Trending Topic

Human brain left x-ray view
17 mins

Trending Topic

Developed by Touch
Mark CompleteCompleted
BookmarkBookmarked

Parkinson’s disease (PD) is a complex neurodegenerative condition that predominantly affects older people, with a rising prevalence worldwide.1,2 There are many on-going challenges and unmet needs in PD: difficulties in making an accurate diagnosis (particularly in the early stages of the disease), troubling side effects associated with the available pharmacological treatments, a lack of effective disease-modifying therapies […]

Multiple Sclerosis Management – A Changing Landscape 2013

Per Soelberg Sørensen, Massimo Filippi, Wolfgang Brück
Share
Facebook
X (formerly Twitter)
LinkedIn
Via Email
Mark CompleteCompleted
BookmarkBookmarked
Copy LinkLink Copied
Download as PDF
Published Online: Jan 27th 2013 European Neurological Review, 2013;8(2):105–114 DOI: http://doi.org/10.17925/ENR.2013.08.02.105
Select a Section…
1

Abstract

Overview

The aims of this educational meeting, held in Vienna, Austria, were to explore the significant advances that have occurred in multiple sclerosis (MS) management over the past two decades, to highlight modern-day perspectives and challenges and to consider the impact of the new oral first-line treatments expected to enter the MS market shortly. The meeting was attended by 372 delegates with neurological interests from 30 countries and was opened by Per Soelberg Sørensen (Copenhagen, Denmark).

Keywords

Multiple sclerosis, magnetic resonance imaging, pathophysiology, role of injectable agents, oral agents, individualised therapy

2

Article

Proceedings of the Meeting: Multiple Sclerosis Management – A Changing Landscape 2013, held on 26–27 April 2013 in Vienna, Austria

Multiple Sclerosis Management 2013
The keynote lecture was given by Fred D Lublin (New York, New York, US), who outlined present and future challenges in the treatment of multiple sclerosis (MS). His theme was: where are we with current MS treatments and at what stage should patients be treated?

There are now 10 marketed disease-modifying agents with seven different modes of action (all anti-inflammatory) for MS treatment and more have been submitted for regulatory approval.1–4 Most of these treatments focus on clinically isolated syndrome (CIS) and relapsing remitting MS (RRMS).5

The biggest challenge in MS treatment is progressive disease: the majority of current treatments are approved for RRMS and are much less effective in progressive disease.6 Consequently, many patients with secondary progressive MS (SPMS) feel abandoned. The most valid treatment strategy currently is to treat early and delay or prevent SPMS.7 There is considerable research effort in progress to address the progressive stage of the disease, but repairing damaged or lost axons is challenging. Therefore, despite the ongoing emergence of new agents, there remain many unmet treatment needs in MS therapy.8

An important question in MS is which patients should be treated and when? Some studies have suggested that certain immunophenotypes and pathophenotypes predispose MS.9,10 The change in the Macdonald criteria, including dissemination of MRI lesions in time and space, has increased diagnostic sensitivity and specificity11 and this has helped determine which patients are at risk and need to start treatment to inhibit or prevent accumulating neurological damage.

In clinical terms, MS progresses in steps of incomplete recovery leading to gradual worsening. In pathological terms, neuronal inflammatory disease leads to degeneration. This progression is driven by exacerbations12 and reducing their incidence through disease-modifying treatment (DMT) has advantageous long-term consequences.

To better reduce relapses and inhibit progression, some investigators have tried concomitant use of first-line DMTs in MS. The CombiRX study was an example of this approach. Sponsored by the US National Institutes of Health (NIH), the CombiRx study, a phase III, three-year-long study included 1,008 patients with RRMS and combined both glatiramer acetate (GA) treatment and interferon beta-1a (IFNb-1a) compared with these agents given separately.13,14 In all of the three different definitions of exacerbations used in the CombiRx trial, GA monotherapy showed a significantly greater reduction on relapse rate compared with IFNb-1a treatment. In the efficacy head-to-head trial arm, GA monotherapy was superior to IFNb-1a in reducing the risk of exacerbation. The combination did show advantages in MRI metrics and, intriguingly, patients with an expanded disability status scale (EDSS) of 0 were three to five times more likely to progress than those with an EDSS >0.

A problem in MS is how to assess the treatment efficacy. Better measures are needed in clinical trials to define exacerbations and relapses, which vary between trials.15 The ultimate goal in MS treatment is reducing disability progression, however, there is disagreement and inconsistency over disability outcomes measures (EDSS is not sensitive enough) and time to clinically definite MS (CDMS) is variable.16 There is also no agreement over diagnostic approaches including: biomarkers, genomics, gene expression models, epigenomics, proteomics, advanced magnetic resonance imaging (MRI) metrics and clinical MRI biomarker profiles.17–20

The recent approval of several effective treatments in MS (including oral agents) and the imminent approval of more have increased the exciting potential of personalised medicine in MS in the near future.17,21 With the entry of a number of first-line oral treatments for RRMS expected to enter the market, there is a need for a structured approach. The next steps in the development of such an approach are to: determine who will respond to which agent and, after starting treatment, identify responders and nonresponders. It will also be necessary to create an updated MS classification paradigm (radiologically isolated syndrome [RIS], CIS, RRMS), whether there is full or stepwise recovery from relapse and the change in annualised relapse rates (ARR) over time.

There are, however, challenges for the evaluation of future treatments in MS. Disease progression is not inevitable and up to a third of patients do not worsen, which is a problem when evaluating the effectiveness of new treatments in clinical trials.22 Other concerns include the design of the trial (head-to-head versus observational studies), choice of therapies, dosing frequencies, parameters of the disease to monitor, how to compare groups (what statistical inferences to use), identifying and controlling bias and ensuring sufficient statistical power.23,24 In such trials, the criteria for choosing therapies and monitoring response should include MRI lesion load, disease activity and increasing use of biomarkers e.g. immune factors such as interleukin (IL)-17, IL-21 and genetic/genomic markers.

With an increasing choice of effective drugs becoming available and treatments starting earlier, the ‘disease-free’ concept in MS in which patients have no relapses, progression or gadolinium (Gd)-enhancing lesions, is now becoming a usable measure in assessing therapies.25 Future studies may increasingly measure this as a pre-planned endpoint.

Overall, MS is, to some extent, treatable. The therapies given in MS have manifold objectives in aiming to: reduce relapse rates, slow disability progressions, modify the disease, treat acute exacerbations, provide enhanced recovery and function, treat symptoms, provide neuroprotection, enable repair and improve quality of life. The current DMTs do not achieve all these goals but they do alleviate symptoms or enhance function and delay progression. Some therapies are also becoming available that enhance repair. The prognosis for the newly diagnosed patient with MS therefore has substantially improved in recent years and the release of new treatments will make further improvements possible.

Twenty Years of Progress in Multiple Sclerosis

Second-line Options for Multiple Sclerosis Following First-line Treatment Failure
Bernd C Kieseier (Dusseldorf, Germany) outlined the lack of good evidence and guidelines in circulation considering second-line therapies after firstline treatment failure in MS and stressed the need to critically reassess the practice of monitoring disease progression and activity in MS patients.

When first-line treatment is apparently ineffective, it is important to determine whether the failure is transient or permanent. To assess this, it is necessary to have a robust definition of failure (such as increase of 1 point on EDSS scale): currently not available. In addition, there is no consensus to guide treatment of patients with first-line treatment failure.26–29

There is also a general lack of class 1 evidence supporting the use of alternatives, such as fingolimod, mitoxantrone and natalizumab. It is not possible to decide whether these are more effective than ‘platform therapy’ (IFNb or GA). The pivotal studies for the assessment of fingolimod, mitoxantrone and natalizumab in MS (FREEDOMS, MIMS and AFFIRM studies) used patients with highly active disease, not those who had experienced treatment failure30–32 and, as a result, it is not possible to determine how well these treatments function when switching to second-line therapy.

As a result of cost constraints, current treatment algorithms in MS are driven by medical authorities, not by patients, and this restricts treatments that can be offered, especially when a switch is needed to more-effective treatment. In second-line therapy it would be valuable to identify which patients are potential responders to target treatment and avoid giving inappropriate medications.

Second-line therapy has shown superior efficacy over first line. An example is the TRANSFORMS study (1,292 patients) with RRMS who were treated with 1.25 mg or 0.5 mg fingolimod/day or 30 μg IFNb-1a intramuscular/ week.33 There was a significant reduction in ARR for both fingolimod doses (p>0.001) and improvements in MRI findings. In an extension study, switching patients previously treated with IFNb-1a to fingolimod produced significant efficacy improvements. There are, however, safety concerns with second-line drugs. Both binding and neutralising antibodies (NAbs) have been reported,34 which can reduce long-term efficacy. Serious adverse events (SAEs), particularly progressive multifocal leukoencephalopathy (PML), are well known with natalizumab and to be anticipated.35 The risk of John Cunningham (JC) virus infection with natalizumab can be stratified since the virus is present in 50–60 % of population.36 Adverse events (AEs) associated with fingolimod (cardiac effects, infection and macular oedema37) and with mitoxantrone (amenorrhoea, nausea and vomiting, alopecia and urinary tract infections38) are difficult to predict and long-term safety data is limited. Some potential markers for fingolimod efficacy have been proposed, in particular, L-selectin (CD62L) (Schwab et al. in press), but there is a lack of surrogate markers in MS and a lack of prognostic factors. There is, therefore, an urgent clinical need to further investigate escalating therapy in MS. Switching to alternative medications may reduce disease activity but patient-specific factors and the risk–benefit profile of the new drug must be considered. Neurologists have traditionally been slow to switch treatments in MS patients, but this is improving.

Benefit–Risk of Injectable Multiple Sclerosis Therapies
The injectable therapies in MS have been available for 20 years and much experience on their efficacy and safety has been gained during that time. Franz Fazekas (Graz, Austria) provided his impressions and thoughts on these treatments, noting that their efficacy ranges from modest to good when started early. Overall, ARR from a systematic review of multiple clinical trials using IFNb-1a intramuscular or subcutaneous, IFNb-1b subcutaneous and GA range from 1.43–1.93 versus 2.32 for placebo.39 Similar reductions are seen in assessments of disability.

Axonal loss is a major pathological process that is responsible for irreversible neurological disability in patients with MS40 – surrogate markers are needed to monitor this loss and early therapy should be initiated before it has become extensive. Several large studies have emphasised the importance of the early commencement of various treatments. These include the CHAMPS study for IFNb-1a intramuscular, the long-running BENEFIT study for IFNb-1a41 and the PreCISE trial for GA.42

Injectable IFNb-1a therapies are associated with flu-like symptoms in 50 % of patients, skin reactions in 26 % and administration site reactions in 10 %,43 but these tend to diminish with time and generally, early treatment does not compromise well-being.44 Injectable IFNb-1a treatments can also result in Nabs that reduce efficacy in some cases. Screening for these should be integrated with routine clinical and imaging indicators to guide treatment decisions.

The long-term use of injectable therapy has proved effective and safe.41,45 The optimum duration of injectable therapy, however, is unclear. It has demonstrated effectiveness over many years but after 10 years or more the therapy should be stopped if the patient requests it. In many cases, the efficacy range of injectable therapy is too limited and treatment escalation to a more effective medication is necessary. When side effects occur, treatments should either be stopped or de-escalated.46,47

Various head-to-head studies have compared the relative efficacies of injectable treatments. A comparison of IFNb-1a intramuscular 30 μg versus IFNb-1a subcutaneous 44 μg in the EVIDENCE trial showed fewer relapses with IFNb-1a subcutaneous.48 The INCOMIN trial compared IFNb1-a intramuscular 30 μg with IFNb-1a subcutaneous 8 MIU and showed similar efficacy in both.49 A comparison of IFNb-1b intramuscular 30 μg versus 60 μg showed that there was no efficacy difference between doses of this weekly intramuscular therapy.50 More recently, the CombiRx study (discussed above) showed that a combination of both IFNb-1a intramuscular 30 μg and GA 20 mg showed no clinical benefit but that GA alone was superior to IFNb alone in reducing exacerbation risk.13 In addition, the REGARD trial, a head-to-head clinical trial of GA mg and IFNb-1a 44 μg found no significant difference in relapse rates in response to these treatments (hazard ratio 0.943; p=0.643 for the difference in time-to-first relapse).51

Injectable MS therapies are a valuable option in RRMS. Their long-term use is safe despite some inconveniences and associated AEs. It is likely that these therapies will remain in use for a long time; neurologists understand how they work and how to use them. New oral and other agents may eventually replace the current injectables but safety profiles must be considered first.

Understanding and Communicating Risks and Benefits in Chronic Diseases
Angela Fagerlin (Ann Arbor, Michigan, US) delivered the guest lecture that explored problems in communicating risks and benefits of treatments to patients with MS. She stressed that in terms of patient communication it is not what you say but the way you say it. In MS and many other diseases it is important that the patient is given all the information and that decision-making is shared as preference-sensitive decision-making brings significant health benefits.52 When discussing treatments with patients, it is important to consider literacy and numeracy.

Graphical formats are useful in changing patient perception but the appropriate type of graph has to be selected; creating educational materials (e.g. decision aids) or using decision-coaching methods can improve patient decision-making. Overall, physicians should avoid giving too much information to patients at any one time and follow the principle that ‘less is more’.

Emerging Insights into Disease Pathophysiology from Studies of Primary Progressive, Secondary Progressive and Progressive Relapsing Multiple Sclerosis
The state of knowledge around the more progressive forms of MS was considered by Hans Lassmann (Vienna, Austria), who reminded the audience that there are two established views of MS pathology: the plaque-centred view,53 which is over 100 years old, and the immunological view,54 which is based on encephalomyelitis models. MS is an inflammatory disease but it is different from that seen in the autoimmune encephalitis models;55 in progressive disease patient response differs to that during relapsing disease.

The pathological features of MS change over the course of the disease. In the early stages there are predominantly focal lesions in the white matter but in progressive stages there is more cortical and diffuse white matter injury.56 Inflammatory processes are more pronounced in acute and relapsing stages and in the progressive stage. However, it is not clear why these not seen on MRI and why treatments are less effective in progressive disease. In progressive forms the inflammation is increasingly trapped within the central nervous system and drugs have to pass the blood–brain barrier (BBB) and are less effective.

Insights and Challenges for Modern Multiple Sclerosis Management
Assessing Disease Progression – Physician and Patient Perspectives
Alan Thompson (London, UK) described the current focus on progressive disease as “very key”. Of the MS patient population, 55–60 % have progressive disease57 and feel neglected; they sense that as most drug treatments are for RRMS and not for progressive disease. In progressive MS, 44 % of patients want disease stabilisation; 18 % want recovery.

Progressive MS presents several challenges. First, there is no agreed definition: in clinical terms it is accumulation of disability; in MRI terms it is increased number of lesions; and in pathological terms it is abundant axonal damage and atrophy. It can be assessed from patient and physician perspectives, both of which can be measured scientifically using Short Form (SF)-3658 and other scales. Measuring disease is another factor: measures range from the medical model to the psychosocial model (the US Food and Drug Administration [FDA] recommends that patient-related outcomes should be increasingly used in clinical trials). There is a need to identify the concept and the framework of what is being measured, but a stronger underpinning of outcomes in progressive MS is required. Measuring the impact and influence of interactions is problematic: measures need to be robust and responsive (many current measures are not). Patient-reported outcomes need to be valid and should be qualitative and quantitative, such as the Fatigue Impact Scale.59 Therefore, a clearer understanding of which variables are progressing and responder analyses are needed to properly assess progression.

The MS Outcomes Assessment Consortium (MSOAC) is developing new clinical outcome measures of indicating disability in MS, using seven clinical trial datasets. This includes the Critical Path Institute (‘C-Path’) who operate under the auspices of the FDA.60 The Progressive MS Collaborative aims to expedite treatments for progressive MS and has identified five roadblocks to treatment.

Advanced Magnetic Resonance Imaging Techniques
Recent advances in MRI techniques and their implications on understanding the pathology of MS were discussed by Massimo Filippi (Milan, Italy). Over the course of MS the pathological profile changes (see Figure 1).61–63 In progression, there is an increased heterogeneity in white matter lesions and more centrifugal and centripetal lesions.64,65 In addition, cortical lesions66,67 are strongly predictive of disease status and tend to be more apparent later in the disease course. Diffuse grey matter damage has been shown to correlate with EDSS increase (69 %) and cognitive deterioration (97 %).68,69

Multiparametric MRI approaches have shown that the brain can compensate for damage in MS using cognitive reserves;70–72 patients with higher brain or cognitive reserves fare better. Functional MRI approaches have revealed spinal cord changes in MS that are different in PPMS compared with SPMS;73 they have also revealed cortical reorganisation74 and general disorganisation that is related to cognition.75

MS involves a complex balance between tissue damage, repair and cortical reorganisation accompanied by increasing structural destruction in the brain and spinal cord. Newer MRI techniques have enabled the specific detection of different pathologies in MS and these show better correlation with disease course than conventional MRI.

The Advent of Oral Multiple Sclerosis Agents
Mechanism of Action of Oral Agents for Multiple Sclerosis
Wolfgang Brück (Gottingen, Germany) assessed the recently introduced and emerging oral agents for MS that are significantly changing the treatment landscape. Most oral drugs are small molecules that can cross the BBB into the CNS. Drugs that are effective once they enter CNS, however, are currently lacking. The first approved oral agent in MS, fingolimod, is a modulator of sphingosine-1 phosphate (S1P) receptors that are expressed at various sites in the CNS (including neurons and glia).76,77 This action prevents lymphocytes from exiting lymph nodes and thus inhibits inflammatory processes. Fingolimod also suppresses peripheral lymphocyte activity. The cuprizone experimental model has shown that S1P receptors are involved in controlling response to injury and that fingolimod may contribute to this.78

Another approved oral agent for use in RRMS, teriflunomide, is the active metabolite of the prodrug leflunomide that inhibits nucleotide synthesis by blocking dihydroorotate dehydrogenase. It is not a selective agent and affects all rapidly proliferating cells. Teriflunomide is believed to have the potential advantage of not increasing the risk of infection (as with other MS agents) due to its limited effects on the immune system.79 BG-12 dimethyl fumarate was also recently approved for use in RRMS, it is believed to cause glutathione depletion leading to induction of the anti-inflammatory stress protein HO-1 and increased secretion of nuclear factor (Nrf2) followed by an antioxidant response. These effects induce type II dendritic cells and anti-proliferative effects.80,81 Recent data suggest that BG-12 also acts as an antioxidant and improves mitochondrial function in diseased brain tissue.82

Laquinimod, quinolone 3-carboxamide, is in late-stage development and is believed to act against MS by inhibiting both Th1 and Th17 responses83 and switching a pro-inflammatory to an anti-inflammatory response. It also prevents T-cells from entering the CNS. Laquinimod affects antigen presentation, decreases IL-17 production but increases levels of protective proteins in the brain. This drug reduces activation of astrogliosis and inhibits NFKB gene transcription resulting in reduced demyelination and axonal damage. Laquinimod has a clear effect on both peripheral and CNS immune cells that is dose-dependent.

The magnitude of the effect of these four oral agents in the CNS and peripherally varies, as shown in Table 1. Dr Brück raised the intriguing possibility that these new oral drugs increase the possibility of polytherapy in MS. A medication that has a peripheral effect may be combined with one that has a neuroprotective effect in the CNS to create a combination that has complementary properties and possibly greater efficacy.

Recent Clinical Investigations with New Oral Agents for Multiple Sclerosis
The clinical trial data supporting fingolimod, BG-12, teriflunomide and laquinimod were outlined by Giancarlo Comi (Milan, Italy). Fingolimod has shown notable efficacy advantages in recent clinical trials. In the FREEDOMS trial, fingolimod (0.5 mg versus 1.25 mg versus placebo in 1,272 patients with RRMS) significantly reduced the ARR versus placebo (p<0.001) and significantly reduced the risk of disability progression (p=0.02) over the 24-month period.84 Over 2 years of treatment, fingolimod significantly reduced the overall rate of brain atrophy by 36 % compared with placebo (–0.84 % versus –1.31 %).85,86 In this study, fingolimod also showed greater reductions in T2 lesion numbers and Gd-enhancing lesions but similar reductions in brain volume compared with IFNb-1a as previously used in the REFORMS trial (see Table 2). In the FREEDOMS II trial (fingolimod 0.5 mg and 1.25 mg versus placebo in 1,083 patients with RRMS), fingolimod reduced long-term ARR by approximately 50 % compared with placebo but had little effect on disability.87 AEs associated with fingolimod include: bradycardia, macular oedema, elevated blood pressure (BP), liver enzyme increase, risks to pregnancy and infection (should vaccinate against varicella zoster virus [VZV]).

BG-12 has also shown impressive efficacy performance in pivotal clinical trials. The DEFINE study showed that the ARR was reduced by approximately 50 % with BG-12 versus placebo and produced a large reduction in MRI activity.88 The CONFIRM study showed ARR reductions of 44 % and 50.5 % for BID and TID for BG-12 but 28.6 % for the comparator, GA. It also showed improved times to disability progression (21 %, 24 % and 7 %) and new or enlarging T2 lesions (71 %, 73 % and 54 %).89 Common AEs with BG-12 include flushing, diarrhoea, nausea, upper respiratory tract infection (URTI), abdominal pain and proteinuria. In addition a few cases of PML have also been reported.

Teriflunomide has been evaluated in various clinical trials (TEMSO, TOWER, TENERE, TOPIC, TERACLES). The TEMSO study showed that teriflunomide 7 mg or 14 mg/day produced 31.2 % and 31.5 % ARR relative risk reductions versus placebo (p<0.001 for both).90 This study also showed that teriflunomide significantly reduced disability progression (at the higher dose), and MRI evidence of disease activity compared with placebo. In the TOWER study there were 36.3 % (p<0.0001) and 22.3 % (p=0.02) reductions in ARR for the 14 mg and 7 mg/day doses of teriflunomide, respectively.91 AEs included: diarrhoea, nausea, hair thinning, elevated liver enzymes and serious infections. Teriflunomide should not be used in pregnancy.90–92 Using the higher dose in the TENERE study (teriflunomide 14 mg and 7 mg versus IFNb-1a subcutaneous 44 μg) has shown no effect on disability.93

Professor Comi described laquinimod as having an unusual mode of action with manifold pharmacodynamic activities in MS and has been evaluated in a series of clinical trials (ALLEGRO, BRAVO and CONCERTO). Pooled analyses of ARR from ALLEGRO and BRAVO showed that laquinimod reduced the relapse rate by 21.4 % (p=0.0005).94 These analyses also showed that the risk of confirmed disability progression sustained for 3 months was significantly reduced with laquinimod compared with placebo, 34.2 % (p=0.0017) and was 46 % in 6 months (p<0.0001). Such reductions have only been previously seen with alemtuzumab.94 MRI findings in these trials showed a 30 % reduction in Gd-enhancing lesions and 30 % reduction in brain volume loss for laquinimod versus placebo. The data suggest that laquinimod offers unique advantages in MS, with a favourable safety profile (only elevations in liver enzymes reported as AEs), and good efficacy in terms of brain atrophy and disability progression (see Figure 2). Overall, the new oral agents in MS have shown encouraging efficacy in clinical trials, but there remains a need for a structured approach to determine their use in the in regular clinical setting.

Prospects for Individualised Therapy for Multiple Sclerosis
The introduction of several new oral agents and the impending introduction of several more have increased the choice of MS therapies available to neurologists. Patients respond differently to MS therapies and the drugs are suited to different disease phases of the disease and/ or patient status. Gavin Giovannoni (London, UK) wondered whether it is time to consider individualised therapy in MS. A major aspect of this is who should decide whether to treat early and actively? To answer this, a series of questions were put to patients with MS at St Bartholomews Hospital, London, UK to assess their attitudes to treatment:

    Would you choose aggressive first-line treatment over safer firstline treatments? yes: 63 %, maybe: 25 %, no: 11 %. Who should make the decision on accessibility of treatments to earlyphase patients? Regulators: 5 %, payers (governments, insurance companies): 0 %, neurologists: 44 %, patients: 41 %, other: 11 %. What chance of a serious life-threatening AE would you accept as a complication of early aggressive treatment? Risk of 0.01 %: 20 %; risk of 0.1 %: 33 %; risk of 1 %: 20 %.

It is now well-recognised that delaying active treatment can hasten disease progression compared with early intervention, but such a strategy is not always provided in many territories. The UK NHS ‘doughnut’ model of treatment shows that more aggressive treatments are reserved for more active disease and this is decided on an institutional or health authority basis (see Figure 3). Dr Giovannoni suggested that all treatments should be available from the start and the patients, guided by neurologists, should decide which was appropriate for their individual needs. Experience from recent alemtuzumab trials shows that this therapy is more effective when started early95,96 (see Figure 4) and that alemtuzumab decreases relapse rates and decreases disability progression to a greater extent than IFNb-1a. This raises the question: is it fair or ethical to make MS patients wait 20 years for the outcome of an experiment?

Studies have shown that relapses,97,98 MRI activity,99,100 and disease progression101,102 are all significant and are predictive of greater disease activity. Treatment with DMTs decreases disease activity and improves outcomes. With greater treatment success, an acceptable definition of a cure in MS is needed. The terms ‘no evidence of detectable disease’ (NEDD), ‘treat-to-target’ (T2T) and ‘disease-activityfree’ (DAF) are entering the lexicon. With improved diagnostic methods and an increasing selection of drug therapies, Dr Giovannoni left the audience with the question: ‘have we finally entered the era of individualised therapy for MS?’

Workshop Sessions
A series of 10 workshops tackled various aspects of MS and treatment. These were divided into the themes of symptomatic treatment, management of special populations, treatment concerns and pathology. An overview of the sessions and main points is given in Table 3.

Debate – With the Introduction of Oral Agents, Injectables Will Have No Place in the Modern Management of Multiple Sclerosis
The meeting finished with a timely debate about whether new oral treatments would oust the pre-existing injectable therapies in MS management. Óscar Fernandez (Malaga, Spain) argued in favour.

Argument in Favour
Over the past 20 years DMTs have had variable success in MS. However, the new orals have improved efficacy and safety and their introduction has started a new era in MS management. The timeline of MS treatments shows that the 1990s saw the introduction of IFNb-1b, IFNb-1a, the 2000s saw the introduction of GA, mitoxantrone and natalizumab and the 2010s are seeing the introduction of oral agents: fingolimod, teriflunomide, BG-12 and laquinimod. Other new treatments are likely to follow.

The efficacy of current first-line injectables is about 30 %.14,84 The efficacy, safety and tolerability of the oral agents compare well with those of the injectables or improve on it (see Table 4). Safety concerns of injectables include injection site reactions, flu-like symptoms (IFNb), subcutaneous lipoatrophy, myalgia, depression and chest tightness (GA). In clinical studies, up to 40 % of patients discontinue within 2 years and 32 % of patients will have ≥1 injection-related reaction.103 In addition, one-third of the reasons for missing doses was the injection itself and only 4 % had a medication possession ratio >85 % (time a patient has access to medication), in a range of 72–76 %.104

Despite the availability of automatic injection devices that make administration easier and less traumatic, among patients who had received 5 years of treatment, 100 % said they would prefer an oral treatment. Oral therapies increase the possibility of personalised treatment and treatment combinations. Dr Fernandez conceded that despite the obvious advantages of oral therapy, injectable treatments were likely to remain in use for some time.

Argument Against
Per Soelberg Sørensen (Copenhagen, Denmark) countered these arguments and suggested that there was life left in injectable therapies in MS for the foreseeable future. There are currently two types of injectables (IFNb and GA) and there will soon be four types of oral agents. Some patients have an excellent response to injectable therapies and there is consequently no need to transfer them to less well-known treatments. With the existing injectable treatments, 30 % show an excellent response, 40 % have a moderate response and only 30 % have an unsatisfactory response. Future treatment approaches may differ between patients who are established on a therapy and those who have not been treated; switching them may not be justified if they are well controlled.

The advantage of injectable therapies in MS was demonstrated by the increases in disease activity-free patients being better for IFNb-1a and natalizumab in the PRISMS and AFFIRM studies than for fingolimod and cladribine in the FREEDOMS and CLARITY studies (see Figure 5).100,105,106 IFNb has been used in MS for over 20 years and there are millions of years of patient observation and experience supporting its use. Flu-like symptoms and injection site reactions decrease with time and few are severe. There is only a modest increase in liver enzymes and some NAbs. Furthermore, the efficacy of IFNbs can be increased with add-on therapies and PEGylation.

GA has been used for 22 years and AEs are rare: it is not associated with NAbs, or drug–drug interactions and there are no long-term safety signals.107 Safety concerns associated with some oral agents, however, need to be considered when selecting an appropriate therapy.108 Fingolimod has a risk of cardiovascular events, macular oedema, potential teratogenicity, influenza and herpes infections, gastroenteritis and bronchitis.37 With BG-12 a few cases of PML have been reported, and it is associated with flushing and gastrointestinal events.89,109 Teriflunomide increases the incidence of diarrhoea, nausea, hair thinning and raises levels of alanine transaminase (ALT)90 and laquinimod is associated with ALT elevations.110

Many patients will likely remain on injectables, but, in future, new patients may be given oral agents as first-line therapy. There is a general lack of experience with oral agents so some neurologists may be unwilling to use them immediately. Injectables, therefore, will not disappear but will become part of the mix. Neurologists, however, must take account of their patients’ opinions when choosing MS therapies.

The audience was asked if they agreed with the statement before and after hearing the arguments. It was therefore generally believed that injectables will continue to have a place in MS management.

Agree – Before Debate – 17 % and After Debate – 23 %
Disagree – Before Debate – 83 % and After Debate – 77 %

Conclusion
Per Soelberg Sørensen concluded the meeting. He asserted that this is an exciting time in MS, the treatment landscape is changing rapidly with many new therapies and diagnostic improvements arriving in quick succession. The overall prognosis for the patient newly diagnosed with MS is better now that it was 20 years ago when DMTs first became available. It is now possible to limit the numbers of patients developing the secondary progressive disease phase, the importance of early therapy is now recognised and more patients are receiving DMTs. Intensive effort is being directed towards effective therapies in SPMS for which treatment is much more challenging and there continues to be fewer options available. The new orals appear to offer equivalent or improved efficacy than the IFNbs with greater convenience, but experience with side effects could change the picture. They also raise the possibility of combination therapy, which could increase efficacy if complementary drugs are used. At the same time, however, the use of injectable therapies in MS continues to grow so those are unlikely to disappear anytime soon. Clinical experience will determine which of the new orals provide genuine new benefits in MS treatment and which have positive effects on long-term patient outcomes.

Definitions of Multiple Sclerosis Treatment Study Names
AFFIRM: Natalizumab Safety and Efficacy in Relapsing Remitting Multiple Sclerosis; ALLEGRO: Oral Laquinimod in Preventing Progression in Multiple Sclerosis; BENEFIT: Betaferon®/Betaseron® in Newly Emerging Multiple Sclerosis for Initial Treatment Study; BEYOND: Betaferon Efficacy Yielding Outcomes of a New Dose study; BRAVO: Laquinimod Double Blind Placebo Controlled Study in RRMS Patients With a Rater Blinded Reference Arm of Interferon β-1a (Avonex®); CHAMPS: Controlled High Risk Avonex Multiple Sclerosis Study; CLARITY: Cladribine Tablets Treating Multiple Sclerosis Orally; CombiRx: Combination Therapy in Patients With Relapsing-Remitting Multiple Sclerosis; CONCERTO: Efficacy and Safety and Tolerability of Laquinimod in Subjects With Relapsing Remitting Multiple Sclerosis; CONFIRM: Efficacy and Safety Study of Oral BG00012 With Active Reference in Relapsing- Remitting Multiple Sclerosis; DEFINE: Efficacy and Safety of Oral BG00012 in Relapsing-Remitting Multiple Sclerosis; EVIDENCE: EVidence of Interferon Dose-response: European North American Comparative Efficacy; FREEDOMS: Efficacy and Safety of Fingolimod in Patients With Relapsing-remitting Multiple Sclerosis; INCOMIN: Every-other-day Interferon Beta-1b Versus Once-weekly Interferon Beta-1a for Multiple Sclerosis; MIMS: Mitoxantrone in Multiple Sclerosis; PreCISe: Early Glatiramer Acetate Treatment in Delaying Conversion to Clinically Definite Multiple Sclerosis of Subjects Presenting With Clinically Isolated Syndrome; PRISMS: Prevention of Relapses and disability by Interferon beta-1a Subcutaneously in Multiple Sclerosis; REFORM: Tolerability of Rebif (New Formulation) (IFN Beta-1a) and Betaseron (IFN Beta-1b) in IFN-naive Subjects With Relapsing Remitting Multiple Sclerosis; REGARD: Rebif vs. Glatiramer Acetate in Relapsing MS Disease Study; TEMSO: Teriflunomide in Reducing the Frequency of Relapses and Accumulation of Disability in Patients With Multiple Sclerosis; TENERE: Study Comparing the Effectiveness and Safety of Teriflunomide and Interferon Beta-1a in Patients With Relapsing Multiple Sclerosis; TERACLES: Efficacy and Safety of Teriflunomide in Patients with Relapsing Multiple Sclerosis and Treated with Interferon-beta; TOPIC: Phase III Study With Teriflunomide Versus Placebo in Patients With First Clinical Symptom of Multiple Sclerosis; TOWER: Efficacy Study of Teriflunomide in Patients With Relapsing Multiple Sclerosis; TRANSFORMS: Trial Assessing Injectable Interferon versus FTY720 Oral in Relapsing–Remitting Multiple Sclerosis.

2

References

  1. National Multiple Sclerosis Society Medications for
    Modifying the Disease Course, 2013. Available at: http://
    www.nationalmssociety.org/about-multiple-sclerosis/whatwe-
    know-about-ms/treatments/index.aspx# (accessed 25
    September 2013).

  2. Fox EJ, Rhoades RW, New treatments and treatment goals
    for patients with relapsing-remitting multiple sclerosis, Curr
    Opin Neurol, 2012;(Suppl. 25):S11–19.

  3. Keegan BM, Therapeutic decision making in a new drug era
    in multiple sclerosis, Semin Neurol, 2013;33:5–12.

  4. Marta M, Giovannoni G, Disease modifying drugs in multiple
    sclerosis: mechanisms of action and new drugs in the
    horizon, CNS Neurol Disord Drug Targets, 2012;11:610–23.

  5. Granberg T, Martola J, Kristoffersen-Wiberg M, et al.,
    Radiologically isolated syndrome – incidental magnetic
    resonance imaging findings suggestive of multiple sclerosis,
    a systematic review, Mult Scler, 2013;19:271–80.

  6. Rommer PS, Stuve O, Management of secondary progressive
    multiple sclerosis: prophylactic treatment-past, present, and
    future aspects, Curr Treat Options Neurol, 2013;15:241–58.

  7. Scalfari A, Neuhaus A, Daumer M, et al., Onset of secondary
    progressive phase and long-term evolution of multiple
    sclerosis, J Neurol Neurosurg Psychiatry, 2013 [Epub ahead
    of print].

  8. Rovaris M, Confavreux C, Furlan R, et al., Secondary
    progressive multiple sclerosis: current knowledge and future
    challenges, Lancet Neurol, 2006;5:343–54.

  9. McKay FC, Swain LI, Schibeci SD, et al., CD127
    immunophenotyping suggests altered CD4+ T cell regulation
    in primary progressive multiple sclerosis, J Autoimmun,
    2008;31:52–8.

  10. Reyes-Palomares A, Rodríguez-López R, Ranea JAG, et al.,
    Global analysis of the human pathophenotypic similarity
    gene network merges disease module components, PLoS
    One, 2013;8(2):e56653.

  11. Polman CH, Reingold SC, Banwell B, et al., Diagnostic criteria
    for multiple sclerosis: 2010 revisions to the McDonald
    criteria, Ann Neurol, 2011;69:292–302.

  12. Tullman MJ, Oshinsky RJ, Lublin FD, et al., Clinical
    characteristics of progressive relapsing multiple sclerosis,
    Mult Scler, 2004;10:451–4.

  13. Lublin FD, Cofield SS, Cutter GR, et al., Randomized study
    combining interferon and glatiramer acetate in multiple
    sclerosis, Ann Neurol, 2013;73:327–40.

  14. Wolinsky J, Salter AR, Narayana P, et al., MRI Outcomes in
    CombiRx: Blinded, 7-Year Extension Results 65th Annual
    Meeting of the American Academy of Neurology, Honolulu,
    Hawaii, 9–16 April 2011.

  15. Hyland M, Rudick RA, Challenges to clinical trials in multiple
    sclerosis: outcome measures in the era of disease-modifying
    drugs, Curr Opin Neurol, 2011;24:255–61.

  16. van Winsen LM, Kragt JJ, Hoogervorst EL, et al., Outcome
    measurement in multiple sclerosis: detection of clinically
    relevant improvement, Mult Scler, 2010;16:604–10.

  17. Comabella M, Vandenbroeck K, Pharmacogenomics and
    multiple sclerosis: moving toward individualized medicine,
    Curr Neurol Neurosci Rep, 2011;11:484–91.

  18. Katsavos S, Anagnostouli M, Biomarkers in Multiple Sclerosis:
    An Up-to-Date Overview, Mult Scler Int, 2013;2013:340508.

  19. Komori M, Matsuyama Y, Nirasawa T, et al., Proteomic
    pattern analysis discriminates among multiple sclerosisrelated
    disorders, Ann Neurol, 2012;71:614–23.

  20. Rajasekharan S, Bar-Or A, Autoimmunity CMNiC, From bench
    to MS bedside: challenges translating biomarker discovery to
    clinical practice, J Neuroimmunol, 2012;248:66–72.

  21. Pravica V, Markovic M, Cupic M, et al., Multiple sclerosis:
    individualized disease susceptibility and therapy response,
    Biomark Med, 2013;7:59–71.

  22. Swingler RJ, Compston DA, The morbidity of multiple
    sclerosis, Q J Med, 1992;83:325–37.

  23. Lucas R, Ponsonby AL, McMichael A, et al., Observational
    analytic studies in multiple sclerosis: controlling bias through
    study design and conduct. The Australian Multicentre Study
    of Environment and Immune Function, Mult Scler,
    2007;13:827–39.

  24. Signori A, Baccino A, Sormani MP, The quality of reports of
    randomized trials in multiple sclerosis: a review, Mult Scler,
    2012;18:776–81.

  25. Havrdova E, Galetta S, Stefoski D, et al., Freedom from
    disease activity in multiple sclerosis, Neurology,
    2010;74(Suppl. 3):S3–7.

  26. Berkovich R, Treatment of acute relapses in multiple
    sclerosis, Neurotherapeutics, 2013;10:97–105.

  27. Castillo-Trivino T, Mowry EM, Gajofatto A, et al., Switching
    multiple sclerosis patients with breakthrough disease to
    second-line therapy, PLoS One, 2011;6:e16664.

  28. Portaccio E, Zipoli V, Siracusa G, et al., Switching to secondline
    therapies in interferon-beta-treated relapsing-remitting
    multiple sclerosis patients, Eur Neurol, 2009;61:177–82.

  29. Waubant E, Overview of treatment options in multiple
    sclerosis, J Clin Psychiatry, 2012;73:e22.

  30. Devonshire V, Havrdova E, Radue EW, et al., Relapse and
    disability outcomes in patients with multiple sclerosis
    treated with fingolimod: subgroup analyses of the doubleblind,
    randomised, placebo-controlled FREEDOMS study,
    Lancet Neurol, 2012;11:420–28.

  31. Hartung HP, Gonsette R, Konig N, et al., Mitoxantrone in
    progressive multiple sclerosis: a placebo-controlled, doubleblind,
    randomised, multicentre trial, Lancet, 2002;360:2018–
    25.

  32. Havrdova E, Galetta S, Hutchinson M, et al., Effect of
    natalizumab on clinical and radiological disease activity
    in multiple sclerosis: a retrospective analysis of the
    Natalizumab Safety and Efficacy in Relapsing-Remitting
    Multiple Sclerosis (AFFIRM) study, Lancet Neurol,
    2009;8:254–60.

  33. Cohen JA, Barkhof F, Comi G, et al., Oral fingolimod or
    intramuscular interferon for relapsing multiple sclerosis,
    N Engl J Med, 2010;362:402–15.

  34. Perini P, Calabrese M, Biasi G, et al., The clinical impact of
    interferon beta antibodies in relapsing-remitting MS,
    J Neurol, 2004;251:305–9.

  35. Polman CH, O’Connor PW, Havrdova E, et al., A randomized,
    placebo-controlled trial of natalizumab for relapsing multiple
    sclerosis, N Engl J Med, 2006;354:899–910.

  36. Sorensen PS, Bertolotto A, Edan G, et al., Risk stratification
    for progressive multifocal leukoencephalopathy in patients
    treated with natalizumab, Mult Scler, 2012;18:143–52.

  37. Willis MA, Cohen JA, Fingolimod therapy for multiple
    sclerosis, Semin Neurol, 2013;33:37–44.

  38. Martinelli Boneschi F, Vacchi L, Rovaris M, et al.,
    Mitoxantrone for multiple sclerosis, Cochrane Database Syst
    Rev, 2013;5:CD002127.

  39. Roskell NS, Zimovetz EA, Rycroft CE, et al., Annualized
    relapse rate of first-line treatments for multiple sclerosis:
    a meta-analysis, including indirect comparisons versus
    fingolimod, Curr Med Res Opin, 2012;28:767–80.

  40. Trapp BD, Ransohoff R, Rudick R, Axonal pathology in
    multiple sclerosis: relationship to neurologic disability,
    Curr Opin Neurol, 1999;12:295–302.

  41. Kappos L, Freedman MS, Polman CH, et al., Long-term
    effect of early treatment with interferon beta-1b after a first
    clinical event suggestive of multiple sclerosis: 5-year active
    treatment extension of the phase 3 BENEFIT trial, Lancet
    Neurol, 2009;8:987–97.

  42. Comi G, Martinelli V, Rodegher M, et al., Effects of early
    treatment with glatiramer acetate in patients with clinically
    isolated syndrome, Mult Scler, 2012;19:1074–83.

  43. Devonshire V, Lapierre Y, MacDonell R, et al., The Global
    Adherence Project – A multicentre observational study
    on adherence to disease-modifying therapies in patients
    suffering from relapsing-remitting multiple sclerosis, 22nd
    Congress of the European Committee for the Treatment
    and Research in Multiple Sclerosis, Madrid, Spain, 27–30
    September 2006.

  44. Fazekas F, Baumhackl U, Berger T, et al., Decision-making
    for and impact of early immunomodulatory treatment: the
    Austrian Clinically Isolated Syndrome Study (ACISS), Eur J
    Neurol, 2010;17:852–60.

  45. Goodin DS, Reder AT, Ebers GC, et al., Survival in MS: a
    randomized cohort study 21 years after the start of the
    pivotal IFNbeta-1b trial, Neurology, 2012;78:1315–22.

  46. Moses H, Jr., Brandes DW, Managing adverse effects of
    disease-modifying agents used for treatment of multiple
    sclerosis, Curr Med Res Opin, 2008;24:2679–90.

  47. Rieckmann P, Heidenreich F, Sailer M, et al., Treatment deescalation
    after mitoxantrone therapy: results of a phase IV,
    multicentre, open-label, randomized study of subcutaneous
    interferon beta-1a in patients with relapsing multiple
    sclerosis, Ther Adv Neurol Disord, 2012;5:3–12.

  48. Schwid SR, Panitch HS, Full results of the Evidence of
    Interferon Dose-Response-European North American
    Comparative Efficacy (EVIDENCE) study: a multicenter,
    randomized, assessor-blinded comparison of low-dose
    weekly versus high-dose, high-frequency interferon beta-1a
    for relapsing multiple sclerosis, Clin Ther, 2007;29:2031–48.

  49. Durelli L, Verdun E, Barbero P, et al., Every-other-day
    interferon beta-1b versus once-weekly interferon beta-1a
    for multiple sclerosis: results of a 2-year prospective
    randomised multicentre study (INCOMIN), Lancet,
    2002;359:1453–60.

  50. Leary SM, Miller DH, Stevenson VL, et al., Interferon beta-1a
    in primary progressive MS: an exploratory, randomized,
    controlled trial, Neurology, 2003;60:44–51.

  51. May TS, REGARD: Glatiramer acetate, interferon-beta equally
    effective for MS 23rd Congress of the European Committee
    for Treatment and Research in Multiple Sclerosis, Prague,
    Czech Republic, 2007. Available at: http://www.medscape.
    com/viewarticle/564944 (accessed 12 November 2013).

  52. Mendel R, Traut-Mattausch E, Frey D, et al., Do physicians’
    recommendations pull patients away from their preferred
    treatment options?, Health Expect, 2012;15:23–31.

  53. DeLuca GC, Williams K, Evangelou N, et al., The contribution
    of demyelination to axonal loss in multiple sclerosis, Brain,
    2006;129:1507–16.

  54. McLaughlin KA, Wucherpfennig KW, B cells and
    autoantibodies in the pathogenesis of multiple sclerosis and
    related inflammatory demyelinating diseases, Adv Immunol,
    2008;98:121–49.

  55. Handel AE, Lincoln MR, Ramagopalan SV, Of mice and
    men: experimental autoimmune encephalitis and multiple
    sclerosis, Eur J Clin Invest, 2011;41:1254–8.

  56. Stadelmann C, Bruck W, Interplay between mechanisms
    of damage and repair in multiple sclerosis, J Neurol,
    2008;255(Suppl. 1):12–18.

  57. Robertson N, Hirst C, Epidemiology of Progressive Multiple
    Sclerosis. In: Wilkins A (eds.), Progressive Multiple Sclerosis,
    London, UK: Springer-Verlag, 2013.

  58. Rothwell PM, McDowell Z, Wong CK, et al., Doctors and
    patients don’t agree: cross sectional study of patients’
    and doctors’ perceptions and assessments of disability in
    multiple sclerosis, BMJ, 1997;314:1580–83.

  59. Hobart J, Cano S, Baron R, et al., Achieving valid patientreported
    outcomes measurement: a lesson from fatigue in
    multiple sclerosis, Mult Scler, 2013; [Epub ahead of print].

  60. Critical Path Institute (‘C-Path’) – Multiple Sclerosis Outcome
    Assessments Consortium (MSOAC) 2013. Avaialble at:
    http://c-path.org/MSOAC.cfm (accessed 5 June 2013).

  61. Fisniku LK, Brex PA, Altmann DR, et al., Disability and T2 MRI
    lesions: a 20-year follow-up of patients with relapse onset of
    multiple sclerosis, Brain, 2008;131:808–17.

  62. Rocca MA, Mastronardo G, Rodegher M, et al., Long-term
    changes of magnetization transfer-derived measures from
    patients with relapsing-remitting and secondary progressive
    multiple sclerosis, AJNR Am J Neuroradiol, 1999;20:821–7.

  63. Vellinga MM, Castelijns JA, Barkhof F, et al., Postwithdrawal
    rebound increase in T2 lesional activity in natalizumabtreated
    MS patients, Neurology, 2008;70:1150–51.

  64. Deloire MS, Salort E, Bonnet M, et al., Cognitive impairment
    as marker of diffuse brain abnormalities in early relapsing
    remitting multiple sclerosis, J Neurol Neurosurg Psychiatry,
    2005;76:519–26.

  65. Preziosa P, Rocca MA, Mesaros S, et al., Intrinsic damage
    to the major white matter tracts in patients with different
    clinical phenotypes of multiple sclerosis: a voxelwise
    diffusion-tensor MR study, Radiology, 2011;260:541–50.

  66. Calabrese M, Filippi M, Gallo P, Cortical lesions in multiple
    sclerosis, Nat Rev Neurol, 2010;6:438–44.

  67. Mainero C, Benner T, Radding A, et al., In vivo imaging of
    cortical pathology in multiple sclerosis using ultra-high field
    MRI, Neurology, 2009;73:941–8.

  68. Agosta F, Rovaris M, Pagani E, et al., Magnetization transfer
    MRI metrics predict the accumulation of disability 8 years
    later in patients with multiple sclerosis, Brain,
    2006;129:2620–7.

  69. Rovaris M, Judica E, Gallo A, et al., Grey matter damage
    predicts the evolution of primary progressive multiple
    sclerosis at 5 years, Brain, 2006;129:2628–34.

  70. Christodoulou C, Krupp LB, Liang Z, et al., Cognitive
    performance and MR markers of cerebral injury in
    cognitively impaired MS patients, Neurology, 2003;60:1793–8.

  71. Mainero C, De Stefano N, Iannucci G, et al., Correlates of MS
    disability assessed in vivo using aggregates of MR quantities,
    Neurology, 2001;56:1331–4.

  72. Sumowski JF, Wylie GR, Leavitt VM, et al., Default network
    activity is a sensitive and specific biomarker of memory in
    multiple sclerosis, Mult Scler, 2013;19:199–208.

  73. Valsasina P, Rocca MA, Absinta M, et al., Cervical cord FMRI
    abnormalities differ between the progressive forms of
    multiple sclerosis, Hum Brain Mapp, 2012;33:2072–80.

  74. Rocca MA, Colombo B, Falini A, et al., Cortical adaptation in
    patients with MS: a cross-sectional functional MRI study of
    disease phenotypes, Lancet Neurol, 2005;4:618–26.

  75. Rocca MA, Valsasina P, Ceccarelli A, et al., Structural and
    functional MRI correlates of Stroop control in benign MS,
    Hum Brain Mapp, 2009;30:276–90.

  76. Brinkmann V, Cyster JG, Hla T, FTY720: sphingosine
    1-phosphate receptor-1 in the control of lymphocyte
    egress and endothelial barrier function, Am J Transplant,
    2004;4:1019–25.

  77. Schwab SR, Cyster JG, Finding a way out: lymphocyte egress
    from lymphoid organs, Nat Immunol, 2007;8:1295–301.

  78. Kim HJ, Miron VE, Dukala D, et al., Neurobiological effects
    of sphingosine 1-phosphate receptor modulation in the
    cuprizone model, FASEB J, 2011;25:1509–18.

  79. Claussen MC, Korn T, Immune mechanisms of new
    therapeutic strategies in MS: teriflunomide, Clin Immunol,
    2012;142:49–56.

  80. Linker RA, Lee DH, Ryan S, et al., Fumaric acid esters exert
    neuroprotective effects in neuroinflammation via activation
    of the Nrf2 antioxidant pathway, Brain, 2011;134:678–92.

  81. van Horssen J, Witte ME, Schreibelt G, et al., Radical changes
    in multiple sclerosis pathogenesis, Biochim Biophys Acta,
    2011;1812:141–50.

  82. Scannevin RH, Chollate S, Jung MY, et al., Fumarates promote
    cytoprotection of central nervous system cells against
    oxidative stress via the nuclear factor (erythroid-derived
    2)-like 2 pathway, J Pharmacol Exp Ther, 2012;341:274–84.

  83. Schulze-Topphoff U, Shetty A, Varrin-Doyer M, et al.,
    Laquinimod, a quinoline-3-carboxamide, induces type
    II myeloid cells that modulate central nervous system
    autoimmunity, PLoS One, 2012;7:e33797.

  84. Kappos L, Radue EW, O’Connor P, et al., A placebocontrolled
    trial of oral fingolimod in relapsing multiple
    sclerosis, N Engl J Med, 2010;362:387–401.

  85. Cohen J, Radue E-W, F B, et al., Fingolimod-Effect on Brain
    Atrophy and Clinical/MRI Correlations in Three Phase 3
    Studies – TRANSFORMS, FREEDOMS and FREEDOMS II, 65th
    Annual Meeting of the American Academy of Neurology, San
    Diego, USA, 16–23 March 2013.

  86. Kappos L, Radue E-W, O’Connor P, et al., Long-Term Efficacy
    and Safety of Fingolimod (FTY720) in Relapsing-Remitting
    Multiple Sclerosis (RRMS): Results from the Extension of the
    Phase III FREEDOMS Study (S41.004), 64th Annual Meeting
    of the Anmerican Academy of Neurology, New Orleans, USA,
    22–27 April 2012.

  87. Goodin D, Jeffery D, Kappos L, et al., Fingolimod Reduces
    Annualized Relapse Rate in Patients with Relapsing-
    Remitting Multiple Sclerosis: FREEDOMS II Study Subgroup
    Analysis (P07.102), 65th annual meeting of the American
    Academy of Neurology, San Diego, USA, 16–23 March 2013.

  88. Gold R, Kappos L, Arnold DL, et al., Placebo-controlled
    phase 3 study of oral BG-12 for relapsing multiple sclerosis,
    N Engl J Med, 2012;367:1098–107.

  89. Fox RJ, Miller DH, Phillips JT, et al., Placebo-controlled phase
    3 study of oral BG-12 or glatiramer in multiple sclerosis,
    N Engl J Med, 2012;367:1087–97.

  90. O’Connor P, Wolinsky JS, Confavreux C, et al., Randomized
    trial of oral teriflunomide for relapsing multiple sclerosis,
    N Engl J Med, 2011;365:1293–303.

  91. Kappos L, Comi G, Confavreux C, et al., The efficacy and
    safety of teriflunomide in patients with relapsing MS: results
    from TOWER, a phase III, placebo-controlled study, 28th
    Congress of the European Committee for Treatment and
    Research in Multiple Sclerosis, Lyon, France, 10–13
    October 2012.

  92. Singer B, Comi G, Miller A, et al., Frequency of Infections
    during Treatment with Teriflunomide: Pooled Data from Three
    Placebo-Controlled Teriflunomide Studies P01.171, 65th
    Annual Meeting the American Academy of Neurology, San
    Diego, USA, 2013.

  93. Vermersch P, Czlonkowska A, Grimaldi LM, et al.,
    Teriflunomide versus subcutaneous interferon beta-1a in
    patients with relapsing multiple sclerosis: a randomised,
    controlled phase 3 trial, Mult Scler, 2013 [Epub ahead of
    print].

  94. Vollmer T, Comi G, Solberg Sorensen P, et al., Clinical Efficacy
    of Laquinimod for the Treatment of Multiple Sclerosis; Pooled
    Analyses from the ALLEGRO and BRAVO Phase III Trials
    64th AAN Annual Meeting of the American Academy of
    Neurology, New Orleans, USA, 21–28 April 2012.

  95. Coles AJ, Cox A, Le Page E, et al., The window of therapeutic
    opportunity in multiple sclerosis: evidence from monoclonal
    antibody therapy, J Neurol, 2006;253:98–108.

  96. Coles AJ, Twyman CL, Arnold DL, et al., Alemtuzumab for
    patients with relapsing multiple sclerosis after diseasemodifying
    therapy: a randomised controlled phase 3 trial,
    Lancet, 2012;380:1829–39.

  97. Bosca I, Coret F, Valero C, et al., Effect of relapses over early
    progression of disability in multiple sclerosis patients treated
    with beta-interferon, Mult Scler, 2008;14:636–9.

  98. Lublin FD, Baier M, Cutter G, Effect of relapses on
    development of residual deficit in multiple sclerosis,
    Neurology, 2003;61:1528–32.
    99. Bermel RA, Fox RJ, MRI in multiple sclerosis, Continuum
    (Minneap Minn), 2010;16:37–57.

  99. Bermel RA, You X, Foulds P, et al., Predictors of long-term
    outcome in multiple sclerosis patients treated with
    interferon beta, Ann Neurol, 2013;73:95–103.

  100. Goodin DS, Traboulsee A, Knappertz V, et al., Relationship
    between early clinical characteristics and long term
    disability outcomes: 16 year cohort study (follow-up) of the
    pivotal interferon beta-1b trial in multiple sclerosis, J Neurol
    Neurosurg Psychiatry, 2012;83:282–7.

  101. Rio J, Nos C, Tintore M, et al., Defining the response to
    interferon-beta in relapsing-remitting multiple sclerosis
    patients, Ann Neurol, 2006;59:344–52.
    Daugherty KK, Butler JS, Mattingly M, et al., Factors leading
    patients to discontinue multiple sclerosis therapies, J Am
    Pharm Assoc (2003), 2005;45:371–5.

  102. Steinberg SC, Faris RJ, Chang CF, et al., Impact of adherence
    to interferons in the treatment of multiple sclerosis: a nonexperimental,
    retrospective, cohort study, Clin Drug Investig,
    2010;30:89–100.

  103. Rio J, Castillo J, Rovira A, et al., Measures in the first year
    of therapy predict the response to interferon beta in MS,
    Mult Scler, 2009;15:848–53.

  104. Rio J, Comabella M, Montalban X, Predicting responders
    to therapies for multiple sclerosis, Nat Rev Neurol,
    2009;5:553–60.

  105. Johnson KP, Glatiramer acetate for treatment of relapsingremitting
    multiple sclerosis, Expert Rev Neurother,
    2012;12:371–84.

  106. Killestein J, Rudick RA, Polman CH, Oral treatment for
    multiple sclerosis, Lancet Neurol, 2011;10:1026–34.

  107. van Oosten BW, Killestein J, Barkhof F, et al., PML in a patient
    treated with dimethyl fumarate from a compounding
    pharmacy, N Engl J Med, 2013;368:1658–9.

  108. Comi G, Jeffery D, Kappos L, et al., Placebo-controlled trial
    of oral laquinimod for multiple sclerosis, N Engl J Med,
    2012;366:1000–1009.
3

Article Information

Disclosure

Multiple Sclerosis Management: A Changing Landscape 2013 was an independent educational meeting funded by an educational grant from Teva Pharmaceuticals Inc.

Correspondence

Per Soelberg Sørensen, Danish Multiple Sclerosis Center, Department of Neurology 2082, Rigshospitalet, DK-2100 Copenhagen, Denmark. E: pss@rh.dk

Support

The publication of this article was supported by Teva Pharmaceuticals Inc. The views and opinions expressed are those of the expert presenters and not necessarily those of Teva Pharmaceuticals Inc.
An erratum to this article can be found below.

Acknowledgements

Editorial assistance was provided by James Gilbart at Touch Medical Media. The following expert presenters also reviewed the content of these proceedings prior to publication: Julián Benito-Léon (Madrid, Spain), Angie Fagerlin (Ann Arbor, Michigan, US), Franz Fazekas (Graz, Austria), Oscar Fernandez (Málaga, Spain), Gavin Giovannoni (London, UK), Maria Houtchens (Boston, Massachusetts, US), Raj Kapoor (London, UK), Michael Khalil (Graz, Austria), Bernd Kieseier (Düsseldorf, Germany), Hans Lassmann (Vienna, Austria), Fred D Lublin (New York, New York, US), Gianluigi Mancardi (Genoa, Italy), Lauren Strober (West Orange, New Jersey, US), Marc Tardieu (Paris, France), Alan Thompson (London, UK) and Mauro Zaffaroni (Gallarate, Italy).

Received

2013-11-01T00:00:00

4

Further Resources

Share
Facebook
X (formerly Twitter)
LinkedIn
Via Email
Mark CompleteCompleted
BookmarkBookmarked
Copy LinkLink Copied
Download as PDF

This Functionality is for
Members Only

Explore the latest in medical education and stay current in your field. Create a free account to track your learning.

Close Popup