Skip to content Skip to sidebar Skip to footer

Disease-modifying Treatments for Multiple Sclerosis - a Review of Approved Medications

  • Journal List
  • Wiley-Blackwell Online Open
  • PMC4670697

Eur J Neurol. 2016 Jan; 23(Suppl i): 18–27.

Disease-modifying treatments for multiple sclerosis – a review of approved medications

Ø Torkildsen

aDepartment of Clinical Medicine, KG Jebsen MS Research Middle, Academy of Bergen, Bergen, Norway

bDepartment of Neurology, Norwegian Multiple Sclerosis Competence Centre, Haukeland Academy Hospital, Bergen, Norway

Thou-M Myhr

aDepartment of Clinical Medicine, KG Jebsen MS Enquiry Centre, University of Bergen, Bergen, Norway

cSection of Neurology, Norwegian Multiple Sclerosis Registry and Biobank, Haukeland Academy Infirmary, Bergen, Norway

L Bø

aSection of Clinical Medicine, KG Jebsen MS Inquiry Centre, University of Bergen, Bergen, Kingdom of norway

bSection of Neurology, Norwegian Multiple Sclerosis Competence Middle, Haukeland University Hospital, Bergen, Norway

Find articles by Fifty Bø

Abstract

Background and purpose

There is nevertheless no curative treatment for multiple sclerosis (MS), but during the last 20 years eight dissimilar disease-modifying compounds have been approved for relapsing−remitting MS (RRMS).

Methods

A literature search was conducted on published randomized controlled phase III trials indexed in PubMed on the canonical medications until 21 May 2015.

Results

In this review the fashion of activity, documented handling effects and side furnishings of the approved MS therapies are briefly discussed.

Conclusions

Based on current knowledge of risk−do good of the approved MS medications, including factors influencing adherence, information technology is suggested that oral treatment with dimethyl fumarate or teriflunomide should be preferred as a starting therapy among the beginning-line preparations for de novo RRMS. In the case of breakthrough disease on showtime-line therapy, or quickly evolving severe RRMS, second-line therapy with natalizumab, fingolimod or alemtuzumab should be chosen based on careful risk−benefit stratification.

Keywords: disease-modifying, multiple sclerosis, review, treatment

Introduction

Multiple sclerosis (MS) is a common cause of disability in immature adults. Irreversible axonal damage occurs even in the primeval phases of affliction evolution ane. Although some people with relapsing−remitting MS (RRMS) have a 'benign' illness grade with minimal illness activity and impairment, most patients experience increasing disability over time and somewhen convert to secondary progressive MS (SPMS). There is still no curative treatment, but during the concluding twenty years eight different therapies have go available including interferon beta, glatiramer acetate, teriflunomide, dimethyl fumarate, natalizumab, fingolimod, alemtuzumab and mitoxantrone, and several new compounds are in development. All the approved medications have mainly anti-inflammatory effects and increasing evidence indicates that all of them are more constructive in the early on phases of disease development ii , 3. With the development of more effective treatments, the aim of treatment has changed dramatically in the concluding decades, from merely reducing relapse rates and slowing of disability progression to preventing all show of new disease activity 4. In the current review, the fashion of action and documented result of the current immunomodulatory MS therapies are briefly discussed.

Methods

The commodity is based on English language-language original clinical treatment trials and selected review articles, identified through a literature search in PubMed using the search term 'multiple sclerosis' combined with 'interferon beta', 'glatiramer acetate', 'teriflunomide', 'dimethyl fumarate', 'natalizumab', 'fingolimod', 'mitoxantrone' and 'alemtuzumab'. The search was terminated on 21 May 2015. Titles and abstracts accept been reviewed, and full-text versions of manufactures examined in the bulk of cases. Particular emphasis has been placed on randomized controlled stage III studies.

First-line medications

Interferon beta

Interferon beta is a naturally occurring polypeptide predominantly produced by fibroblasts. Its anti-inflammatory furnishings are largely believed to result from the inhibition of T-lymphocyte proliferation, a shift of cytokine response from an inflammatory response to an anti-inflammatory profile, and reduced migration of inflammatory cells across the blood–brain barrier 5. Interferon beta is available for MS handling in recombinant forms, as interferon beta-1a or interferon beta-1b. Interferon beta-1b is given as a dose of 250 μg subcutaneously every other 24-hour interval; interferon beta-1a is given as a dose of thirty μg intramuscularly once weekly or subcutaneously at doses of 22 or 44 μg three times a calendar week.

Phase III trials of all the interferon beta preparations take shown benign effects in reducing the annualized relapse rate (ARR) by about 30%–34%, reducing the progression of disability in RRMS likewise as magnetic resonance imaging (MRI) disease activity 6 9. A recent report on peginterferon beta-1a, given in one case every 2 weeks, found comparable results, with a reduction in ARR at 36% x. Studies of all interferon beta preparations 11 14 accept likewise reported a reduced gamble of new illness activity amongst people with clinically isolated syndrome (CIS), every bit shown past a significantly prolonged time to a 2nd relapse and reduction in new MRI lesions, in some cases too a delayed progression of disability. All the interferon beta preparations have been evaluated in the treatment of SPMS. The first interferon beta-1b written report showed efficacy of the treatment equally measured past both relapse charge per unit and inability progression 15, just later studies of both interferon beta-1b and interferon beta-1a could only detect handling effects on the relapse rate sixteen 18. Thus, information technology seems that only SPMS patients with superimposed relapses benefit from interferon beta handling xv xviii. Interferons have non been documented to be effective in main progressive MS (PPMS) 19.

Well-nigh patients (50%–75%) experience flu-like symptoms, including muscle aches, fever, chills, headache and back pain, that usually announced 2–8 h afterwards an injection and resolve inside 24 h. Liver enzymes may be elevated and bone marrow function may be depressed, which warrants periodic surveillance of liver function and claret counts before starting therapy and every vi months thereafter xi fourteen. Isolated cases of severe injection-site reactions involving infection or necrosis too as severe cases of acute liver failure and pancreatitis have been reported. Long-fourth dimension exposure to interferon beta does not seem to increase the risk of cancer 20 , 21 or infections.

Interferon beta treatment may induce formation of specific neutralizing antibodies (NABs). NAB germination is less likely during treatment with intramuscular interferon beta-1a 22. The NABs ordinarily announced within half-dozen–eighteen months of treatment, and evidence is accumulating that the efficacy of treatment is reduced in the presence of NABs. Accordingly, it is recommended to test all patients for the presence of NABs every 6 months during the first 2 years of therapy, and treatment should be switched in patients who are confirmed to exist NAB positive 22. In cases of clinically stable affliction, switches to other non-interferon first-line treatments are recommended, simply second-line handling should be considered in cases of quantum disease.

Glatiramer acetate

Glatiramer acetate is a puddle of synthetic peptides, resembling sequences of myelin basic protein, with an average length of 40–100 residues. The mechanisms of activeness have not been fully antiseptic just are probably largely related to anti-inflammatory effects past promoting Th2 difference under the development of Th2 glatiramer acetate reactive CD4+ T cells. These can accumulate in the primal nervous system (CNS) and promote eyewitness suppression by releasing anti-inflammatory cytokines 23. Glatiramer acetate is administered as subcutaneous injections of twenty mg once a day.

Glatiramer acetate treatment trials in RRMS 24 showed a meaning reduction in ARR (29%) and a reduction in gadolinium-enhanced MRI activity 25. In a treatment trial of CIS with silent MRI lesions, glatiramer acetate treatment was found to significantly prolong time to a second relapse and to reduce the risk of new MRI lesions 26. Glatiramer acetate has not been investigated for the treatment of SPMS and has non shown significant do good in PPMS patients 27.

Glatiramer acetate is commonly well tolerated, just virtually patients (65%) feel injection-site reactions (hurting, erythema, swelling and pruritus). About 15% report a transient self-limited systemic reaction (immediately after injection) of facial flushing and chest tightness, accompanied at times past palpitation, anxiety and dyspnoea. Other reported side effects are lymphadenopathy, dyspnoea and lipoatrophy 24 26. Lipoatrophy is permanent and is perhaps the most severe side effect. At that place have non been reports of increased cancer risk or increased risk of infections with prolonged employ of glatiramer acetate.

Teriflunomide

Teriflunomide is an immunomodulatory amanuensis that selectively and reversibly inhibits the mitochondrial enzyme dihydroorotate dehydrogenase, required for de novo pyrimidine synthesis. This leads to reduced proliferation of dividing cells that demand de novo synthesis of pyrimidine to expand. The therapeutic effect in MS is non fully understood but it is probably mediated by a reduced number of circulating lymphocytes 28. Teriflunomide is administered as tablets, xiv mg once daily.

2 phase 3 trials in RRMS 29 , 30 showed that teriflunomide 14 mg once daily, compared to placebo, reduced the ARR by 31%–36%, the charge per unit of inability progression by 26%–27% and MRI gadolinium-enhancing lesions by about 80%. Some other stage Three trial of teriflunomide 14 mg once daily, compared to interferon beta-1a 44 μg subcutaneously three times weekly, showed similar furnishings on the ARR (0.26 and 0.22 respectively) and on time to a new relapse or termination of handling 31. Teriflunomide 14 mg once daily has been tested in a randomized, double-bullheaded, placebo-controlled trial of CIS patients with silent MRI lesions. Teriflunomide treatment was associated with significantly prolonged time to a 2d relapse and a reduction in new MRI lesions 32. Teriflunomide has not been studied for the handling of progressive MS.

Common adverse events include upper respiratory tract infection, urinary tract infection, paraesthesia, diarrhoea, nausea, hair thinning, alanine aminotransferase increase, reduction in claret leucocytes and increase in blood pressure 29 , thirty. Relatively frequent (every second week) alanine aminotransferase screening during the first half dozen months of treatment is recommended and thereafter every second month 29 , thirty. Teriflunomide treatment should be stopped if liver transaminase levels increase three times in a higher place upper normal levels. Regular measurements of claret pressure, white blood cells and platelet counts are also recommended. Teriflunomide has a long half-life. Elimination with cholestyramine or activated charcoal for eleven days tin can accelerate teriflunomide elimination, leading to more than than 98% subtract in teriflunomide plasma concentrations. Liver function needs to be carefully monitored during teriflunomide treatment, and discontinuation of therapy should be considered if a serum transaminase increase more than than three times the upper normal level is confirmed. Rare cases of pancytopenia have been reported with the utilize of leflunomide; this should also lead to treatment termination.

Dimethyl fumarate

Dimethyl fumarate is an immunomodulatory amanuensis with anti-inflammatory backdrop, but the mechanism of action in MS is just partially understood. Pre-clinical studies bespeak that dimethyl fumarate responses are primarily mediated through activation of the nuclear factor (erythroid-derived 2)-like ii (Nrf2) transcriptional pathway. Dimethyl fumarate has also been shown to upregulate Nrf2-dependent antioxidant genes in patients 33. Dimethyl fumarate is administered as a 240 mg capsule twice daily.

2 phase Iii trials of RRMS 34 , 35 showed that dimethyl fumarate 240 mg twice daily, compared to placebo, reduced the ARR past 44%–53%, the charge per unit of disability progression by 22%–32% and MRI gadolinium-enhancing lesions by near 75%–94%. Compared to glatiramer acetate every bit an active comparator in i of the trials 35, dimethyl fumarate 240 mg twice daily reduced the ARR by 24% and the rate of disability progression by 17%. These differences were not pregnant and the study was not powered to observe statistically significant differences in treatment outcome. The number of new and enlarging MRI T2 lesions was significantly reduced past about 36%. Dimethyl fumarate has not been studied for the treatment of CIS or progressive MS.

Mutual adverse events include flushing, nausea, diarrhoea and abdominal hurting 34 , 35. The handling may also reduce white blood cell counts and requite elevations of hepatic transaminases; regular blood tests are therefore recommended 34 , 35. Dimethyl fumarate should be stopped if liver transaminase levels increase iii times above upper normal levels. Recently, a case of John Cunningham virus (JCV) induced progressive multifocal leucoencephalopathy (PML) was reported in a patient who had received dimethyl fumarate 36. An additional four PML cases have been previously reported in psoriasis patients who had received fumaderm 37. Prolonged severe lymphopaenia (<500 cells per cubic millimetre) that persists for more than 6 months has been suggested every bit a gamble factor for PML. In the case of persistent lymphopaenia, dimethyl fumarate should exist terminated in JCV-positive patients.

Second-line medications

Fingolimod

Fingolimod is an oral sphingosine 1-phosphate receptor (S1PR) modulator that subsequent to its phosphorylation binds with high affinity to S1PR, which in turn leads to an internalization and degradation of the receptor in unlike tissues and prison cell types, including lymphocytes. As a consequence, fingolimod inhibits the power of autoreactive lymphocytes to egress from the lymph nodes towards the CNS. Fingolimod 0.5 mg capsules are given orally once daily 38.

Ii phase Three trials in RRMS 38 , 39 showed that fingolimod 0.v mg one time daily, compared to placebo, reduced the ARR by 48%–55%, the rate of disability progression past 25%–30% and MRI gadolinium-enhancing lesions by more than than eighty%. Another study comparing fingolimod 0.5 mg one time daily to interferon beta-1a 30 μg intramuscularly one time weekly showed a reduced ARR by 52%, a reduced charge per unit of disability progression by 25% and a reduced number of MRI gadolinium-enhancing lesions by more than than fifty% amongst those who received fingolimod twoscore. Fingolimod is currently not documented to be effective confronting CIS, SPMS or PPMS.

Common adverse events include upper respiratory tract infection, headache, coughing, diarrhoea and back pain 38 , 39. Fingolimod may also crusade a transient bradycardia and atrioventricular block. It is therefore recommended to monitor patients continuously with an electrocardiogram for six h after the first dose, and to extend the monitoring of patients who develop specific clinically relevant signs of heart arrhythmia (http://www.fda.gov/Drugs/DrugSafety/ucm303192.htm). More often than not, fingolimod should not be used by patients with known cardiac arrhythmias or patients using other medications known to induce bradycardia. Rare adverse events of elevated liver enzymes and macular oedema may occur, and regular claret sampling and a routine eye examination later on 3 months of treatment are therefore recommended. Ane decease due to a fulminant primary varicella zoster infection was reported in one of the phase Three trials twoscore. Therefore a blood sample for screening of previous varicella zoster infection is advised, and in the example of a negative screening test vaccination is recommended prior to treatment initiation.

Natalizumab

Natalizumab is a monoclonal antibody against α4-integrin, blocking the interaction with its ligands. The machinery of action is largely through preventing adherence of activated leucocytes to inflamed endothelium, thus inhibiting the migration of inflammatory cells into the CNS. Natalizumab is administered as a 300 mg intravenous infusion every iv weeks 41.

The pivotal phase III trial of RRMS showed that natalizumab monotherapy reduced the ARR by 68%, the rate of disability progression by 54% and MRI gadolinium-enhancing lesions by more than than 90% compared to placebo 41. Another written report 42 constitute that treatment with natalizumab added to interferon beta-1a was significantly more effective than interferon beta-1a alone in reducing ARR, new T2 lesions and disability progression. Natalizumab is currently not documented to be effective against CIS, SPMS or PPMS.

Although natalizumab is more often than not well tolerated, the treatment is associated with an increased run a risk of developing PML 43. This is a potentially life-threatening CNS infection of oligodendrocytes by the JCV. Therefore all patients receiving natalizumab should be screened for previous JCV infection. The risk for PML in JCV-negative patients is low (<0.09/thou) and is probably associated with recent seroconversion (estimated as 2%–3% each year) or a false negative test. Amongst the JCV-positive patients the take chances of developing PML is influenced by treatment duration and previous immunosuppressive handling. The chance is relatively depression during the start 2 years of treatment and increases thereafter. The highest run a risk is found amongst JCV-positive patients who previously have too received immunosuppressive treatment after 2 years of handling (∼1/60) 44. Anti-JCV antibody levels seem besides to differentiate PML risk in anti-JCV antibody positive patients with no prior immunosuppressant utilise 45. Equally a general rule, it is recommended that JCV-positive patients who take been treated with natalizumab for more than 2 years should be switched to another 2d-line therapy. Based on current cognition, a washout time of 8 weeks seems to reduce the risk of rebound effect compared to longer washout periods. In the case of a depression JCV index (<i.5), natalizumab treatment may in some cases be continued after thorough information is given to the patient and under careful evaluation for new symptoms that may stand for PML 44. Three-monthly JCV alphabetize evaluation and MR examination is then recommended. It is recommended to retest JCV-negative patients every 6 months and JCV-positive patients should be carefully informed about the risk for PML at handling initiation and after 2 years of handling.

Natalizumab treatment may induce an immune response, with the formation of persistent NABs (∼4%–half dozen%) confronting the preparation. NABs ordinarily appear within the first 12 months of handling, reduce the efficacy of the handling and are associated with higher rates of infusion-related adverse events. Accordingly, patients should be tested for NABs at 6 and 12 months of therapy and after for infusion-related agin events or treatment failure. NABs tin can occur transiently and positive findings should therefore exist confirmed within 3 months before deciding to switch therapy. Testing can be discontinued in patients who remain NAB negative during the first year of therapy.

Alemtuzumab

Alemtuzumab is a recombinant, humanized monoclonal antibiotic directed against CD52, a jail cell surface antigen present at high levels on especially T and B lymphocytes. Alemtuzumab acts through antibody-dependent cellular cytolysis and complement-mediated lysis following prison cell surface bounden. The mechanism by which alemtuzumab exerts its therapeutic effects in MS is suggested to be by a depletion and repopulation of lymphocytes that reduces the potential for relapses and thereby delays disease progression 46. Alemtuzumab is administered by intravenous infusion for 2 handling courses. The initial handling course is 12 mg/solar day for v consecutive days (60 mg full dose), and the 2d handling course is 12 mg/day for three consecutive days (36 mg full dose) administered 12 months afterwards the initial treatment course. Boosted courses may exist given 12 months later the latest treatment course if necessary. Based on the European Medicines Bureau (EMA) licence alemtuzumab has indication every bit a first-line medication in agile RRMS. Because the treatment increases the chance of secondary autoimmunity, most European neurologists would utilize this drug as a second-line grooming, yet.

Two phase III trials of RRMS accept shown that alemtuzumab 12, compared to interferon beta-1a 44 μg administered subcutaneously iii times weekly, reduced the ARR by 49%–55%, the charge per unit of disability progression by 30%–42% and MRI gadolinium-enhancing lesions by 61%–63% 47 , 48. Alemtuzumab has currently non been studied in patients with CIS or PPMS and has not been demonstrated to be effective in SPMS 49 , 50.

Patients usually experience infusion-associated reactions including flushing, nausea, headache, tachycardia, urticaria, rash, pruritus, pyrexia and fatigue 47 50. Oral antiviral prophylaxis with aciclovir 200 mg twice daily (or equivalent) should be administered and continued for a minimum of i month afterward the last dose. Alemtuzumab treatment is associated with increased risk of upper respiratory tract infection and urinary tract infection. Alemtuzumab treatment may also consequence in the formation of autoantibodies and increased adventure of autoimmune-mediated weather condition (occurring a median of 32 months after the first treatment), including thyroid disorders (41%), immune thrombocytopenic purpura (3.5%) or, rarely, nephropathies (due east.g. anti-glomerular basement membrane disease) (<ane%) 51. Based on the risk of autoimmune-mediated conditions, monthly blood and urine analyses are recommended for iv years subsequently the last dosing of alemtuzumab.

Mitoxantrone

Mitoxantrone is a synthetic anthracenedione derivative and is mostly used in treating various malignancies. It interacts with nuclear Deoxyribonucleic acid and is a potent immunosuppressive agent targeting proliferating allowed cells, inhibiting proliferation and inducing apoptosis of T lymphocytes, B lymphocytes, macrophages and other antigen-presenting cells.

Express efficacy information are bachelor, but controlled studies of highly active RRMS take shown significant efficacy of the handling, as shown by a 60%–70% reduction in the relapse rate (compared with placebo or intravenous methylprednisolone) every bit well as reduced inability progression and MRI disease action 52 , 53. The largest phase 3 investigator-blinded written report randomized patients with worsening RRMS and SPMS for 5 or 12 mg of mitoxantrone per square metre of trunk surface or placebo every iii months for 2 years 54. The treatment showed a 66% reduction in the ARR in the high-dose arm compared with placebo, and reduced disability progression and MRI disease activeness. Mitoxantrone has not been included in treatment trials of patients with CIS or PPMS.

Side effects such equally transient nausea, fatigue, mild hair loss (for days to a calendar week) and menstrual disturbances are frequent (lx%–70%) 54. Additional side effects are urinary tract infection (about xxx%) as well as elevated liver enzymes and leucopenia (about 15%–20%). Mitoxantrone-induced amenorrhoea and acute promyelocytic leukaemia take likewise been reported. The treatment induces transient leucopenia, with a nadir later on almost 10 days, and thus follow-up blood command is needed. Although not in the phase Iii trial, lethal congestive heart failure and therapy-related leukaemia take been reported, even years later on treatment ends 55 , 56. Due to the potential cardiotoxicity, the maximum cumulative dose is restricted to 120–140 mg/yard2 of torso surface, and echocardiograms should be done earlier, during and after treatment. Mitoxantrone is teratogenic and is absolutely contraindicated in pregnancy. The apply of mitoxantrone has rapidly decreased due to the risk of astringent complications and the increasing number of alternative highly effective and less toxic treatment options.

Suggested handling strategies

Individualized therapy is advocated; the ideal treatment option would be the safest treatment that eliminates clinical and radiological evidence of disease action 3. Most patients would start on a first-line therapy only then be inverse quickly to a 2nd-line medication in the case of quantum disease activity (Fig. one). Testify of clinical disease activity (relapses and/or accumulating inability) with or without new MRI lesions is in general accepted as an indication for switching to more potent 2nd-line therapies. In this context, models like the Rio or modified Rio score 57 have been increasingly accepted every bit a tool to monitor the handling issue. The score is based on evaluation of treatment response by the combination of clinical (relapse and disability progression) and MRI disease activity (Rio score) or relapse and MRI affliction activeness only (modified Rio score) during the first twelvemonth of interferon treatment. Increasing evidence indicates that patients experiencing a new clinical relapse with significant influence on disability and/or new signs of radiological disease activity (≥3 active MRI lesions) during the concluding year, whilst on outset-line medications, should be considered for switching to more than potent medications 58 , 59.

An external file that holds a picture, illustration, etc.  Object name is ene0023-0018-f1.jpg

Treatment algorithm for handling-naïve patients with RRMS. *Patients with quickly evolving severe RRMS should first directly on a second-line therapy. Breakthrough illness action is defined as one new clinical relapse with meaning influence on disability and/or new signs of radiological disease activity (≥3 active MRI lesions) during the terminal year whilst on commencement-line medication.

Another possible treatment strategy is induction handling, which consists of early on apply of immunosuppressive medications followed by long-term maintenance therapy sixty. This treatment government has been used with success for patients with aggressive RRMS, using mitoxantrone 61. The use of mitoxantrone is declining because of its long-term safety profile. Alemtuzumab is approved as a first-line therapy, and could besides be considered an consecration treatment because of its long-term effects on the immune arrangement. This could be an attractive handling selection for patients with a highly active disease course, as 70.1% of the alemtuzumab-treated patients in the Intendance-MS written report remained costless of new lesions and MRI activity in year four, despite most receiving their last treatment course 3 years prior 62.

Choosing amongst the beginning-line medications

The main findings from the pivotal and phase 3 studies performed are given in Tables i and 2. Although the placebo-controlled trials indicate numerically college efficacy on ARR from dimethyl fumarate (about 44%–53%) compared to the other outset-line preparations (about xxx%–35%), information on direct comparisons of the agents are limited. Head-to-head comparison between dimethyl fumarate and glatiramer acetate indicated numerically (although not statistically significant) better event from dimethyl fumarate 35. Head-to-head comparison between teriflunomide and high-frequency interferon beta-1a showed comparable effects on the ARR 29. Head-to-caput comparisons between intramuscular depression-dose and low-frequency interferon beta-1a and subcutaneous high-dose and loftier-frequency interferon beta-1a and interferon beta-1b take shown that loftier-dose and high-frequency interferon beta regimens have short-term benefits on the relapse charge per unit and MRI activity 63 , 64. Limitations in the blueprint of these studies have been widely discussed, however, and the long-term differences in efficacy may be reduced by a significantly lower frequency of NAB germination with low-dose and low-frequency interferon beta-1a. Caput-to-head comparisons of glatiramer acetate and subcutaneous loftier-dose and high-frequency interferon beta-1a and interferon beta-1b have shown similar clinical benefit from the treatments, with some MRI parameters in favour of the interferon beta preparations 65 , 66.

Table 1

Randomized placebo-controlled stage III clinical trials of the canonical relapsing−remitting multiple sclerosis medications

Medication N Trial name (reference) ARR Disability progression
Relative reduction ARR Relative reduction EDSS progression
First-line
 Interferon beta-1b 124 vs. 123 MSSG 6 34% 0.84 vs. 1.27 29% (Due north.S.) 0.20 vs. 0.28
 Interferon beta-1a i.chiliad. 158 vs. 143 MSCRG seven 18% 0.67 vs. 0.82 37% 0.22a vs. 0.35a
 Interferon beta-1a s.c. 184 vs. 187 PRISMS eight 32% 1.73 vs. 2.56 32% 0.26 vs. 0.38
 Peginterferon-1a 500 vs. 512 Advance 10 36% 0.26 vs. 0.40 36% 0.07 vs. 0.11
 Glatiramer acetate 125 vs. 126 CMSSG 24 29% ane.19 vs. 1.68 12% (Due north.Southward.) 0.22 vs. 0.25
 Teriflunomide 358 vs. 363 TEMSO 29 31% 0.37 vs. 0.54 26% 0.20 vs. 0.27
 Teriflunomide 370 vs. 388 TOWER xxx 36% 0.32 vs. 0.50 24% 0.sixteen vs. 0.21
 Dimethyl fumarate 410 vs. 408 Define 34 53% 0.17 vs. 0.36 41% 0.xvi vs. 0.27
 Dimethyl fumarate 359 vs. 363 CONFIRM 35 44% 0.22 vs. 0.40 24% (North.S.) 0.13 vs. 0.17
Second-line
 Fingolimod 425 vs. 418 FREEDOMS 38 55% 0.18 vs. 0.40 28% 0.18 vs. 0.25
 Fingolimod 358 vs. 355 FREEDOMS-two 39 48% 0.21 vs. 0.40 xiv% (N.S.) 0.25 vs. 0.29
 Natalizumab 627 vs. 315 Affirm 41 68% 0.23 vs. 0.73 42% 0.17 vs. 0.29
 Mitoxantroneb lx vs. 64 MIMS 54 66% 0.35 vs. ane.02 64% 0.08 vs. 0.22

Table 2

Randomized controlled phase Three clinical trials of the canonical relapsing−remitting multiple sclerosis medications, where the medications have been compared head-to-caput with another active multiple sclerosis medication

Medication Compared to Northward Trial proper noun (reference) ARR Disability progression
Relative reduction ARR Relative reduction EDSS progression
Get-go-line
 Interferon beta-1b Interferon beta-1a i.m. 92 vs. 96 INCOMIN 63 24% 0.v vs. 0.7 44% 0.xiii vs. 0.30
 Interferon-beta-1a s.c. Interferon-beta-1a i.m. 339 vs. 338 EVIDENCE 64 xvi%a 0.54 vs. 0.64 xiii% (N.S.) 0.xiii vs. 0.15
 Interferon beta-1a s.c. Glatiramer acetate 386 vs. 378 REGARD 65 3% (Due north.S) 0.30 vs. 0.29 25% (N.S.) 0.12 vs. 0.09
 Interferon-beta-1b Glatiramer acetate 899 vs. 448 BEYOND 66 3% (N.S) 0.33 vs. 0.34 5% (Due north.Southward.) 0.22 vs. 0.xx
 Teriflunomide Interferon-beta 1a southward.c. 111 vs. 104 TENERE 31 four% (N.S) 0.26 vs. 0.22 - -
 Dimethyl fumarate Glatiramer acetate 359 vs. 350 CONFIRM 35 24% (N.S) 0.22 vs. 0.29 17% (N.Southward.) 0.13 vs. 0.16
Second-line
 Fingolimod Interferon beta-1a i.m. 431 vs. 435 TRANSFORMS 40 52%a 0.16 vs. 0.33 25% (N.Due south.) 0.06 vs. 0.08
 Alemtuzumab Interferon beta-1a south.c. 376 vs. 202 CARE MS-one 47 55% 0.18 vs. 0.39 thirty% (N.S.) 0.08b vs. 0.11b
 Alemtuzumab Interferon beta-1a s.c. 426 vs. 202 Intendance MS-2 48 49% 0.26 vs. 0.52 42% 0.13b vs. 0.21b

Disease-modifying handling for MS is a long-lasting therapy for most patients. Adherence to treatment is thus crucial, and many patients may therefore adopt oral treatment. Consequently starting with an oral first-line drug is suggested. In the example of intolerability or unacceptable side effects, switching between the oral preparations or with 1 of the injectable preparations should exist considered. The injectable medications take been used for a longer period than the oral medications, and more long-term safety data are therefore available 20 , 21. Similarly, there are more long-term prophylactic data on pregnancies occurring during treatments with both glatiramer acetate and interferons, pointing to a relative safety of utilize 67. These could be good reasons for still choosing an injectable medication as beginning-line treatment. Information technology is of import to continuously evaluate the treatment regimen, aiming for optimal adherence, because both the administration form and side-effect profiles.

Choosing amidst the 2nd-line medications

In the case of breakthrough disease activity despite a full and adequate grade of a first-line training, switching to natalizumab, fingolimod or alemtuzumab should be considered. Although alemtuzumab is licensed as a first-line medication in active RRMS, many European neurologists would use this drug as a second-line preparation, due to potential side furnishings. Second-line therapy should also be considered in the case of patients with rapidly evolving severe RRMS defined by two or more disabling relapses in 1 year, and with gadolinium-enhancing lesions on brain MRI or a meaning increase in T2 lesion load compared to a previous recent MRI. Careful risk stratification for potential adverse effects is important, and most neurologists would prefer fingolimod or alemtuzumab for patients who are JCV positive. Similarly, in the instance of contraindications for fingolimod or alemtuzumab, ane of the other second-line treatment options should be considered. The use of mitoxantrone has become less frequent due to the relatively high adventure of serious side effects, and may only exist used in some cases of SPMS. Stratification for differences in clinical issue is hard due to the lack of treatment studies with head-to-head comparisons of second-line therapies. Some neurologists would prefer natalizumab for JCV-negative patients due to the numerically college reduction of ARR in pivotal trials, although treatment effect cannot be direct compared between different study populations.

For a small grouping of patients who do not reply to the approved 2nd-line treatments, off-label treatments like rituximab 68 or ofatumumab 69 or experimental therapy with autologous haematopoietic stalk cell transplantation 70 may be considered. These treatment options have currently non been tested in large phase 3 trials, but phase II trials or instance series reports have shown promising results. These treatment options besides seem to exist effective against RRMS and non the progressive forms of the illness 71.

Conclusions and future challenges

Although the last decade has shown a revolution in treatment options for patients with MS, this has mainly benefited newly diagnosed patients with an RRMS disease class. None of the approved medications or experimental therapies has shown convincing evidence of slowing down or preventing disease progression in patients with SPMS or PPMS. Thus there is an urgent demand to also amend the handling options for patients who take entered a progressive phase.

Disclosure of conflicts of involvement

Ø. Torkildsen has participated on a scientific advisory board for Biogen Idec, Merck-Serono and Genzyme and received speaker honoraria and travel grants from Genzyme, Merck-Serono, Novartis and Biogen Idec. G.-Chiliad. Myhr has participated on scientific informational boards for Novartis Norway, Biogen Idec and Genzyme; received funding for travel from Bayer, Novartis, Merck-Serono and Biogen Idec; received speaker honoraria from Bayer, Genzyme, Sanofi-Aventis, Novartis, Merck-Serono and Biogen Idec; and received unrestricted research back up from Bayer, Sanofi-Aventis, Novartis, Merck-Serono, Biogen Idec, Pronova Biocare and the Norwegian MS Social club. 50. Bø has participated on scientific advisory boards for Novartis Norway; received funding for travel from Sanofi-Aventis, Novartis, Merck-Serono and Biogen Idec; received speaker honoraria from Bayer, Genzyme, Sanofi-Aventis, Novartis, Merck-Serono and Biogen Idec; and received unrestricted research support from Bayer, Sanofi-Aventis, Novartis, Merck-Serono and Biogen Idec.

References

  • Trapp BD, Peterson J, Ransohoff RM, Rudick R, Mörk South, Bö L. Axonal transection in the lesions of multiple sclerosis. Due north Engl J Med. 1998;338:278–285. [PubMed] [Google Scholar]
  • Coles AJ, Cox A, Le Page Due east, et al. The window of therapeutic opportunity in multiple sclerosis: evidence from monoclonal antibiotic therapy. J Neurol. 2006;253:98–108. [PubMed] [Google Scholar]
  • Hauser SL, Chan JR, Oksenberg JR. Multiple sclerosis: prospects and hope. Ann Neurol. 2013;74:317–327. [PubMed] [Google Scholar]
  • Havrdova E, Galetta S, Stefoski D, Comi G. Liberty from disease activity in multiple sclerosis. Neurology. 2010;74(Suppl. iii):S3–S7. [PubMed] [Google Scholar]
  • Dhib-Jalbut S, Marks S. Interferon-β mechanisms of action in multiple sclerosis. Neurology. 2010;74(Suppl. one):S17-24. [PubMed] [Google Scholar]
  • The IFNB Multiple Sclerosis Written report Grouping. 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. [PubMed] [Google Scholar]
  • Jacobs LD, Cookfair DL, Rudick RA, et al. Intramuscular interferon beta1a for disease progression in relapsing multiple sclerosis. Ann Neurol. 1996;39:285–294. [PubMed] [Google Scholar]
  • Ebers GC PRISMS (Prevention of Relapses Disability by Interferon β-1a Subcutaneously in Multiple Sclerosis) Study Group. Randomised double-blind placebo controlled report of interferon β-1a in relapsing/remitting multiple sclerosis. Lancet. 1998;352:1498–1504. [PubMed] [Google Scholar]
  • PRISMS Study Group and the Academy of British Columbia MS/MRI Analysis Group. PRISMS4: Long-term efficacy of interferon-β-1a in relapsing MS. Neurology. 2001;56:1628–1636. [PubMed] [Google Scholar]
  • 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;13:657–665. Jul; [PubMed] [Google Scholar]
  • Kappos L, Polman CH, Freedman MS, et al. Handling with interferon beta-1b delays conversion to clinically definite and McDonald MS in patients with clinically isolated syndromes. Neurology. 2006;67:1242–1249. [PubMed] [Google Scholar]
  • Jacobs LD, Beck RW, Simon JH, et al. Intramuscular interferon beta-1a therapy initiated during a commencement demyelinating consequence in multiple sclerosis. Northward Engl J Med. 2000;343:898–904. [PubMed] [Google Scholar]
  • Comi G, Filippi M, Barkhof F, et al. Effect of early interferon handling on conversion to definite multiple sclerosis: a randomised study. Lancet. 2001;357:1576–1582. [PubMed] [Google Scholar]
  • Comi G, De SN, Freedman MS, et al. Comparing of ii dosing frequencies of subcutaneous interferon beta-1a in patients with a beginning clinical demyelinating event suggestive of multiple sclerosis (REFLEX): a phase 3 randomised controlled trial. Lancet Neurol. 2012;eleven:33–41. . Erratum in: Lancet Neurol 2012; eleven: 125. [PubMed] [Google Scholar]
  • Kappos L European Study Grouping on Interferon β-1b in Secondary Progressive MS. Placebo-controlled multicentre randomised trial of interferon β-1b in treatment of secondary progressive multiple sclerosis. Lancet. 1998;352:1491–1497. [PubMed] [Google Scholar]
  • Secondary Progressive Efficacy Clinical Trial of Recombinant Interferon-Beta-1a in MS (SPECTRIMS) Written report Group. Randomized controlled trial of interferon-beta-1a in secondary progressive MS. Clinical results. Neurology. 2001;56:1496–1504. [PubMed] [Google Scholar]
  • Cohen JA, Cutter GR, Fischer JS, et al. Benefit of interferon β-1a on MSFC progression in secondary progressive MS. Neurology. 2002;59:679–687. [PubMed] [Google Scholar]
  • Panitch H, Miller A, Paty D, et al. Interferon beta-1b in secondary progressive MS: results from a 3-year controlled report. Neurology. 2004;63:1788–1795. [PubMed] [Google Scholar]
  • Leary SM, Miller DH, Stevenson VL, et al. Interferon β-1a in primary progressive MS: an exploratory, randomized, controlled trial. Neurology. 2003;lx:44–51. [PubMed] [Google Scholar]
  • Sandberg-Wollheim M, Kornmann G, Bischof D, Moraga MS, Hennessy B, Alteri Eastward. The risk of malignancy is not increased in patients with multiple sclerosis treated with subcutaneous interferon beta-1a: analysis of data from clinical trial and post-marketing surveillance settings. Mult Scler. 2011;17:431–440. [PubMed] [Google Scholar]
  • Kingwell Eastward, Evans C, Zhu F, Oger J, Hashimoto Due south, Tremlett H. Assessment of cancer risk with β-interferon treatment for multiple sclerosis. J Neurol Neurosurg Psychiatry. 2014;85:1096–1102. [PubMed] [Google Scholar]
  • Sørensen PS, Deisenhammer F, Duda P, et al. Guidelines on utilise of anti-IFN-β antibody measurements in multiple sclerosis: written report of an EFNS Task Force on IFN-β antibodies in multiple sclerosis. Eur J Neurol. 2005;12:817–827. [PubMed] [Google Scholar]
  • Racke MK, LovettRacke AE, Karandikar NJ. The machinery of action of glatiramer acetate treatment in multiple sclerosis. Neurology. 2010;74(Suppl. 1):S2530. [PubMed] [Google Scholar]
  • Johnson KP, Brooks BR, Cohen JA, et al. Copolymer 1 reduces relapse rate and improves inability in relapsing−remitting multiple sclerosis: results of a phase Iii multicenter, double-blind, placebo-controlled trial. Neurology. 1995;45:1268–1276. [PubMed] [Google Scholar]
  • Comi Thou, Filippi M, Wolinsky JS. European/Canadian multicenter, double-blind, randomized, placebo-controlled written report of the effects of glatiramer acetate on magnetic resonance imaging – measured disease action and burden in patients with relapsing multiple sclerosis. European/Canadian Glatiramer Acetate Study Grouping. Ann Neurol. 2001;49:290–297. [PubMed] [Google Scholar]
  • Comi M, Martinelli V, Rodegher Chiliad, et al. Issue of glatiramer acetate on conversion to clinically definite multiple sclerosis in patients with clinically isolated syndrome (PreCISe report): a randomised, double-bullheaded, placebo-controlled trial. Lancet. 2009;374:150311. [PubMed] [Google Scholar]
  • Wolinsky JS, Narayana PA, O'Connor P, et al. Glatiramer acetate in primary progressive multiple sclerosis: results of a multinational, multicenter, double-blind, placebo-controlled trial. Ann Neurol. 2007;61:14–24. [PubMed] [Google Scholar]
  • Papadopoulou A, Kappos L, Sprenger T. Teriflunomide for oral therapy in multiple sclerosis. Proficient Rev Clin Pharmacol. 2012;v:617–628. November;. doi: x.1586/ecp.12.56. [PubMed] [Google Scholar]
  • O'Connor P, Wolinsky JS, Confavreux C, et al. Randomized trial of oral teriflunomide for relapsing multiple sclerosis. N Engl J Med. 2011;365:1293–1303. . doi: 10.1056/NEJMoa1014656. [PubMed] [Google Scholar]
  • Confavreux C, O'Connor P, Comi G, et al. Oral teriflunomide for patients with relapsing multiple sclerosis (TOWER): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Neurol. 2014;13:247–256. . doi: 10.1016/S1474-4422(13)70308-nine. Epub 2014 Jan 23. [PubMed] [Google Scholar]
  • 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. 2014;20:705–716. [PubMed] [Google Scholar]
  • Miller AE, Wolinsky JS, Kappos L, et al. Oral teriflunomide for patients with a first clinical episode suggestive of multiple sclerosis (TOPIC): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Neurol. 2014;xiii:977–986. [PubMed] [Google Scholar]
  • Linker RA, Golden R. Dimethyl fumarate for treatment of multiple sclerosis: mechanism of activity, effectiveness, and side effects. Curr Neurol Neurosci Rep. 2013;thirteen:394. [PubMed] [Google Scholar]
  • Gold R, Kappos Fifty, Arnold DL, et al. Placebo-controlled phase 3 study of oral BG-12 for relapsing multiple sclerosis. North Engl J Med. 2012;367:1098–1107. [PubMed] [Google Scholar]
  • Fox RJ, Miller DH, Phillips JT, et al. Placebo-controlled phase 3 written report of oral BG-12 or glatiramer in multiple sclerosis. North Engl J Med. 2012;367:1087–1097. [PubMed] [Google Scholar]
  • Rosenkranz T, Novas One thousand, Terborg C. PML in a patient with lymphocytopenia treated with dimethyl fumarate. N Engl J Med. 2015;372:1476–1478. [PubMed] [Google Scholar]
  • Sweetser MT, Dawson KT, Bozic C. Manufacturer's response to case reports of PML. Northward Engl J Med. 2013;368:1659–1661. [PubMed] [Google Scholar]
  • Kappos Fifty, Radue EW, O'Connor P, et al. A placebo controlled trial of oral fingolimod in relapsing multiple sclerosis. North Engl J Med. 2010;362:387401. [PubMed] [Google Scholar]
  • Calabresi PA, Radue EW, Goodin D, et al. Safety and efficacy of fingolimod in patients with relapsing−remitting multiple sclerosis (FREEDOMS II): a double-blind, randomised, placebo-controlled, stage iii trial. Lancet Neurol. 2014;13:545–556. [PubMed] [Google Scholar]
  • Cohen JA, Barkhof F, Comi K, et al. Oral fingolimod or intramuscular interferon for relapsing multiple sclerosis. N Engl J Med. 2010;362:40215. [PubMed] [Google Scholar]
  • 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. [PubMed] [Google Scholar]
  • Rudick RA, Stuart WH, Calabresi PA, et al. Natalizumab plus interferon beta-1a for relapsing multiple sclerosis. N Engl J Med. 2006;354:911–923. [PubMed] [Google Scholar]
  • Clifford DB, De Luca A, Simpson DM, Arendt Grand, Giovannoni Yard, Nath A. Natalizumab-associated progressive multifocal leukoencephalopathy in patients with multiple sclerosis: lessons from 28 cases. Lancet Neurol. 2010;ix:438–446. [PubMed] [Google Scholar]
  • Bloomgren G, Richman S, Hotermans C, et al. Risk of natalizumab-associated progressive multifocal leukoencephalopathy. N Engl J Med. 2012;366:1870–1880. [PubMed] [Google Scholar]
  • Plavina T, Subramanyam G, Bloomgren G, et al. Anti-JC virus antibody levels in serum or plasma further define take a chance of natalizumab-associated progressive multifocal leukoencephalopathy. Ann Neurol. 2014;76:802–812. [PMC free article] [PubMed] [Google Scholar]
  • Brown JW, Coles AJ. Alemtuzumab: evidence for its potential in relapsing−remitting multiple sclerosis. Drug Des Devel Ther. 2013;7:131–138. . doi: ten.2147/DDDT.S32687. Epub 2013 Mar 6. [PMC free article] [PubMed] [Google Scholar]
  • 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;380:1819–1828. [PubMed] [Google Scholar]
  • Coles AJ, Twyman CL, Arnold DL, et al. Alemtuzumab for patients with relapsing multiple sclerosis after affliction-modifying therapy: a randomised controlled phase 3 trial. Lancet. 2012;380:1829–1839. [PubMed] [Google Scholar]
  • Coles AJ, Wing MG, Molyneux P, et al. Monoclonal antibody treatment exposes three mechanisms underlying the clinical course of multiple sclerosis. Ann Neurol. 1999;46:296–304. [PubMed] [Google Scholar]
  • Paolillo A, Coles AJ, Molyneux PD, et al. Quantitative MRI in patients with secondary progressive MS treated with monoclonal antibody Campath 1H. Neurology. 1999;53:751–757. [PubMed] [Google Scholar]
  • Tuohy O, Costelloe Fifty, Colina-Cawthorne G, et al. Alemtuzumab treatment of multiple sclerosis: long-term safety and efficacy. J Neurol Neurosurg Psychiatry. 2015;86:208–215. [PubMed] [Google Scholar]
  • Millefiorini E, Gasperini C, Pozzilli C, et al. Randomized placebo-controlled trial of mitoxantrone in relapsing−remitting multiple sclerosis: 24 calendar month clinical and MRI outcome. J Neurol. 1997;244:153–159. [PubMed] [Google Scholar]
  • Martinelli V, Radaelli Thou, Straffi L, et al. Mitoxantrone: benefits and risks in multiple sclerosis patients. Neurol Sci. 2009;30(Suppl. 2):S167–170. [PubMed] [Google Scholar]
  • Hartung HP, Gonsette R, König N, et al. Mitoxantrone in progressive multiple sclerosis: a placebo controlled, double-blind, randomised, multicentre trial. Lancet. 2002;360:2018–2025. [PubMed] [Google Scholar]
  • Martinelli V, Cocco East, Capra R, et al. Acute myeloid leukemia in Italian patients with multiple sclerosis treated with mitoxantrone. Neurology. 2011;77:1887–1895. [PubMed] [Google Scholar]
  • Ellis R, Brown Southward, Boggild M. Therapy-related acute leukaemia with mitoxantrone: four years on, what is the risk and can it be limited? Mult Scler. 2015;21:642–645. [PubMed] [Google Scholar]
  • Sormani MP, Rio J, Tintorè Thou, et al. Scoring treatment response in patients with relapsing multiple sclerosis. Mult Scler. 2013;19:605–612. [PubMed] [Google Scholar]
  • Río J, Rovira A, Tintoré G, et al. Relationship betwixt MRI lesion activity and response to IFN-β in relapsing−remitting multiple sclerosis patients. Mult Scler. 2008;fourteen:479–484. [PubMed] [Google Scholar]
  • Bermel RA, You X, Foulds P, et al. Predictors of long-term outcome in multiple sclerosis patients treated with interferon β Ann Neurol. 2013;73:95–103. [PubMed] [Google Scholar]
  • Edan G, Le Page Due east. Induction therapy for patients with multiple sclerosis: Why? When? How? CNS Drugs. 2013;27:403–409. [PMC gratuitous commodity] [PubMed] [Google Scholar]
  • Edan M, Comi G, Le PE, Leray Due east, Rocca MA, Filippi M French–Italian Mitoxantrone Interferon-beta-1b Trial Grouping. Mitoxantrone prior to interferon beta-1b in ambitious relapsing multiple sclerosis: a three-yr randomised trial. J Neurol Neurosurg Psychiatry. 2011;82:1344–1350. [PubMed] [Google Scholar]
  • Arnold D, Traboulsee A, Coles A, et al. Durable consequence of alemtuzumab on MRI activity in treatment-naive active relapsing-remitting multiple sclerosis patients: four-year follow-upward of Intendance-MS I (P7.246) Neurology. 2015;84 ; 14 Supplement P7.246. [Google Scholar]
  • 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 ii-year prospective randomised multicentre study (INCOMIN) Lancet. 2002;359:1453–1460. [PubMed] [Google Scholar]
  • Panitch H, Goodin DS, Francis G, et al. Bear witness of Interferon Dose−response: European Due north American Compartative Efficacy; University of British Columbia MS/MRI Research Group. Randomized, comparative report of interferon β-1a handling regimens in MS: the EVIDENCE Trial. Neurology. 2002;59:1496–1506. [PubMed] [Google Scholar]
  • Mikol DD, Barkhof F, Chang P, et al. Comparison of subcutaneous interferon beta-1a with glatiramer acetate in patients with relapsing multiple sclerosis (the REbif vs Glatiramer Acetate in Relapsing MS Disease [REGARD] study): a multicentre, randomised, parallel, open-label trial. Lancet Neurol. 2008;7:903–914. [PubMed] [Google Scholar]
  • O'Connor P, Filippi 1000, Arnason B, et al. 250 μg or 500 μg interferon beta-1b versus 20 mg glatiramer acetate in relapsing−remitting multiple sclerosis: a prospective, randomised, multicentre report. Lancet Neurol. 2009;8:889–897. [PubMed] [Google Scholar]
  • Amato MP, Portaccio E. Fertility, pregnancy and childbirth in patients with multiple sclerosis: touch on of disease-modifying drugs. CNS Drugs. 2015;29:207–220. [PubMed] [Google Scholar]
  • Hauser SL, Waubant E, Arnold DL, et al. B-cell depletion with rituximab in relapsing−remitting multiple sclerosis. N Engl J Med. 2008;358:676–688. [PubMed] [Google Scholar]
  • Sorensen PS, Lisby Due south, Grove R, et al. Safety and efficacy of ofatumumab in relapsing−remitting multiple sclerosis: a phase ii study. Neurology. 2014;82:573–581. [PubMed] [Google Scholar]
  • Burman J, Iacobaeus East, Svenningsson A, et al. Autologous haematopoietic stalk jail cell transplantation for aggressive multiple sclerosis: the Swedish experience. J Neurol Neurosurg Psychiatry. 2014;85:1116–1121. [PubMed] [Google Scholar]
  • Atkins HL, Freedman MS. Hematopoietic stalk cell therapy for multiple sclerosis: top 10 lessons learned. Neurotherapeutics. 2013;10:68–76. [PMC gratis article] [PubMed] [Google Scholar]

finlaydrined.blogspot.com

Source: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4670697/

Post a Comment for "Disease-modifying Treatments for Multiple Sclerosis - a Review of Approved Medications"