J. Neuroimmunol. 2015, 278, 277–279. [Google Scholar] [CrossRef]
  82. Pelfrey, C.M.; Cotleur, A.C.; Zamor, N.; Lee, J.C.; Robert, R.J. Immunological studies of mitoxantrone in primary progressive multiple sclerosis. J. Neuroimmunol. 2006, 175, 192–199. [Google Scholar] [CrossRef]
  83. Cotte, S.; Von Ahsen, N.; Kruse, N.; Huber, B.; Winkelmann, A.; Zettl, U.K.; Starck, M.; König, N.; Téllez, N.; Dorr, J.; et al. ABC-transporter gene-polymorphisms are potential pharmacogenetic markers for mitoxantrone response in multiple sclerosis. Brain 2009, 132, 2517–2530. [Google Scholar] [CrossRef] [Green Version]
  84. US Food and Drug Administration. Press Announcement−FDA Approves New Oral Drug to Treat Multiple Sclerosis. Office of the Commissioner. Available online: https://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm634469.htm (accessed on 2 April 2019).
  85. Giovannoni, G.; Comi, G.; Cook, S.; Rammohan, K.; Rieckmann, P.; Sørensen, P.S.; Vermersch, P.; Chang, P.; Hamlett, A.; Musch, B.; et al. A Placebo-Controlled Trial of Oral Cladribine for Relapsing Multiple Sclerosis. N. Engl. J. Med. 2010, 362, 416–426. [Google Scholar] [CrossRef]
  86. Montalban, X.; Leist, T.P.; Cohen, B.A.; Moses, H.; Campbell, J.; Hicking, C.; Dangond, F. Cladribine tablets added to IFN-β in active relapsing multiple sclerosis. Neurol. Neuroimmunol. NeuroInflamm. 2018, 5, 477. [Google Scholar] [CrossRef]
  87. Leist, T.P.; Comi, G.; Cree, B.A.C.; Coyle, P.K.; Freedman, M.S.; Hartung, H.-P.; Vermersch, P.; Casset-Semanaz, F.; Scaramozza, M. Effect of oral cladribine on time to conversion to clinically definite multiple sclerosis in patients with a first demyelinating event (ORACLE MS): A phase 3 randomised trial. Lancet Neurol. 2014, 13, 257–267. [Google Scholar] [CrossRef]
  88. Sipe, J. Cladribine in treatment of chronic progressive multiple sclerosis. Lancet 1994, 344, 9–13. [Google Scholar] [CrossRef]
  89. Rice, G.P.A.; Filippi, M.; Comi, G. Cladribine and progressive MS: Clinical and MRI outcomes of a multicenter controlled trial. Neurology 2000, 54, 1145–1155. [Google Scholar] [CrossRef]
  90. Filippi, M.; Rovaris, M.; Iannucci, G.; Mennea, S.; Sormani, M.P.; Comi, G. Whole brain volume changes in patients with progressive multiple sclerosis treated with cladribine. Neurology 2000, 55, 1714–1718. [Google Scholar] [CrossRef]
  91. Polman, C.; Pozzilli, C.; Thompson, A.; Beckmann, K.; Dahlke, F. Final analysis of the European multicenter trial on IFNbeta-1b in secondary-progressive multiple sclerosis. Neurology 2001, 57, 1969–1975. [Google Scholar]
  92. The North American Study Group. Interferon beta-1b in secondary progressive MS: Results from a 3-year controlled study. Neurology 2004, 63, 1788–1795. [Google Scholar] [CrossRef]
  93. Kappos, L.; Weinstock-Guttman, B.; Pozzilli, C.; Thompson, A.J.; Dahlke, F.; Beckmann, K.; Polman, C.; McFarland, H.; European (EU-SPMS) Interferon beta-1b in Secondary Progressive Multiple Sclerosis Trial Steering Committee and Independent Advisory Board; North American (NA-SPMS) Interferon beta-1b in Secondary Progressive Multiple Sclerosis Trial Steering Committee and Independent Advisory Board. Interferon beta-1b in secondary progressive multiple sclerosis. A combined analysis of the two trials. Neurology 2004, 63, 1779–1787. [Google Scholar] [CrossRef]
  94. Secondary Progressive Efficacy Clinical Trial of Recombinant Interferon-Beta-1a in MS (SPECTRIMS) Study Group. Randomized controlled trial of interferon- beta-1a in secondary progressive MS: Clinical results. Neurology 2001, 56, 1496–1504. [Google Scholar] [CrossRef]
  95. Andersen, O.; Elovaara, I.; Färkkilä, M.; Hansen, H.J.; Mellgren, S.; Myhr, K.-M.; Sandberg-Wollheim, M.; Soelberg, S. Multicentre, randomised, double blind, placebo controlled, phase III study of weekly, low dose, subcutaneous interferon beta-1a in secondary progressive multiple sclerosis. J. Neurol. Neurosurg. Psychiatry 2004, 75, 706–710. [Google Scholar] [CrossRef] [Green Version]
  96. Leary, S.M.; Miller, D.H.; Stevenson, V.L.; Brex, P.A.; Chard, D.T.; Thompson, A.J. Interferon beta-1a in primary progressive MS: An exploratory, randomized, controlled trial. Neurology 2003, 60, 44–51. [Google Scholar] [CrossRef]
  97. Montalban, X. Overview of European pilot study of interferon beta-Ib in primary progressive multiple sclerosis. Mult. Scler. 2004, 10, S62–S64. [Google Scholar]
  98. Montalban, X.; Garriga, J.S.; Tintoré, M.; Brieva, L.; Aymerich, F.; Rio, J.; Porcel, J.; Borràs, C.; Nos, C.; Rovira, À.; et al. A single-center, randomized, double-blind, placebo-controlled study of interferon beta-1b on primary progressive and transitional multiple sclerosis. Mult. Scler. J. 2009, 15, 1195–1205. [Google Scholar] [CrossRef]
  99. Wolinsky, J.S.; Narayana, P.A.; O’Connor, P.; Coyle, P.K.; Ford, C.; Johnson, K.; Miller, A.; Pardo, L.; Kadosh, S.; Ladkani, D.; et al. Glatiramer acetate in primary progressive multiple sclerosis: Results of a multinational, multicenter, double-blind, placebo-controlled trial. Ann. Neurol. 2007, 61, 14–24. [Google Scholar] [CrossRef]
  100. Wolinsky, J.S.; Shochat, T.; Weiss, S.; Ladkani, D. Glatiramer acetate treatment in PPMS: Why males appear to respond favorably. J. Neurol. Sci. 2009, 286, 92–98. [Google Scholar] [CrossRef]
  101. Ho, P.-R.; Campbell, N.; Chang, I.; Deykin, A.; Forrestal, F.; Lucas, N.; Yu, B.; Arnold, D.L.; Hartung, H.-P.; Miller, A.; et al. Effect of natalizumab on disease progression in secondary progressive multiple sclerosis (ASCEND): A phase 3, randomised, double-blind, placebo-controlled trial with an open-label extension. Lancet Neurol. 2018, 17, 405–415. [Google Scholar]
  102. Graves, D.; Vernino, S. Immunotherapies in Neurologic Disorders. Med. Clin. N. Am. 2012, 96, 497–523. [Google Scholar] [CrossRef]
  103. Salzer, J.; Svenningsson, R.; Alping, P.; Novakova, L.; Bjorck, A.; Fink, K.; Islam-Jakobsson, P.; Malmestrom, C.; Axelsson, M.; Vagberg, M. Rituximab in multiple sclerosis: A retrospective observational study on safety and efficacy. Neurology 2016, 87, 2074–2081. [Google Scholar] [CrossRef]
  104. Komori, M.; Lin, Y.C.; Cortese, I.; Blake, A.; Ohayon, J.; Cherup, J.; Maric, D.; Kosa, P.; Wu, T.; Bielekova, B. Insufficient disease inhibition by intrathecal rituximab in progressive multiple sclerosis. Ann. Clin. Transl. Neurol. 2016, 3, 166–179. [Google Scholar] [CrossRef]
  105. Bhargava, P.; Wicken, C.; Smith, M.D.; Strowd, R.E.; Cortese, I.; Reich, D.S.; Calabresi, P.A.; Mowry, E.M. Trial of intrathecal rituximab in progressive multiple sclerosis patients with evidence of leptomeningeal contrast enhancement. Mult. Scler. Relat. Disord. 2019, 30, 136–140. [Google Scholar] [CrossRef]
  106. Bergman, J.; Burman, J.; Gilthorpe, J.D.; Zetterberg, H.; Jiltsova, E.; Bergenheim, T.; Svenningsson, A. Intrathecal treatment trial of rituximab in progressive MS: An open-label phase 1b study. Neurology 2018, 91, e1893–e1901. [Google Scholar] [CrossRef]
  107. Topping, J.; Dobson, R.; Lapin, S.; Maslyanskiy, A.; Kropshofer, H.; David, L.; Giovannoni, G.; Evdoshenko, E. The effects of intrathecal rituximab on biomarkers in multiple sclerosis. Mult. Scler. Relat. Disord. 2016, 6, 49–53. [Google Scholar] [CrossRef]
  108. British and Dutch Multiple Sclerosis Azathioprine Trial Group. Double-masked trial of azathioprine in multiple sclerosis. Lancet 1988, 2, 179–183. [Google Scholar]
  109. Kappos, L.; Pätzold, U.; Dommasch, D.; Poser, S.; Haas, J.; Krauseneck, P.; Malin, J.-P.; Fierz, W.; Graffenried, B.U.; Gugerli, U.S. Cyclosporine versus azathioprine in the long-term treatment of multiple sclerosis? results of the german multicenter study. Ann. Neurol. 1988, 23, 56–63. [Google Scholar] [CrossRef]
  110. Uccelli, A.; Capello, E.; Fenoglio, D.; Incagliato, M.; Valbonesi, M.; Mancardi, G.L. Intravenous immunoglobulin, plasmalymphocytapheresis and azathioprine in chronic progressive multiple sclerosis. Neurol. Sci. 1994, 15, 49–53. [Google Scholar] [CrossRef]
  111. Perini, P.; Calabrese, M.; Tiberio, M.; Ranzato, F.; Battistin, L.; Gallo, P. Mitoxantrone versus cyclophosphamide in secondary-progressive multiple sclerosis: A comparative study. J. Neurol. 2006, 253, 1034–1040. [Google Scholar] [CrossRef]
  112. Zephir, H.; De Seze, J.; Duhamel, A.; Debouverie, M.; Hautecoeur, P.; Lebrun, C.; Malíková, I.; Pelletier, J.; Sénéchal, O.; Vermersch, P. Treatment of progressive forms of multiple sclerosis by cyclophosphamide: A cohort study of 490 patients. J. Neurol. Sci. 2004, 218, 73–77. [Google Scholar] [CrossRef]
  113. Brochet, B.; Deloire, M.S.A.; Perez, P.; Loock, T.; Baschet, L.; Debouverie, M.; Pittion, S.; Ouallet, J.-C.; Clavelou, P.; De Seze, J.; et al. Double-Blind Controlled Randomized Trial of Cyclophosphamide versus Methylprednisolone in Secondary Progressive Multiple Sclerosis. PLoS ONE 2017, 12, e0168834. [Google Scholar] [CrossRef]
  114. Weiner, H.L.; Mackin, G.A.; Orav, E.J.; Hafler, D.A.; Dawson, D.M.; Lapierre, Y.; Herndon, R.; Lehrich, J.R.; Hauser, S.L.; Turel, A.; et al. Intermittent cyclophosphamide pulse therapy in progressive multiple sclerosis: Final report of the Northeast Cooperative Multiple Sclerosis Treatment Group. Neurology 1993, 43, 910. [Google Scholar] [CrossRef]
  115. Hommes, O.R.; Sørensen, P.S.; Fazekas, F.; Enriquez, M.M.; Koelmel, H.W.; Fernández, Ó.; Pozzilli, C.; O’Connor, P. Intravenous immunoglobulin in secondary progressive multiple sclerosis: Randomised placebo-controlled trial. Lancet 2004, 364, 1149–1156. [Google Scholar] [CrossRef]
  116. Goodkin, D.; Rudick, R.; Medendorp, S.V.; Daughtry, M.; Van Dyke, C. Low-dose oral methotrexate in chronic progressive multiple sclerosis: Analyses of serial MRIs. Neurology 1996, 47, 1153–1157. [Google Scholar] [CrossRef]
  117. Goodkin, D.E.; Rudick, R.A.; Medendorp, S.V.; Daughtry, M.M.; Schwetz, K.M.; Fischer, J.; Van Dyke, C. Low-dose (7.5 mg) oral methotrexate reduces the rate of progression in chronic progressive multiple sclerosis. Ann. Neurol. 1995, 37, 30–40. [Google Scholar] [CrossRef]
  118. Lugaresi, A.; Caporale, C.; Farina, D.; Marzoli, F.; Bonanni, L.; Muraro, P.A.; De Luca, G.; Iarlori, C.; Gambi, D. Low-dose oral methotrexate treatment in chronic progressive multiple sclerosis. Neurol. Sci. Off. J. Ital. Neurol. Soc. Ital. Soc. Clin. Neurophysiol. 2001, 22, 209–210. [Google Scholar] [CrossRef]
  119. The Multiple Sclerosis Study Group. Efficacy and toxicity of cyclosporine in chronic progressive multiple sclerosis: A randomized, double-blinded, placebo-controlled clinical trial. Ann Neurol. 1990, 27, 591–605. [Google Scholar] [CrossRef]
  120. Fakih, R.; Matiello, M.; Chitnis, T.; Stankiewicz, J.M. Efficacy and safety of mycophenolate mofetil in progressive multiple sclerosis patients. J. Neurol. 2018, 265, 2688–2694. [Google Scholar] [CrossRef]
  121. Barkhof, F.; Giovannoni, G.; Hartung, H.P.; Cree, B.; Uccelli, A.; Sormani, M.P.; Krieger, S.; Uitdehaag, B.; Vollmer, T.; Montalban, X. ARPEGGIO: A randomized, placebo-controlled study to evaluate oral laquinimod in patients with primary progressive multiple sclerosis (PPMS) (P7.210). Neurology 2015, 84, P7.210. [Google Scholar]
  122. Freedman, M.S.; Bar-Or, A.; Oger, J.; Traboulsee, A.; Patry, D.; Young, C.; Olsson, T.; Li, D.; Hartung, H.P.; Krantz, M.; et al. A phase III study evaluating the efficacy safety of MBP8298 in secondary progressive multiple sclerosis. Neurology 2011, 77, 1551–1560. [Google Scholar] [CrossRef]
  123. Deshmukh, V.A.; Tardif, V.; Lyssiotis, C.A.; Green, C.C.; Kerman, B.; Kim, H.J.; Padmanabhan, K.; Swoboda, J.G.; Ahmad, I.; Kondo, T.; et al. A regenerative approach to the treatment of multiple sclerosis. Nature 2013, 502, 327–332. [Google Scholar] [CrossRef] [Green Version]
  124. Mei, F.; Fancy, S.P.J.; Shen, Y.A.; Niu, J.; Zhao, C.; Presley, B.; Miao, E.; Lee, S.; Mayoral, S.R.; Redmond, S.A.; et al. Micropillar arrays as a high-throughput screening platform for therapeutics in multiple sclerosis. Nat. Med. 2014, 20, 954–960. [Google Scholar] [CrossRef] [Green Version]
  125. Green, A.J.; Gelfand, J.M.; Cree, B.A.; Bevan, C.; Boscardin, W.J.; Mei, F.; Inman, J.; Arnow, S.; Devereux, M.; Abounasr, A.; et al. Clemastine fumarate as a remyelinating therapy for multiple sclerosis (ReBUILD): A randomised, controlled, double-blind, crossover trial. Lancet 2017, 390, 2481–2489. [Google Scholar] [CrossRef]
  126. Jepson, S.; Vought, B.; Gross, C.H.; Gan, L.; Austen, D.; Frantz, J.D.; Zwahlen, J.; Lowe, D.; Markland, W.; Krauss, R. LINGO-1, a Transmembrane Signaling Protein, Inhibits Oligodendrocyte Differentiation and Myelination through Intercellular Self-interactions. J. Boil. Chem. 2012, 287, 22184–22195. [Google Scholar] [CrossRef] [Green Version]
  127. Mi, S.; Miller, R.H.; Lee, X.; Scott, M.L.; Shulag-Morskaya, S.; Shao, Z.; Chang, J.; Thill, G.; Levesque, M.; Zhang, M.; et al. LINGO-1 negatively regulates myelination by oligodendrocytes. Nat. Neurosci. 2005, 8, 745–751. [Google Scholar] [CrossRef]
  128. Cadavid, D.; Balcer, L.; Galetta, S.; Aktas, O.; Ziemssen, T.; Vanopdenbosch, L.; Frederiksen, J.; Skeen, M.; Jaffe, G.J.; Butzkueven, H.; et al. Safety and efficacy of opicinumab in acute optic neuritis (RENEW): A randomised, placebo-controlled, phase 2 trial. Lancet Neurol. 2017, 16, 189–199. [Google Scholar] [CrossRef]
  129. Mellion, M.; Edwards, K.R.; Hupperts, R.; Drulović, J.; Montalban, X.; Hartung, H.P.; Brochet, B.; Calabresi, P.A.; Rudick, R.; Ibrahim, A. Efficacy Results from the Phase 2b SYNERGY Study: Treatment of Disabling Multiple Sclerosis with the Anti-LINGO-1 Monoclonal Antibody Opicinumab (S33.004). Neurology 2017, 88, S33.004. [Google Scholar]
  130. .McCroskery, P.; Selmaj, K.; Fernandez, O.; Grimaldi, L.; Silber, E.; Pardo, G.; Freedman, M.S.; Zhang, Y.; Xu, L.; Cadavid, D. Safety and Tolerability of Opicinumab in Relapsing Multiple Sclerosis. Neurology 2017, 88, P5.369. [Google Scholar]
  131. Cho, Y.; Crichlow, G.V.; Vermeire, J.J.; Leng, L.; Du, X.; Hodsdon, M.E.; Bucala, R.; Cappello, M.; Gross, M.; Gaeta, F.; et al. Allosteric inhibition of macrophage migration inhibitory factor revealed by ibudilast. Proc. Natl. Acad. Sci. USA 2010, 107, 11313–11318. [Google Scholar] [CrossRef] [Green Version]
  132. Fox, R.J.; Coffey, C.S.; Conwit, R.; Cudkowicz, M.E.; Gleason, T.; Goodman, A.; Klawiter, E.C.; Matsuda, K.; McGovern, M.; Naismith, R.T.; et al. Phase 2 Trial of Ibudilast in Progressive Multiple Sclerosis. N. Engl. J. Med. 2018, 379, 846–855. [Google Scholar] [CrossRef]
  133. Greenwood, J.; Steinman, L.; Zamvil, S.S. Statin therapy and autoimmune disease: From protein prenylation to immunomodulation. Nat. Rev. Immunol. 2006, 6, 358–370. [Google Scholar] [CrossRef]
  134. Chataway, J.; Schuerer, N.; Alsanousi, A.; Chan, D.; MacManus, D.; Hunter, K.; Anderson, V.; Bangham, C.R.M.; Clegg, S.; Nielsen, C.; et al. Effect of high-dose simvastatin on brain atrophy and disability in secondary progressive multiple sclerosis (MS-STAT): A randomised, placebo-controlled, phase 2 trial. Lancet 2014, 383, 2213–2221. [Google Scholar] [CrossRef]
  135. Rocamonde, B.; Paradells, S.; Barcia, J.; Barcia, C.; García-Verdugo, J.M.; Miranda, M.; Gómez, F.R.; Soria, J. Neuroprotection of lipoic acid treatment promotes angiogenesis and reduces the glial scar formation after brain injury. Neuroscience 2012, 224, 102–115. [Google Scholar] [CrossRef]
  136. Rochette, L.; Ghibu, S.; Richard, C.; Zeller, M.; Cottin, Y.; Vergely, C. Direct and indirect antioxidant properties of alpha-lipoic acid and therapeutic potential. Mol. Nutr. Food Res. 2013, 57, 114–125. [Google Scholar] [CrossRef]
  137. Spain, R.; Powers, K.; Murchison, C.; Heriza, E.; Winges, K.; Yadav, V.; Cameron, M.; Kim, E.; Horak, F.; Simon, J.; et al. Lipoic acid in secondary progressive multiple sclerosis: A randomized controlled pilot trial. Neur. Neuroimmunol. Neuroinflamm. 2017, 4, e374. [Google Scholar] [CrossRef]
  138. Lo, A.C.; Saab, C.Y.; Black, J.A.; Waxman, S.G. Phenytoin Protects Spinal Cord Axons and Preserves Axonal Conduction and Neurological Function in a Model of Neuroinflammation in Vivo. J. Neurophysiol. 2003, 90, 3566–3571. [Google Scholar] [CrossRef]
  139. Raftopoulos, R.; Hickman, S.J.; Toosy, A.; Sharrack, B.; Mallik, S.; Paling, D.; Altmann, D.R.; Yiannakas, M.C.; Malladi, P.; Sheridan, R.; et al. Phenytoin for neuroprotection in patients with acute optic neuritis: A randomised, placebo-controlled, phase 2 trial. Lancet Neurol. 2016, 15, 259–269. [Google Scholar] [CrossRef]
  140. Cohen, J.A. Mesenchymal Stem Cell Transplantation in Multiple Sclerosis. J. Neurol. Sci. 2013, 333, 43–49. [Google Scholar] [CrossRef]
  141. Bonab, M.M.; Yazdanbakhsh, S.; Lotfi, J.; Alimoghaddom, K.; Talebian, F.; Hooshmand, F.; Ghavamzadeh, A.; Nikbin, B. Does mesenchymal stem cell therapy help multiple sclerosis patients? Report of a pilot study. Iran. J. Immunol. IJI 2007, 4, 50–57. [Google Scholar]
  142. Bonab, M.M.; Sahraian, M.A.; Aghsaie, A.; Karvigh, S.A.; Hosseinian, S.M.; Nikbin, B.; Lotfi, J.; Khorramnia, S.; Motamed, M.R.; Togha, M.; et al. Autologous mesenchymal stem cell therapy in progressive multiple sclerosis: An open label study. Curr. Stem Cell Res. Ther. 2012, 7, 407–414. [Google Scholar] [CrossRef]
  143. Yamout, B.; Hourani, R.; Salti, H.; Barada, W.; El-Hajj, T.; Al-Kutoubi, A.; Herlopian, A.; Baz, E.K.; Mahfouz, R.; Khalil-Hamdan, R.; et al. Bone marrow mesenchymal stem cell transplantation in patients with multiple sclerosis: A pilot study. J. Neuroimmunol. 2010, 227, 185–189. [Google Scholar] [CrossRef]
  144. Lublin, F.D.; Bowen, J.D.; Huddlestone, J.; Kremenchutzky, M.; Carpenter, A.; Corboy, J.R.; Freedman, M.S.; Krupp, L.; Paulo, C.; Hariri, R.J.; et al. Human placenta-derived cells (PDA-001) for the treatment of adults with multiple sclerosis: A randomized, placebo-controlled, multiple-dose study. Mult. Scler. Relat. Disord. 2014, 3, 696–704. [Google Scholar] [CrossRef] [Green Version]
  145. Karussis, D.; Karageorgiou, C.; Vaknin-Dembinsky, A.; Gowda-Kurkalli, B.; Gomori, J.M.; Kassis, I.; Bulte, J.W.M.; Petrou, P.; Ben-Hur, T.; Abramsky, O.; et al. Safety and Immunological Effects of Mesenchymal Stem Cell Transplantation in Patients with Multiple Sclerosis and Amyotrophic Lateral Sclerosis. Arch. Neurol. 2010, 67, 1187–1194. [Google Scholar] [CrossRef]
  146. Harris, V.K.; Vyshkina, T.; Sadiq, S.A. Clinical safety of intrathecal administration of mesenchymal stromal cell–derived neural progenitors in multiple sclerosis. Cytotherapy 2016, 18, 1476–1482. [Google Scholar] [CrossRef]
  147. Harris, V.K.; Stark, J.; Vyshkina, T.; Blackshear, L.; Joo, G.; Stefanova, V.; Sara, G.; Sadiq, S.A. Phase I Trial of Intrathecal Mesenchymal Stem Cell-derived Neural Progenitors in Progressive Multiple Sclerosis. EBioMedicine 2018, 29, 23–30. [Google Scholar] [CrossRef] [Green Version]
  148. Li, J.-F.; Zhang, D.-J.; Geng, T.; Chen, L.; Huang, H.; Yin, H.-L.; Zhang, Y.-Z.; Lou, J.-Y.; Cao, B.; Wang, Y.-L. The Potential of Human Umbilical Cord-Derived Mesenchymal Stem Cells as a Novel Cellular Therapy for Multiple Sclerosis. Cell Transplant. 2014, 23, 113–122. [Google Scholar] [CrossRef]
  149. Liang, J.; Zhang, H.; Hua, B.; Wang, H.; Wang, J.; Han, Z.; Sun, L. Allogeneic mesenchymal stem cells transplantation in treatment of multiple sclerosis. Mult. Scler. J. 2009, 15, 644–646. [Google Scholar] [CrossRef]
  150. Cohen, J.A.; Atkins, H.; Banwell, B.; Bar-Or, A.; Bebo, B.; Bowen, J.; Burt, R.; Calabresi, P.; Cohen, J.; Comi, G.; et al. Cell-based therapeutic strategies for multiple sclerosis. Brain 2017, 140, 2776–2796. [Google Scholar] [Green Version]
  151. Uccelli, A.; Brundin, L.; Clanet, M.; Fernandez, O.; Nabavi, S.M.; Muraro, P.A.; Oliveri, R.S.; Radue, E.W.; Sellner, J.; Sorensen, P.S.; et al. MEsenchymal StEm cells for Multiple Sclerosis (MESEMS): A randomized, double blind, cross-over phase I/II clinical trial with autologous mesenchymal stem cells for the therapy of multiple sclerosis. Trials 2019, 20, 263. [Google Scholar] [CrossRef]
  152. Sedel, F.; Bernard, D.; Mock, D.M.; Tourbah, A. Targeting demyelination and virtual hypoxia with high-dose biotin as a treatment for progressive multiple sclerosis. Neuropharmacology 2016, 110, 644–653. [Google Scholar] [CrossRef] [Green Version]
  153. Sedel, F.; Papeix, C.; Bellanger, A.; Touitou, V.; Lebrun-Frenay, C.; Galanaud, D.; Gout, O.; Lyon-Caen, O.; Tourbah, A. High doses of biotin in chronic progressive multiple sclerosis: A pilot study. Mult. Scler. Relat. Disord. 2015, 4, 159–169. [Google Scholar] [CrossRef] [Green Version]
  154. Tourbah, A.; Lebrun-Frenay, C.; Edan, G.; Clanet, M.; Papeix, C.; Vukusic, S.; De Sèze, J.; Debouverie, M.; Gout, O.; Clavelou, P.; et al. MD1003 (high-dose biotin) for the treatment of progressive multiple sclerosis: A randomised, double-blind, placebo-controlled study. Mult. Scler. J. 2016, 22, 1719–1731. [Google Scholar] [CrossRef] [Green Version]
  155. Birnbaum, G.; Stulc, J. High dose biotin as treatment for progressive multiple sclerosis. Mult. Scler. Relat. Disord. 2017, 18, 141–143. [Google Scholar] [CrossRef]
  156. Baldassari, L.E.; Feng, J.; Clayton, B.L.; Oh, S.-H.; Sakaie, K.; Tesar, P.J.; Wang, Y.; Cohen, J.A. Developing therapeutic strategies to promote myelin repair in multiple sclerosis. Expert Rev. Neurother. 2019, 1–17. [Google Scholar] [CrossRef]
  157. Najm, F.J.; Madhavan, M.; Zaremba, A.; Shick, E.; Karl, R.T.; Factor, D.C.; Miller, T.E.; Nevin, Z.S.; Kantor, C.; Sargent, A.; et al. Drug-based modulation of endogenous stem cells promotes functional remyelination in vivo. Nature 2015, 522, 216–220. [Google Scholar] [CrossRef] [Green Version]
  158. Cohen, J.A.; Baldassari, L.E.; Atkins, H.L.; Bowen, J.D.; Bredeson, C.; Carpenter, P.A.; Corboy, J.R.; Freedman, M.S.; Griffith, L.M.; Lowsky, R.; et al. Autologous Hematopoietic Cell Transplantation for Treatment-Refractory Relapsing Multiple Sclerosis: Position Statement from the American Society for Blood and Marrow Transplantation. Boil. Blood Marrow Transplant. 2019, 25, 845–854. [Google Scholar] [CrossRef] [Green Version]
  159. Marrie, R.A.; Hanwell, H. General health issues in multiple sclerosis: Comorbidities, secondary conditions, and health behaviors. Continuum 2013, 19, 1046–1057. [Google Scholar] [CrossRef]
  160. Ontaneda, D. Inadequate outcome measures are the biggest impediment to successful clinical trials in progressive MS-Commentary. Mult. Scler. 2017, 23, 508–509. [Google Scholar] [CrossRef]
  161. Giovannoni, G.; Airas, L.; Bove, R.; Boyko, A.; Cutter, G.; Hobart, J.; Kuhle, J.; Oh, J.; Tur, C.; Garas, M.; et al. Ocrelizumab Treatment Effect on Upper Limb Function in PPMS Patients with Disability: Subgroup Results of the ORATORIO Study to Inform the ORATORIO-HAND Study Design; ECTRIMS Online Library: London, UK, 2018; p. 619. [Google Scholar]
  162. Sumowski, J.F.; Benedict, R.; Enzinger, C.; Filippi, M.; Geurts, J.J.; Hamalainen, P.; Hulst, H.; Inglese, M.; Leavitt, V.M.; Rocca, M.A.; et al. Cognition in multiple sclerosis State of the field and priorities for the future. Neurology 2018, 90, 278–288. [Google Scholar] [CrossRef]
Figure 1. Clinical course of progressive multiple sclerosis. The orange star indicates the presence of radiological activity (new/enlarging T2 lesions or gadolinium enhancing lesions).
Figure 1. Clinical course of progressive multiple sclerosis. The orange star indicates the presence of radiological activity (new/enlarging T2 lesions or gadolinium enhancing lesions).
Biomedicines 07 00056 g001
Table 1. Potential remyelinating and neuroprotective molecules studied in multiple sclerosis.
Table 1. Potential remyelinating and neuroprotective molecules studied in multiple sclerosis.
TherapyPotential Mechanism of ActionTrialsPrimary EndpointResults
Remyelination strategies
Clemastine fumarateFirst-generation anti-histamine, promotes remyelination and oligodendrocyte differentiation via anti-muscarinic effect [123,124]ReBUILD [125]Shortening of P100 latency delay on visual-evoked potentials at 150 daysImprovement in P100 latency of 1.7 ms/eye (95% CI 0.5 to 2.9, p = 0.0048) with clemastine
OpicinumabAnti-LINGO-1 antibody, promotes remyelination and oligodendrocyte differentiation via blocking of inhibitory adhesion molecule [126,127]RENEW [128] (in acute unilateral optic neuritis)24-week change in optic nerve conduction latency using full-field visual evoked potentialNon-significant trend towards improvement in the intention-to-treat analysis, modest but significant benefit at week 32 in the per-protocol analysis
SYNERGY [129,130] (in RRMS and SPMS with active disease)Percentage of participants with ≥3 month confirmed improvement of composite endpoint (EDSS, T25FW, 9HPT, PASAT) over 72 weeksBenefit seen in those receiving the 30 mg/kg dose.
Neuroprotection strategies
IbudilastPhosphodiesterase-inhibitor, inhibits macrophage migration inhibitory factor, and toll-like receptor 4 [131]SPRINT-MS [132]Progression of whole brain atrophy over 96 weeks48% slowing in the rate of atrophy progression with ibudilast compared to placebo
SimvastatinHMG-CoA reductase inhibitor, inhibit MHCII-restricted antigen presentation, shifts cytokine production from a pro- to an anti-inflammatory response, decreases T-cell proliferation [133]MS-STAT [134]Progression of whole-brain atrophy, change in EDSS and total MS Impact Scale-29 at 24 monthsDecrease in annualized rate of whole brain atrophy compared to placebo, benefit on EDSS and MS Impact Scale-29 as well
Lipoic acidEndogenous antioxidant, various potential mechanisms, including free radical scavenging, oxidative damage repair, downregulation of inflammatory cytokines, T-cell migration in CNS inhibition [135,136]Spain et al. [137].Annual percent change of brain volume68% reduction in the rate of brain atrophy compared to placebo over 24 months
PhenytoinSelective sodium-channel inhibitor, reverses sodium influx, which drives calcium influx via reverse operation of the sodium/calcium exchanger after axonal injury [138]Raftopoulos et al. [139].RNFL thickness in the affected eye30% reduction in the extent of RNFL loss with phenytoin compared with placebo at 6 months
Mesenchymal stem cells *Pluripotent non-hematopoietic precursor cells (isolated from bone marrow or adipose tissue), release of soluble trophic factors that promote intrinsic tissue repair mechanisms [140]Multiple small clinical trials and open label studies using variable route of administration and dosing regimens [141,142,143,144,145,146,147,148,149]Variable endpoints depending on trialGood safety and tolerability, efficacy not yet established [150]
Phase II randomized, double-blind trial, MESEMS (NCT01854957) [151]Safety, reduction in the total number of contrast-gadolinium enhancing lesionsOngoing
Open-label study, MSC-NTF Cells (NCT03799718)Safety, T25FW change from baseline, changes in neurotrophic factorsOngoing
High-dose biotin (MD 1003) *Essential co-factor for five carboxylases involved in fatty acid synthesis and energy production, promotes remyelination, and reduces axonal hypoxia [152]Sedel et al. (pilot study) [153]Shortening of P100 latency on visual-evoked potentialsImprovement or normalization of P100 latency
Tourbah et al. (randomized, double-blind placebo-controlled trial) [154]Proportion of patients with disability reversal on EDSS or T25FW at month 9, confirmed at month 122.6% of treated patients achieved the primary endpoint versus none of the placebo-treated patients (p = 0.005)
Birnbaum et al. (open-label study of compound medication, not MD 1003) [155]EDSS worsening or improvement while on treatment (3 to 12 months)No benefits observed
* possibly dual effect on remyelination and neuroprotection. 9HPT: 9-hole-peg test, EDSS: Expanded disability status scale, LINGO-1: Leucine-rich repeat and immunoglobulin domain-containing neurite outgrowth inhibitor receptor interacting protein-1, PASAT: Paced auditory serial addition test, RRMS: Relapsing–remitting multiple sclerosis, RNFL: Retinal nerve fiber layer, SPMS: Secondary progressive multiple sclerosis, T25FW: Timed-25-foot walk.

Share and Cite

MDPI and ACS Style

Macaron, G.; Ontaneda, D. Diagnosis and Management of Progressive Multiple Sclerosis. Biomedicines 2019, 7, 56. https://doi.org/10.3390/biomedicines7030056

AMA Style

Macaron G, Ontaneda D. Diagnosis and Management of Progressive Multiple Sclerosis. Biomedicines. 2019; 7(3):56. https://doi.org/10.3390/biomedicines7030056

Chicago/Turabian Style

Macaron, Gabrielle, and Daniel Ontaneda. 2019. "Diagnosis and Management of Progressive Multiple Sclerosis" Biomedicines 7, no. 3: 56. https://doi.org/10.3390/biomedicines7030056

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop