2,178
Views
2
CrossRef citations to date
0
Altmetric
Neurology

Management of relapsing–remitting multiple sclerosis in Qatar: an expert consensus

, , , , , , , & show all
Pages 251-260 | Received 29 May 2019, Accepted 16 Sep 2019, Published online: 07 Oct 2019

Abstract

Healthcare systems vary greatly between countries. International, evidence-based guidelines for the management of multiple sclerosis (MS) may need to be adapted for use in particular countries. Two years ago, the authors published a comprehensive consensus guideline for the management of MS in Qatar. Since that time, the availability of disease-modifying treatments for relapsing–remitting MS (RRMS), and our understanding of how to apply those treatments, has increased. The authors present an update to our guidance, focussing on the management of relapsing–remitting RRMS. In particular, the authors consider the optimal use of different DMTs in patients presenting with mild, medium or high disease activity.

Introduction

A number of new disease-modifying therapies (DMTs) for relapsing–remitting multiple sclerosis (RRMS) have been introduced in recent years, broadening and complicating the design of therapeutic interventions for an individual patient. “First-line” or “platform” DMTs (interferons, glatiramer acetate, dimethyl fumarate and teriflunomide) have been supplanted to some extent by a new generation of highly potent DMTs (alemtuzumab, cladribine tablets 10 mg1, fingolimod, natalizumab and ocrelizumab) that has changed the landscape of care for people with RRMSCitation1. Indeed, these drugs have enabled long periods (years) of disease activity-free remission for substantial proportions of patientsCitation2. Challenging tolerability profiles and/or paucity of information on long-term outcomes with newer agents have led to some caution on the part of regulatory authorities concerning their therapeutic use.

The debate continues as to when to prescribe a given high efficacy DMT for a given patient. Free healthcare is available for all Qatari nationals, and restrictions on prescribing a given DMT for a given type of patient are less strict than in other regions. Thus, clinical practice in Qatar is well placed to benefit from the increase in the number of efficacious DMTs. Two years ago, an expert group from Qatar published its consensus recommendations on the management of RRMS in that countryCitation3. This article seeks to extend and update this earlier guidance. Our primary focus here is on the optimal management of RRMS, within the context of our current understanding of how disease activity and other medical, personal and social factors impact on treatment goals and decisions.

Epidemiology and characteristics of multiple sclerosis in Qatar

Qatar is a relatively small nation, with a population estimated at about 2.7 million in December 2018Citation4. All new cases of MS are assessed at a single tertiary referral centre, the Hamad General Hospital (HGH), and all treatments for MS are dispensed from there. Accordingly, the HGH provides an appropriate data source for measuring the prevalence of RRMS, and its treatment, in Qatari nationals. This was done in two publications, published in 2013Citation5,Citation6.

Identification of 154 patients with MS after exclusion of patients with neuromyelitis optica and isolated transverse myelitis yielded a crude prevalence of MS of 65/100,000 population (95% CI: 58–70)Citation6. Accordingly, Qatar is an area of medium-to-high prevalence of MS, according to the classical Kurtzke classificationCitation7. The female-to-male ratio was 1.33, which was broadly similar to other populations within and outside the regionCitation6. An analysis restricted to 142 newly diagnosed patients at this institution demonstrated a relatively low median EDSS score of 2 (20%) and a preponderance towards sensory symptoms (63%) and visual symptoms (45%), rather than motor (43%), cerebellar (32%), brainstem (27%), cord (14%), or bladder or bowel (10%) symptoms. The authors considered that these findings suggested a milder clinical presentation of MS, but a severe radiologic presentation, compared with nearby countries.

Analysis of hospital records showed that roughly similar numbers of patients with MS in Qatar were treated with interferon β (44%) or were untreated (45%), due to either non-prescription of MS treatments or non-adherence to treatment. Otherwise, patients had been treated with fingolimod (10%) or natalizumab (1%). However, these data reflect the availability of DMTs for MS in 2010, and a number of new DMTs for the management of MS have become available recently.

Defining the activity of relapsing–remitting multiple sclerosis

There are no consensus criteria for defining the activity of RRMS. Moreover, a recent guideline from expert European societies on the pharmacologic management of RRMS advocates treatment based on the level of disease activity, without providing objective criteria for assessing thisCitation8. The European Medicines Agency (EMA) recommends consideration of progression of the clinical burden of disease (number of relapses, worsening disability and increasing radiologic burden) in defining RRMS activityCitation9. For our discussions, we have defined highly active disease and mild disease (moderate disease would show features of both) that follow the criteria used within the EMA’s evaluation of cladribine tablets, fingolimod and natalizumabCitation10–14. The criteria for these definitions are shown in Box 1.

These criteria provide a useful and practical guide to assessing disease activity, although individual patient or prognostic factors should be taken into account, such as older age at presentation, incomplete recovery from relapses, the presence of motor relapses at presentation, rapid progression of disability (e.g. increase of at least one EDSS point in 1 year), or presentation with spinal, or cerebellar or brainstem lesions. The clinical judgement of the individual neurologist will always play an important part in defining the activity of RRMS for a given patient.

Box 1. Criteria for defining highly active relapsing–remitting multiple sclerosis (RRMS).

Highly active RRMS

Treatment-naive patients:

≥2 relapses in one year

And ≥1 gadolinium-enhancing lesions on brain magnetic resonance imaging (MRI)

Or a significant increase in T2 lesion load compared to a previous recent MRI

Patients treated with at least one disease modifying therapya:

≥1 relapse in the previous year while on therapy

And ≥9 T2 hyperintense lesions in cranial MRI

Or ≥1 gadolinium-enhancing lesion

Mild RRMS

Treatment-naive patients:

≤1 disabling relapse in 1 year

Or no gadolinium-enhancing lesions, and no significant increase in T2 lesion load compared with a recent MRI

Patients treated with at least one disease modifying therapya:

Decreased relapse rate and no ongoing severe relapses

No relapses in the past year

Or no Gd + lesions and no more than 8 T2 lesions

aTreatment usually for at least 1 year.

Management of relapsing–remitting multiple sclerosis according to disease activity

Overview of current disease-modifying treatments for relapsing–remitting multiple sclerosis

Purpose of this section

The consensus guidance we published in 2017 included a review of individual treatments for RRMS that were available at that time: alemtuzumab, dimethyl fumarate, fingolimod, glatiramer acetate, interferons, mitoxantrone, natalizumab, ocrelizumab and teriflunomideCitation3. Accordingly, only a brief account of the properties of these medications will be given here, based on published reviews and the European labelling for these agents, with discussion focussed on new information that has appeared since our earlier consensus publication. Cladribine has become available for the management of RRMS since the previous consensus, and some of its therapeutic properties are described briefly below.

Efficacy

Interferons were the first DMT to be introduced in the management of RRMS in the early 1990s, and rapidly became the standard of care for RRMS following demonstration of reduced risk of relapses and reduced frequency of MRI lesions, and reductions in the accumulation of disability, as reviewed previouslyCitation3. Indeed, the widespread exposure of people with RRMS to these agents may have contributed to a decline in the average severity of RRMS over time, as shown by a declining background relapse rate in randomized, controlled trialsCitation15.

summarizes principal results from randomized evaluations of newer, high efficacy DMTsCitation16–34. Caution must always be applied when comparing results across clinical trials, due to differences in their designs and patient populations. Nevertheless, it is clear that substantial reductions in relapse rates were observed with DMTs in placebo-controlled trials, often with evidence of disability, and with significantly increased achievement of “no evidence of disease activity” (NEDA) outcomes compared with placebo. Few randomized, head-to-head phase 3 comparisons between different DMTs are available, and all of these included a formulation of interferon as a comparator. Randomization to alemtuzumab, ocrelizumab or fingolimod was associated with greater efficacy compared with interferon β-1a in these trials, as shown by larger reductions in relapse rates, MRI lesions, and greater achievement of NEDA over time periods up to three years ().

Table 1. Overview of principal randomized phase 3 evaluations of disease-modifying therapies (DMTs) for multiple sclerosis.

Tolerability and safety

The safety profile of first-line DMTs, interferon β, glatiramer acetate and dimethyl fumarate, has been reviewed elsewhereCitation35–40. Briefly, the main side-effects of interferon β (flu-like symptoms and injection site reactions) rarely cause treatment discontinuation if they are not accompanied by more serious safety issues during long-term treatmentCitation35–37. Moreover, 20 years of post-trial follow-up has demonstrated a possible reduction in mortality in interferon β-treated patients, compared with patients originally randomly allocated to placeboCitation37. Glatiramer acetate is associated with immediate post-injection reactions, which may include including flushing, palpitations and dyspnoeaCitation38,Citation39.

Flushing and gastrointestinal side-effects are common in patients initiating dimethyl fumarate (DMF) and may cause treatment discontinuationCitation41. Five cases of progressive multifocal leukoencepalopathy (PML) in patients with RRMS receiving DMF have been reported in the literature, with an additional 14 cases observed in people receiving DMF for psoriasis; most of these cases occurred in patients who developed prolonged lymphopeniaCitation42. Finally, teriflunomide has been associated mainly with increased blood pressure and abnormal liver function testsCitation43.

summarizes the safety profile of the high efficacy DMTs. The safety and tolerability profiles vary widely between individual DMTs. The main side-effects of alemtuzumab are infusion-associated reactions (occurring in more than 90% of patients and occasionally severe), autoimmunity (mainly involving the thyroid), and infections, principally herpes simplex and zoster in the upper respiratory tractCitation44. Pretreatment with corticosteroids (and, optionally, antipyretics) is given to reduce the severity of infusion-associated reactions. Immunization against varicella zoster for patients previously unexposed to this virus and initiation of anti-herpes prophylaxis is recommended before initiating alemtuzumab.

Table 2. Most common side-effects and monitoring burden associated with DMTs, according to their European summaries of product characteristics.

Treatment with cladribine tablets is relatively well tolerated by people with RRMS; its main side-effects are lymphopenia/leukopenia and opportunistic infections, mainly herpes zoster, according to published informationCitation45,Citation46, its European labelling and the experience of physicians. Cladribine tablets do not appear to be associated with increased risk of PMLCitation46, although their European labelling contains a note that PML has occurred in patients taking a different cladribine regimen for the treatment of leukaemia. Patients should be screened for latent tuberculosis or hepatitis to reduce the risk of later activation of these diseases. Patients with very low leukocyte counts (<500/mm3) should be monitored actively for infections. No significant increase in the rate of malignancy was observed in patients with MS in an integrated analysis of clinical evaluations of cladribine tablets, or in the PREMIERE Registry of patients with RRMS receiving this treatmentCitation46, consistent with data reported previouslyCitation10,Citation47.

The main safety concern with natalizumab is progressive multifocal leukoencepalopathy (PML), an opportunistic infection caused by JC virusCitation48,Citation49. The presence of JC virus antibodies, treatment with natalizumab for at least 2 years and prior treatment with an immunosuppressant are all risk factors for PML during treatment with natalizumab. Consideration of these risk factors when planning to prescribe natalizumab has greatly reduced the risk of PML, and researchers are seeking biomarkers to help reduce this risk furtherCitation50.

Infusion reactions are common with ocrelizumab, and dose adjustments or permanent withdrawal of therapy are required where they are potentially life threateningCitation51,Citation52. The European labelling carries a warning about a possible increase in malignancy, including breast cancersCitation51. The risk of PML with ocrelizumab is unknown at present, and cases of PML have not been reported in MS patients receiving this treatmentCitation53. Ocrelizumab is an inhibitor of CD20. PML and other opportunistic infections (including reactivation of hepatitis B) have been observed as rare events in other CD20 inhibitors in patients with rheumatoid arthritis or malignant disease who also received other antineoplastic drugsCitation53–55.

Fingolimod is associated with several principal side-effects, including bradycardia, macular oedema (especially early in therapy), hepatic abnormalities and an increased risk of malignant diseaseCitation56–58. PML has also been reported in patients taking fingolimodCitation59.

Administration and treatment burden

also summarizes the administration regimens and principal requirements for initial and ongoing monitoring associated with these DMTs. Administration is oral for teriflunomide and fingolimod, and by regular, periodic infusions or injections for natalizumab and ocrelizumab. Alemtuzumab and cladribine tablets are hypothesized to be members of the emerging sub-class of immune reconstitution therapies (IRTs)Citation60,Citation61, The rationale and mechanisms underlying IRTs has been reviewed elsewhereCitation62,Citation63. Briefly, suppression of components of the immune system is followed by a gradual rebuilding of the population of T and B cells, without recovery of the clones that gave rise to inflammation and demyelination, where the treatment has been successful. The main hallmark of IRTs is a period of efficacy that long outlasts both the duration of drug administration and the persistence of drug in the plasma.

The administration regimens of alemtuzumab and cladribine tablets require two short courses of treatment 1 year apart. Further treatment with alemtuzumab may be given in years three and four, if required, but there is no requirement for re-treatment with cladribine tablets in years three and four, based on the results of an extension to the CLARITY randomized evaluation of this agentCitation64. Accordingly, these agents demonstrate some features of an IRT. Immune cells recover gradually after treatment with cladribine tablets, without the overshoot in B cells seen with alemtuzumab, which may account for the difference in safety between these agents with regard to activation of autoimmunity. Cladribine also has greater effects on components of the adaptive system, with less effect on the innate immune systemCitation65.

Monitoring requirements during and after initiation of treatments also vary considerably between DMTs (). Of the immune reconstitution therapies, alemtuzumab has a considerably heavier monitoring burden than cladribine tablets, with a requirement for monthly monitoring of blood counts and renal function, and regular monitoring for autoimmune damage to the thyroid. Haematological monitoring requirement with cladribine tablets is required to ensure that lymphocyte counts have recovered sufficiently before administration of the second course of treatment in year 2 of therapy. Lymphocytes should also be measured at months 2 and 6 of each treatment year, and followed thereafter if <500/mm3; patients with lymphocyte counts <500/mm3 should also be monitored for signs of infection, particularly zoster. Monitoring is also required if concomitant treatments that may affect blood counts or exposure to cladribine are taken. Fingolimod and teriflunomide require monitoring for their cardiovascular and hepatic side-effects, respectively. The risk of PML increase with the duration of treatment with natalizumab, and patients should be re-counselled on this after 2 years of treatment.

Choosing a disease-modifying therapy

summarizes the authors’ consensus recommendations for selection of a DMT during different clinical scenarios across the spectrum of disease severity in patients with RRMS. Specifically, we considered treatment of a patient without prior therapy with a DMT, and a patient who has demonstrated sub-optimal clinical response to one or two DMTs. Further discussion of the clinical context surrounding these decisions is given below.

Table 3. Consensus recommendations on the pharmacologic management of RRMS according to disease activity.

Goals of treatment

The goals of management of all people with RRMS relate to the disease (reducing relapse rates, preserving long-term functional status), the treatment (avoiding side-effects as far as possible, minimizing the burden of treatment and its associated monitoring) and the patient’s lifestyle (maintaining quality of life, supporting good adherence to therapy, delivering treatment in a way that fits well with the individual patient’s lifestyle and accommodating the patient’s need to plan a family)Citation66–68. Disease activity may alter priorities to some extent. Where disease activity is low or moderate, convenience and family planning may take a relatively high priority, and the relatively benign first-line DMTs often used in these patients have well understood safety and tolerability profiles. For the patient with high disease activity and a likely adverse long-term prognosis, preventing further relapses, radiologic progression and associated progression of disability is paramount. The more effective suppression of disease activity with high efficacy DMTs may come at a price of more, and potentially serious, side-effects and more monitoring, although this varies between individual drugs as described above. Family planning presents an especially difficult challenge in this setting. The following sections address the impact of disease activity at presentation on the choice of a DMT.

Disease activity and selection of treatment

In Qatar, it is usual practice to prescribe “first-line” or “platform” DMTs where disease activity is low: RRMS in these patients can often be controlled adequately by interferon (preferably at the higher dose) or glatiramer acetate, without need for agents with more challenging safety and tolerability profilesCitation8. High efficacy DMTs are preferred where interferons are considered unlikely to provide sufficient protection from relapses for patients with highly active disease, or patients who have already experienced breakthrough disease while receiving a first-line DMTCitation8. For patients with moderate disease, the choice between a first-line or high efficacy DMT is likely to depend on the outcome of discussions with the patient regarding their personal circumstances and preferences (see below).

Most DMTs induce suppression of immune function, either temporarily (IRT-like DMTs) or continuously (immunosuppressant drugs given regularly throughout their use). Switching between DMTs is problematic where drugs either persist on the body (e.g. teriflunomide) or have long-lasting effects on white cell countsCitation69. If the chosen DMT cannot be administered immediately, for example because of leukopenia persisting from earlier treatment, a period without treatment, or bridging therapy with a corticosteroid, interferon or glatiramer acetate are options for filling this treatment gapCitation70. These strategies are not without risk, however; for example, switching between natalizumab and fingolimod has been shown to result in increased relapse rates, especially in patients with more pronounced disabilityCitation71.

Switching between disease-modifying therapies

Alemtuzumab and cladribine tablets both require two administrations 1 year apart before the full dose has been given. It is unclear whether the occurrence of a relapse subsequent to this time should prompt a treatment switch or retreatment with the original treatment; the debate continues as to how many relapses constitute failure of a DMT with characteristics of an IRT in a patient who probably received this therapy due to high disease activityCitation72.

Family planning

Planning a pregnancy provides a dilemma for patients with RRMS and their physicians, in which the risks of active treatment must be balanced with the risks of relapse if treatment is withdrawnCitation73. Cultural issues around pregnancy may be particularly problematic in Middle-Eastern countries such as Qatar, as there is a widespread preference for achieving pregnancy early in marriage, followed by raising large families, so that some Qatari women may spend a substantial period of their childbearing years in pregnancyCitation74.

Most DMTs are contraindicated during pregnancyCitation75. Most physicians recommend stopping DMT treatment in the event of pregnancyCitation76. Relapse rates reduce during pregnancy for most women, especially in the third trimester. However, women with high relapse rates before pregnancy that required treatment with a high-efficacy DMT, especially where disability is present, may be at risk of a catastrophic relapse in early pregnancyCitation77. A recent expert consensus guideline from the UK recommends that women with RRMS should not delay initiation of DMTs for family planningCitation73. While the reduction in relapse rates that occurs during pregnancy may allow some women to discontinue their DMT when they become pregnant, this expert consensus supports the continuation of DMTs during pregnancy for women with highly active diseaseCitation73.

More research is needed to confirm the role of IRT-like DMTs for women who wish to plan a pregnancy. European labelling requires the avoidance of pregnancy for 6 months after the last dose of cladribine tablets and for 4 months after administration of alemtuzumab. As described above, a prolonged period of freedom from relapses has been observed with cladribine tablets and alemtuzumab in non-pregnant patients that outlasts the period of treatment and persistence of the drug in the body. However, more data are required to demonstrate whether this long-term efficacy is maintained in the setting of pregnancy, and that there are no adverse effects of treatment on maternal or foetal outcomes, before definitive guidance can be proposed regarding the place of IRT-like DMTs in the management of patients planning a family.

The postpartum period is associated with increased risk of relapses, and the need for restarting DMT (if this has been stopped prior to or during pregnancy) needs to be balanced against providing the opportunity for the mother to breastfeedCitation77. New guidance from the UK stresses that women should be encouraged to breastfeed, alongside other treatment considerations, taking the mother’s needs and preferences into accountCitation73. US and European labelling for some DMTs does not contraindicate their use during breastfeeding, according to individualized risk:benefit considerations. At the time of writing, both US and European labelling support cautious use of beta interferons and glatiramer acetate during breastfeeding. US labelling does not rule out use of natalizumab and ocrelizumab during breastfeeding, but European labelling does not support this use.

Patient preferences

Patients with RRMS often deal with their devastating diagnosis by seeking information on the internet and from support groups and other people with RRMS via social mediaCitation78,Citation79. Accordingly, patients are increasingly knowledgeable, motivated and determined to participate actively in their care when they arrive for their consultation. Healthcare professionals tend to focus on adverse aspects of treatments for RRMS (safety issues), while patients may also focus on positive aspects (e.g. beneficial impact on treatment outcomes)Citation79. RRMS is a disease primarily of onset in young adulthood, and some patients may accept the risks of adverse events with a high-activity DMT or IRT-like DMT in return for the prospect of a longer period of freedom from relapses and a good long-term functional outcomeCitation80. Preference for oral vs. injectable treatment, frequency of administration and ability to accommodate monitoring requirements into a patient’s lifestyle may also be importantCitation79,Citation81. Treatment preferences themselves are modified by patients’ current mode of treatment and disability statusCitation82.

Managing relapses

A systematic review of clinical trials in relapsing patients with MS concluded that intravenous or oral methylprednisolone is effective for the management of acute relapses in patients with RRMS, including where acute optic neuritis is present, with use of doses of at least 500 mg/day for 3–5 days, depending on presentationCitation83. The European labelling for powdered formulations of methylpredisolone includes an indication for “acute exacerbations of multiple sclerosis superimposed on a relapsing–remitting background”. An infusion of 1 g methylprednisolone daily, infused over 30 min is recommended for management of acute MS relapses using these preparations. Current guidance for the management of MS supports the use of corticosteroid treatment to reduce the duration and severity of relapses, with appropriate counselling on possible adverse effects of high-dose steroids, such as temporary psychological disturbances, or deterioration of glycemic control in people with diabetesCitation84. Steroid treatment for MS relapses does not preclude breastfeedingCitation73. Plasmapheresis is effective in patients with acute relapses refractory to steroids, and this treatment is supported by US guidelinesCitation85.

Transparency

Declaration of funding

The consensus meeting that gave rise to this article was funded by Merck Serono Middle East FZ-Ltd. This company did not influence the treatment recommendations made here, which are the views of the authors.

Declaration of financial/other relationships

A.B. has disclosed that he is an employee of Merck Serono Middle East FZ-Ltd. D.D. has disclosed that he has served on Advisory Boards of Merck, Novartis, Biologix, Roche and Sanofi and is a member of MENACTRIMS. B.C. has disclosed that she has received travel, speaker and consultant honoraria from Merck, Novartis, Biologix, Roche and Sanofi. O.E. has disclosed that he has received an honorarium from Merck. Y.A. has disclosed that he has participated in Advisory Boards for Merck. M.S.A., G.A., B.M. and G.M. report no duality of interest additional to participation in the consensus meeting funded by Merck Serono Middle East FZ-Ltd, described above. CMRO peer reviewers on this manuscript have no relevant financial or other relationships to disclose.

Acknowledgements

A medical writer (funded by Merck Serono Middle East FZ-Ltd) provided editorial assistance.

Notes

Correction Statement

This article has been republished with minor changes. These changes do not impact the academic content of the article.

Notes

1 Refers to Cladribine Tablets 10 mg (3.5 mg/kg cumulative dose over 2 years, referred to elsewhere in this article as Cladribine Tablets).

References

  • Tintore M, Vidal-Jordana A, Sastre-Garriga J. Treatment of multiple sclerosis – success from bench to bedside. Nat Rev Neurol. 2019;15(1):53–58.
  • Giovannoni G, Bermel R, Phillips T, et al. A brief history of NEDA. Mult Scler Relat Disord. 2018;20:228–230.
  • Deleu D, Mesraoua B, El Khider H, et al. Optimization and stratification of multiple sclerosis treatment in fast developing economic countries: a perspective from Qatar. Curr Med Res Opin. 2017;33(3):439–458.
  • Qatar Ministry of Development and Statistics [Internet] [cited 2019 Mar]. Available from: https://www.mdps.gov.qa/en/Pages/default.aspx
  • Akhtar N, Elsetouhy A, Deleu D, et al. Newly diagnosed multiple sclerosis in state of Qatar. Clin Neurol Neurosur. 2013;115(8):1333–1337.
  • Deleu D, Mir D, Al Tabouki A, et al. Prevalence, demographics and clinical characteristics of multiple sclerosis in Qatar. Mult Scler. 2013;19(6):816–819.
  • Kurtzke JF. A reassessment of the distribution of multiple sclerosis – part 1. Acta Neurol Scand. 2009;51(2):110–136.
  • Montalban X, Gold R, Thompson AJ, et al. ECTRIMS/EAN guideline on the pharmacological treatment of people with multiple sclerosis. Eur J Neurol. 2018;25(2):215–237.
  • European Medicines Agency. Guideline on clinical investigation of medicinal products for the treatment of multiple sclerosis [Internet] [cited 2019 Mar]. Available from: http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2015/03/WC500185161.pdf
  • European Medicines Agency. Assessment report. Mavenclad [Internet] [cited 2019 Mar]. Available from: http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Public_assessment_report/human/004230/WC500234563.pdf
  • Gilenya (fingolimod hydrochloride) European Summary of Product Characteristics [Internet] [cited 2019 Mar]. Available from: https://www.medicines.org.uk/emc/product/4545
  • Committee for Medicinal Products for Human Use (CHMP) of the European Medicines Agency. Summary of opinion (post authorisation). Tysabri. 30 May 2013. Natalizumab [Internet] [cited 2019 Mar]. Available from: https://www.ema.europa.eu/documents/smop/chmp-post-authorisation-summary-positive-opinion-tysabri_en-0.pdf
  • Committee for Medicinal Products for Human Use (CHMP) of the European Medicines Agency. Summary of opinion (initial authorisation). Gilenya. Fingolimod. 20 January 2011. EMA/26661/2011 [Internet] [cited 2019 Mar]. Available from: https://www.ema.europa.eu/documents/press-release/meeting-highlights-committee-medicinal-products-human-use-chmp-17-20-january-2011_en.pdf
  • Tysabri (natalizumab) European Summary of Product Characteristics [Internet] [cited 2019 Mar]. Available from: https://www.medicines.org.uk/emc/product/222
  • Zakaria M. Smoke and mirrors: limited value of relative risk reductions for assessing the benefits of disease-modifying therapies for multiple sclerosis. Mult Scler Relat Disord. 2015;4(3):187–191.
  • PRISMS (Prevention of Relapses and Disability by Interferon beta-1a Subcutaneously in Multiple Sclerosis) Study Group. Randomised double-blind placebo-controlled study of interferon beta-1a in relapsing/remitting multiple sclerosis. Lancet. 1998;352:1498–1504.
  • Jacobs LD, Cookfair DL, Rudick RA, et al. Intramuscular interferon beta-1a for disease progression in relapsing multiple sclerosis. The Multiple Sclerosis Collaborative Research Group (MSCRG). Ann Neurol. 1996;39(3):285–294.
  • IFNB Multiple Sclerosis Study Group. Interferon beta-1b is effective in relapsing–remitting multiple sclerosis. I. Clinical results of a multicenter, randomized, double-blind, placebo-controlled trial. Neurology. 1993;43:655–661.
  • Calabresi PA, Kieseier BC, Arnold DL, et al. Pegylated interferon β-1a for relapsing–remitting multiple sclerosis (ADVANCE): a randomised, phase 3, double-blind study. Lancet Neurol. 2014;13(7):657–665.
  • Johnson KP, Brooks BR, Cohen JA, et al. Copolymer 1 reduces relapse rate and improves disability in relapsing–remitting multiple sclerosis: results of a phase III multicenter, double-blind placebo-controlled trial. The Copolymer 1 Multiple Sclerosis Study Group. Neurology. 1995;45(7):1268–1276.
  • Khan O, Rieckmann P, Boyko A, et al. Three times weekly glatiramer acetate in relapsing–remitting multiple sclerosis. Ann Neurol. 2013;73(6):705–713.
  • Giovannoni G, Gold R, Kappos L, et al. BG-12 Increases the proportion of patients free of clinical and radiologic disease activity in relapsing–remitting multiple sclerosis: findings from the DEFINE Study. Neurology. 2012;78(Meeting Abstracts 1):PD5.005.
  • 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(12):1087–1097.
  • Cohen JA, Coles AJ, Arnold DL, 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(9856):1819–1828.
  • Coles AJ, Twyman CL, Arnold DL, et al. Alemtuzumab for patients with relapsing multiple sclerosis after disease-modifying therapy: a randomised controlled phase 3 trial. Lancet. 2012;380(9856):1829–1839.
  • Giovannoni G, Comi G, Cook S; CLARITY Study Group, et al. A placebo-controlled trial of oral cladribine for relapsing multiple sclerosis. N Engl J Med. 2010;362(5):416–426.
  • Giovannoni G, Cook S, Rammohan K, et al. Sustained disease-activity-free status in patients with relapsing–remitting multiple sclerosis treated with cladribine tablets in the CLARITY study: a post-hoc and subgroup analysis. Lancet Neurol. 2011;10(4):329–337.
  • Cohen JA, Barkhof F, Comi G, t al. Oral fingolimod or intramuscular interferon for relapsing multiple sclerosis. N Engl J Med. 2010;362(5):402–415.
  • Kappos L, Radue EW, O’Connor P, et al. A placebo-controlled trial of oral fingolimod in relapsing multiple sclerosis. N Engl J Med. 2010;362(5):387–401.
  • 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(9):899–910.
  • 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(3):254–260.
  • Hauser SL, Bar-Or A, Comi G, et al. Ocrelizumab versus interferon beta-1a in relapsing multiple sclerosis. N Engl J Med. 2017;376(3):221–234.
  • Traboulsee A, Arnold D, Bar-Or A, et al. Ocrelizumab no evidence of disease activity (NEDA) status at 96 weeks in patients with relapsing multiple sclerosis: analysis of the phase III double-blind, double-dummy, interferon beta-1a-controlled OPERA I and OPERA II studies (PL02.004). Neurology. 2016;86(Suppl 1):Abstract PL02:004.
  • Bevan CJ, Cree BA. Disease activity free status: a new end point for a new era in multiple sclerosis clinical research? JAMA Neurol. 2014;71(3):269–270.
  • Sanford M, Lyseng-Williamson KA. Subcutaneous recombinant interferon-β-1a (Rebif): a review of its use in the treatment of relapsing multiple sclerosis. Drugs. 2011;71(14):1865–1891.
  • Bayas A, Rieckmann P. Managing the adverse effects of interferon-beta therapy in multiple sclerosis. Drug Saf. 2000;22(2):149–159.
  • Reder AT, Oger JF, Kappos L, et al. Short-term and long-term safety and tolerability of interferon β-1b in multiple sclerosis. Mult Scler Relat Disord. 2014;3(3):294–302.
  • Carter NJ, Keating GM. Glatiramer acetate: a review of its use in relapsing–remitting multiple sclerosis and in delaying the onset of clinically definite multiple sclerosis. Drugs. 2010;70(12):1545–1577.
  • Ziemssen T, Neuhaus O, Hohlfeld R. Risk–benefit assessment of glatiramer acetate in multiple sclerosis. Drug Saf. 2001;24(13):979–990.
  • Deeks ED. Dimethyl fumarate: a review in relapsing–remitting MS. Drugs. 2016;76(2):243–254.
  • Gold R, Arnold DL, Bar-Or A, et al. Long-term effects of delayed-release dimethyl fumarate in multiple sclerosis: interim analysis of ENDORSE, a randomized extension study. Mult Scler. 2017;23(2):253–265.
  • Mills EA, Ogrodnik MA, Plave A, et al. Emerging understanding of the mechanism of action for dimethyl fumarate in the treatment of multiple sclerosis. Front Neurol. 2018;9:5.
  • Miller AE. Oral teriflunomide in the treatment of relapsing forms of multiple sclerosis: clinical evidence and long-term experience. Ther Adv Neurol Disord. 2017;10(12):381–396.
  • Guarnera C, Bramanti P, Mazzon E. Alemtuzumab: a review of efficacy and risks in the treatment of relapsing remitting multiple sclerosis. TCRM. 2017;13:871–879.
  • Deeks ED. Cladribine tablets: a review in relapsing MS. CNS Drugs. 2018;32(8):785–796.
  • Cook S, Leist T, Comi G, et al. Safety of cladribine tablets in the treatment of patients with multiple sclerosis: an integrated analysis. Mult Scler Relat Disord. 2019;29:157–167.
  • Pakpoor J, Disanto G, Altmann DR, et al. No evidence for higher risk of cancer in patients with multiple sclerosis taking cladribine. Neurol Neuroimmunol Neuroinflamm. 2015;2(6):e158.
  • Rudick R, Polman C, Clifford D, et al. Natalizumab: bench to bedside and beyond. JAMA Neurol. 2013;70(2):172–182.
  • Clerico M, Artusi CA, Liberto AD, et al. Natalizumab in multiple sclerosis: long-term management. IJMS. 2017;18(5):940.
  • Lanza Cariccio V, Bramanti P, Mazzon E. Biomarkers identification for PML monitoring, during natalizumab (Tysabri) treatment in relapsing–remitting multiple sclerosis. Mult Scler Relat Disord. 2018;20:93–99.
  • Ocrevus (ocrelizumab) European Summary of Product Characteristics [Internet] [cited 2019 Mar]. Available from: https://www.medicines.org.uk/emc/product/8898
  • McCool R, Wilson K, Arber M, et al. Systematic review and network meta-analysis comparing ocrelizumab with other treatments for relapsing multiple sclerosis. Mult Scler Relat Disord. 2019;29:55–61.
  • Sorensen PS, Blinkenberg M. The potential role for ocrelizumab in the treatment of multiple sclerosis: current evidence and future prospects. Ther Adv Neurol Disord. 2016;9(1):44–52.
  • Clifford DB, Ances B, Costello C, et al. Rituximab-associated progressive multifocal leukoencephalopathy in rheumatoid arthritis. Arch Neurol. 2011;68(9):1156–1164.
  • Tuccori M, Focosi D, Blandizzi C, et al. Inclusion of rituximab in treatment protocols for non-Hodgkin’s lymphomas and risk for progressive multifocal leukoencephalopathy. Oncologist. 2010;15(11):1214–1219.
  • Behjati M, Etemadifar M, Abdar Esfahani M. Cardiovascular effects of fingolimod: a review article. Iran J Neurol. 2014;13(3):119–126.
  • Druart C, El Sankari S, van Pesch V. Long-term safety and real-world effectiveness of fingolimod in relapsing multiple sclerosis. PROM. 2017;9:1–10.
  • Mandal P, Gupta A, Fusi-Rubiano W, et al. Fingolimod: therapeutic mechanisms and ocular adverse effects. Eye (Lond). 2017;31(2):232–240.
  • Berger JR, Cree BA, Greenberg B, et al. Progressive multifocal leukoencephalopathy after fingolimod treatment. Neurology. 2018;90(20):e1815–e1821.
  • Boyko AN, Boyko OV. Cladribine tablets’ potential role as a key example of selective immune reconstitution therapy in multiple sclerosis. Degener Neurol Neuromuscul Dis. 2018;8:35–44.
  • Willis MD, Robertson NP. Alemtuzumab for the treatment of multiple sclerosis. Ther Clin Risk Manag. 2015;11:525–534.
  • Fyfe I. Multiple sclerosis: immune reconstitution effective for MS. Nat Rev Neurol. 2016;12(8):429.
  • Karussis D, Petrou P. Immune reconstitution therapy (IRT) in multiple sclerosis: the rationale. Immunol Res. 2018;66(6):642.
  • Giovannoni G, Soelberg Sorensen P, Cook S, et al. Safety and efficacy of cladribine tablets in patients with relapsing–remitting multiple sclerosis: results from the randomized extension trial of the CLARITY study. Mult Scler. 2018;24(12):1594–1604.
  • Leist TP, Weissert R. Cladribine: mode of action and implications for treatment of multiple sclerosis. Clin Neuropharmacol. 2011;34(1):28–35.
  • Newsome SD, Aliotta PJ, Bainbridge J, et al. CME/CNE article: a framework of care in multiple sclerosis, part 1: updated disease classification and disease-modifying therapy use in specific circumstances. Int J MS Care. 2016;18(6):314–323.
  • Newsome SD, Aliotta PJ, Bainbridge J, et al. A framework of care in multiple sclerosis, part 2: symptomatic care and beyond. Int J MS Care. 2017;19(1):42–56.
  • Kamm CP, Muehl S, Mircsof D, et al. Role of family planning in women with multiple sclerosis in switzerland: results of the Women With Multiple Sclerosis Patient Survey. Front Neurol. 2018;9:821.
  • Pardo G, Jones DE. The sequence of disease-modifying therapies in relapsing multiple sclerosis: safety and immunologic considerations. J Neurol. 2017;264(12):2351–2374.
  • Havla J, Kleiter I, Kümpfel T. Bridging, switching or drug holidays – how to treat a patient who stops natalizumab? Ther Clin Risk Manag. 2013;9:361–369.
  • Hoepner R, Havla J, Eienbröker C, et al. Predictors for multiple sclerosis relapses after switching from natalizumab to fingolimod. Mult Scler. 2014;20(13):1714–1720.
  • Giovannoni G, Turner B, Gnanapavan S, et al. Is it time to target no evident disease activity (NEDA) in multiple sclerosis? Mult Scler Relat Disord. 2015;4(4):329–333. l
  • Dobson R, Dassan P, Roberts M, et al. UK consensus on pregnancy in multiple sclerosis: ‘Association of British Neurologists’ guidelines. Pract Neurol. 2019;19(2):106–114.
  • Al-Oballi Kridli S, Ilori OM, Verriest HL, et al. Health beliefs and practices related to pregnancy and childcare in Qatar: a qualitative study. J Nurs Educ Pract. 2013;3:1–10.
  • Alroughani R, Altintas A, Al Jumah M, et al. Pregnancy and the use of disease-modifying therapies in patients with multiple sclerosis: benefits versus risks. Mult Scler Int. 2016;2016:1034912.
  • Wundes A, Pebdani RN, Amtmann D. What do healthcare providers advise women with multiple sclerosis regarding pregnancy? Mult Scler Int. 2014;2014:819216.
  • Coyle PK. Management of women with multiple sclerosis through pregnancy and after childbirth. Ther Adv Neurol Disord. 2016;9(3):198–210.
  • Kantor D, Bright JR, Burtchell J. Perspectives from the patient and the healthcare professional in multiple sclerosis: social media and participatory medicine. Neurol Ther. 2018;7(1):37–49.
  • Members of the MS in the 21st Century Steering Group. Unmet needs, burden of treatment, and patient engagement in multiple sclerosis: a combined perspective from the MS in the 21st Century Steering Group. Mult Scler Relat Disord. 2018;19:153–160.
  • Garcia-Dominguez JM, Muñoz D, Comellas M, et al. Patient preferences for treatment of multiple sclerosis with disease-modifying therapies: a discrete choice experiment. PPA. 2016;10:1945–1956.
  • Mansfield C, Thomas N, Gebben D, et al. Preferences for multiple sclerosis treatments: using a discrete-choice experiment to examine differences across subgroups of US patients. Int J MS Care. 2017;19(4):172–183.
  • Lee Mortensen G, Rasmussen PV. The impact of quality of life on treatment preferences in multiple sclerosis patients. PPA. 2017;11:1789–1796.
  • Sellebjerg F, Barnes D, Filippini G, et al. Chapter 27. Acute relapses of multiple sclerosis. In: Gilhus NE, Barnes MR, Brainin E, editors. European Handbook of Neurological Management. 2nd ed. Vol. 1. Hoboken, NJ: Wiley-Blackwell; 2011.
  • National Institute of Health and Care Excellence. Multiple sclerosis. Scenario: managing relapse [Internet]. 2018 [cited 2019 Sep]. Available from: https://cks.nice.org.uk/multiple-sclerosis#!scenario:1
  • Cortese I, Chaudhry V, So YT, et al. Rae-Grant A. Evidence-based guideline update: plasmapheresis in neurologic disorders: report of the Therapeutics and Technology Assessment Subcommittee of the American Academy of Neurology. Neurology. 2011;76(3):294–300.