Skip to main content
Biologics : Targets & Therapy logoLink to Biologics : Targets & Therapy
. 2013 Nov 27;7:247–258. doi: 10.2147/BTT.S53007

Optimizing therapeutics in the management of patients with multiple sclerosis: a review of drug efficacy, dosing, and mechanisms of action

Kavitha Damal 1, Emily Stoker 1, John F Foley 1,
PMCID: PMC3854923  PMID: 24324326

Abstract

Multiple sclerosis (MS) is a debilitating neurological disorder that affects nearly 2 million adults, mostly in the prime of their youth. An environmental trigger, such as a viral infection, is hypothesized to initiate the abnormal behavior of host immune cells: to attack and damage the myelin sheath surrounding the neurons of the central nervous system. While several other pathways and disease triggers are still being investigated, it is nonetheless clear that MS is a heterogeneous disease with multifactorial etiologies that works independently or synergistically to initiate the aberrant immune responses to myelin. Although there are still no definitive markers to diagnose the disease or to cure the disease per se, research on management of MS has improved many fold over the past decade. New disease-modifying therapeutics are poised to decrease immune inflammatory responses and consequently decelerate the progression of MS disease activity, reduce the exacerbations of MS symptoms, and stabilize the physical and mental status of individuals. In this review, we describe the mechanism of action, optimal dosing, drug administration, safety, and efficacy of the disease-modifying therapeutics that are currently approved for MS therapy. We also briefly touch upon the new drugs currently under investigation, and discuss the future of MS therapeutics.

Keywords: multiple sclerosis, immunomodulation, interferons, glatiramer acetate, monoclonal antibodies, dimethyl fumarate

Introduction

Multiple sclerosis (MS) is a complex and chronic demyelinating autoimmune neurological disorder that manifests through an interaction of environmental and genetic factors.15 The onset of MS occurs at an individual’s most productive years (20–40 years),6,7 and affects considerably more women than men.810 A long-term follow-up study of MS reports a steady rise in the incidence of MS, while the age at onset of disease symptoms has been continuously decreasing.6 Nearly 2.5 million individuals worldwide (nearly one in every 400 individuals) are afflicted with MS, although experts consider this number to be an underestimation of true prevalence. MS is unquestionably a disabling disease that impairs both the physical function and cognitive ability of patients.11 While their longevity is not severely compromised (reduction in life span by 6–7 years),12 quality of life is significantly impacted, as individuals are plagued by MS-associated comorbidities, such as chronic pain, fatigue, depression, sleep disorders, spasticity, gait and coordination imbalances, migraines, sensory organ dysfunctions, and overall cognitive impairment. Since the description of MS by French neurologist Jean-Martin Charcot as a triad of symptoms (nystagmus, intention tremor, and slurred speech)13 in 1868, research on the etiology, pathophysiology, and management of this disease has progressed dramatically, although a conclusive diagnostic marker or curative therapy still remains undefined.

MS diagnosis and subtype classification

Presently, a combination of paraclinical diagnostic investigations, including magnetic resonance imaging (MRI) assessment of brain-lesion dissemination in space and time, presence of oligoclonal bands in cerebrospinal fluid, delayed latencies in visual evoked potentials, and changes in retinal nerve fiber-layer thickness evaluated using optical coherence tomography alongside clinical symptoms, as recommended by the 2010 McDonald criteria, are used to guide MS diagnosis.1416 While several potential biomarkers are being studied to ascertain their utility in diagnosing MS,17,18 none has yet been determined as clinically useful. Furthermore, discovery of markers to establish prognosis based on disease symptoms and treatment trajectories is also wanting.19

A majority of MS patients (∼85%) experience symptomatic attacks between dormant states (“remission”), commonly referred to as the relapsing–remitting type (RRMS), which may initially present as a clinically isolated syndrome. This may segue, after a number of years, into secondary progressive MS, marked by fewer or no relapses and gradual neurological worsening with brain atrophy. Primary progressive MS presents with a continuous neurological worsening from the first onset of symptoms. Despite their similarity, studies have identified distinct pathological differences20 that could be translated to determine treatment decisions and predict the prognosis for patients based on the subtype presentation.21,22 Of note, primary and secondary progressive forms have generally been more resistant to anti-inflammatory therapies when compared to RRMS subtype.

Management of MS

MS therapeutics divides into primary disease treatment using immunomodulating agents, which will be discussed in detail, as well as specific symptom management (eg, spasticity, fatigue, depression, pain, etc), which will not be further addressed in this review.

Primary immunomodulatory therapeutics

The goal of mainstay therapies of MS is to reduce relapses and postpone progression of disability in patients.23,24 To this end, strategies adopted to treat MS are twofold: a short-term treatment to help reduce the accumulation of disease burden after an acute relapse, and a long-term, sustained treatment aimed at stabilizing the disease process.25

Short-term treatment for acute relapse

In the initial stages of an MS relapse, individuals are generally treated with high doses (500–1,000 mg) of intravenous corticosteroids (eg, methylprednisolone) for a short period of 3–5 days. In rare cases, subcutaneous or intramuscular injections of adrenocorticotropic hormone (eg, HP Acthar® gel) are used, specifically for individuals who cannot tolerate or have poor response to intravenous prednisolone.2628 These anti-inflammatory agents accelerate the process of recovery, and reduce duration of the relapse; however, they do not have any bearing on the occurrence of new relapses or on long-term disease progression.2730

Long-term disease management

The fundamental pathogenesis of MS is characterized by two stages of disease development.31 The inception of MS symptoms (clinical and paraclinical) and focal demyelination of neurons occur during the early inflammatory phase. The late neurodegenerative phase is characterized by further demyelination of neurons perpetrated by infiltrating macrophages, microglial cells, and lymphocytes that attack the endogenous myelin sheath proteins as antigens, leading to irreversible axonal loss.32 Given the role played by lymphocytes in advancing MS, long-term disease management is largely directed towards suppressing the immune-inflammatory responses that promote demyelination and neuronal degradation in an effort to prevent any saltatory changes in the status quo of patients.23,24,27,33 Outcomes of MS treatments are evaluated based on a reduction in MS annualized relapse rate (ARR), stabilization, or regression in Expanded Disability Status Scale (EDSS) score, and unchanged brain and spinal cord MRI lesion burden.34

Over the past decade, the disease-fighting armamentarium for MS has rapidly expanded with the discovery of new disease-modifying therapeutics (DMTs), which employ different mechanisms to slow or reverse inflammatory lesion formation. To date, regulatory agencies, such as the US Food and Drug Administration (FDA) and the European Medicines Agency, have approved nine different DMTs (Table 1) to aid with modifying the disease course in MS patients (http://www.accessdata.fda.gov/scripts/cder/drugsatfda/index.cfm). Emerging evidence suggests some DMTs may be able to stabilize and perhaps even improve neurological status; however, they are not capable of completely relieving all symptoms of MS.23 Here, we briefly discuss the mechanism of action, optimal dosing, and efficacy of each of these DMTs.

Table 1.

Dosing and side-effects of currently approved multiple sclerosis disease-modifying therapeutics

DMT Year of approval Route of administration Frequency of dosing Dosage Common adverse effects Severe adverse effects
Interferon beta-1b (Betaseron®) 1993 Subcutaneous injection Every other day 250 mcg Influenza-like symptoms, urticaria, depression, injection-site reactions, leukopenia, headache Hepatic injury, congestive heart failure, seizures
Interferon beta-1a (Avonex®) 1996 Intramuscular injection Once a week 30 mcg Influenza-like symptoms, depression, anemia, urticaria, fever, myalgia, asthenia Hepatic injury, congestive heart failure, anaphylactic shock
Glatiramer acetate (Copaxone®) 1996 Subcutaneous injection Daily 20 mg Injection-site reactions, palpitations, urticaria, dyspnea, chest pain, vasodilation Injection site lipoatrophy and necrosis
Interferon beta-1a (Rebif®) 2002 Subcutaneous injection Three times a week 22 mcg
44 mcg
Influenza-like symptoms, depression, injection-site reactions, urticaria, myalgia, fever, abdominal pain, elevated liver enzymes Hepatic injury, anaphylactic shock
Natalizumab (Tysabri®) 2006 Intravenous infusion Every 4 weeks 300 mg Headache, urinary tract infections, lung infections, abdominal pain, fatigue, joint pain, depression, gastroenteritis, urticaria, arthralgia PML, anaphylactic shock, hepatotoxicity
Fingolimod (Gilenya®) 2010 Oral capsule Daily 0.5 mg Headache, influenza, gastrointestinal discomfort, back pain, abnormal liver-function tests, angina Macular edema, bradyarrhythmia, PML, hypotension, herpes infection
Teriflunomide (Aubagio®) 2012 Oral capsule Daily Dyspnea, hypertension, gastrointestinal discomfort, leukopenia, urticaria, alopecia, paresthesia Hepatotoxicity, peripheral neuropathy hyperkalemia, acute renal failure
Dimethyl fumarate (Tecfidera®) 2013 Oral capsule Twice daily 120 mg
240 mg
Gastrointestinal disorders (abdominal pain, diarrhea, etc), flushing, pruritus, rash, erythema Lymphopenia

Note: Mitoxantrone® not covered in this review.

Abbreviations: DMT, disease-modifying therapy; PML, progressive multifocal leukoencephalopathy.

Interferons

Interferons (IFNs) are proteins that belong to the cytokine network and are involved with the regulation of immune response against microbial and viral antigens.35 Their immunomodulatory properties were leveraged to develop the first DMTs for MS, namely, IFN-β1b (Betaseron®, Bayer HealthCare, Leverkusen, Germany; Extavia, Novartis, Basel, Switzerland), a fermented and purified recombinant protein produced in the bacterium Escherichia coli,36 and two preparations of IFN-β1a (Avonex®, Biogen Idec Inc., Weston, MA, USA; Rebif®, Merck Serono, Geneva, Switzerland), which are recombinant human IFN proteins produced in mammalian cells in a glycosylated form.27,37

In vitro studies using human isogenic T cell lines and murine experimental autoimmune encephalomyelitis (EAE; a disease very similar to MS) models and in vivo studies on humans (clinical trials) have elucidated the mechanism of action of IFN-β to be as follows:3739 IFN-β suppresses the proliferation of myelin-basic protein-specific T cells, reduces the production of proinflammatory cytokines (eg, IFN-γ), and induces anti-inflammatory cytokines, such as interleukin (IL)-10.37,39,40 This results in a cytokine balance that protects neurons from demyelination by preventing the proliferation of T cells that are required for advancing the autoimmune process and inhibiting T cells from crossing the blood–brain barrier (BBB) and entering the central nervous system (CNS).39,40 Both IFN-β1b and IFN-β1a are equivalent with respect to their ability to reduce MS disease activity (Tables 1 and 2), and they reduce ARR by up to 30%, decrease formation of new or enlarging gadolinium-enhancing MRI lesions by 50%, and significantly lower the progression of EDSS scores.27,4144

Table 2.

Efficacy of approved multiple sclerosis therapeutics: Phase III clinical trial results

Disease-modifying therapy Study Study design Treatment Patients, n Study duration ARR (% decrease compared to placebo) EDSS (% decrease compared to placebo) Gd-MRI (% decrease in T2 weighted lesions)
Interferon-β1b (Betaseron) IFNB study group,42 Paty et al41 R, DB, PC Placebo
IFN-β 0.05 mcg
IFN-β 0.25 mcg
123
125
124
36 months 0.92
0.8(13)
0.66 (28)
46
47
35
 
66
63
Interferon-β1a (Avonex) Jacobs et al129 R, DB, PC Placebo
IM-IFN
143
158
104 weeks 0.9
0.61 (32)
34.9
21.9(37)
 
35
Interferon-β1a (Rebif) PRISMS study group130 R, DB, PC Placebo
SC-IFN 22 mcg
SC-IFN 44 mcg
187
189
184
12 months 2.56
1.82 (29)
1.73 (32)
37
29 (22)
26 (30)
 
67
78
Glatiramer acetate (Copaxone) Johnson et al53 R, DB, PC Placebo
GA
126
125
24 months 0.84
0.59 (29)
28.8
20.8 (28)
 
NA
Natalizumab (Tysabri) Polman et al1 R, DB, PC Placebo
Nat
627
942
116 weeks 0.73
0.23 (68)
29
17 (41)
 
83
Dimethyl fumarate (Tecfidera) Gold et al99 R, DB, PC Placebo
2 × DMF
3 × DMF
408
410
416
96 weeks 0.36
0.17 (53)
0.19 (48)
27
16 (38)
18 (34)
 
85
74
Fox et al100 R, DB, PC, GA Placebo
2 × DMF
3 × DMF
GA
333
338
316
304
96 weeks 0.4
0.22 (44)
0.2 (50.5)
0.29 (28.6)
17
13 (21)
13 (24)
16 (7)
 
57
65
41
Teriflunomide (Aubagio) O’Connor et al131 R, DB, PC Placebo
Ter 7 mg
Ter 14 mg
363
365
368
108 weeks 0.54
0.37 (31.2)
0.37 (31.5)
27.3
21.7 (23.7)
20.2 (29.8)
 
16.7
31.3
Fingolimod (Gilenya) Kappos et al124 R, DB, PC Placebo
Fing 0.5 mg
Fing 1.25 mg
418
425
429
24 months 0.4
0.18 (52)
0.16 (62)
24.1
12.5 (37)
11.5 (40)
 
74
74
Cohen et al125 R, DB, IM-IFN IM-IFN
Fing 0.5 mg
Fing 1.25 mg
431
429
420
12 months 0.33
0.16 (52)
0.2 (40)
7.9
5.9 (25)
6.7 (15)
 
35
42

Abbreviations: ARR, annualized relapse rate; EDSS, Expanded Disability Status Scale; Gd-MRI, gadolinium-enhanced magnetic resonance imaging; PRISMS, Prevention of Relapses and Disability by Interferon beta-1a Subcutaneously in Multiple Sderosis; R, randomized; DB, double-blind; PC, placebo-controlled; NA, not available; DMF, dimethyl fumarate; 2×, twice daily; 3×, thrice daily; IFN-β, interferon-β1b (Betaseron); IM-IFN, intramuscular interferon-β1a (Avonex); SC-IFN, subcutaneous interferon-β1a (Rebif); GA, glatiramer acetate; Nat, natalizumab; Ter, teriflunomide; Fing, fingolimod.

Glatiramer acetate

Demyelination of neurons in MS is mediated by activation of helper T cells in response to a specific myelin-basic protein (MBP), which is one of the autoantigens in MS.45,46 Peptides of MBP bind to class II major histocompatibility complex (MHC II) molecules, which are then recognized by T cells as antigens and consequently destroyed.47,48 In 1995, Copolymer 1 (glatiramer acetate [GA]/Copaxone®; Teva Pharmaceuticals, Petach Tikva, Israel) was introduced as an alternate therapy to IFN-β. GA is a synthetic polymer of four amino acids (L-glutamate, L-lysine, L-alanine, and L-tyrosine) that mimics MBP, and hence competes with MBP antigens to bind with MHC II.27,49 Using human Epstein–Barr virus-transformed B cell lines, Fridkis-Hareli et al50 demonstrated in vitro that GA binds to MHC-II molecules with high efficiency as well as at a fast rate. Thus, when the MHC-II molecules are blocked from binding to MBP, T cell responses are automatically diverted away from the myelin, resulting in neuronal protection.49,50 The proliferation of T cells is thus controlled by GA in a dose-dependent manner.51 MHC-II molecules interact with CD4+ molecules that are present on the surface of helper T cells (Th1 and Th2) that produce proinflammatory cytokine (Th1-type cytokines: INF-γ) and anti-inflammatory cytokine (Th2-type cytokines: IL-10) molecules.52 GA preferentially inhibits production of INF-γ, induces regulatory Th2-like T cell populations that cross-react with MBP, and produce anti-inflammatory cytokines, which in turn protects the myelin through a “bystander-suppression” mechanism.51 GA is generally well tolerated and reduces ARR by 29%; however, it was unable to reduce disability progression significantly when compared to placebo (Tables 1 and 2).5355

Given their safety profile, low toxicity, reasonable efficacy, and relative tolerability, IFN and GA are often prescribed as first-line MS therapies.23,27 While a meta-analysis study in 2004 indicated that GA was not useful in treating MS,55 a more recent study substantiated its utility in treating relapses related to RRMS, but reiterated its limited impact on disability progression.56

Natalizumab

In the early 1990s, Yednock et al57 identified that monocytic cells selectively bound to inflamed blood vessels in the brain; the inflammation was caused by EAE. Using an EAE mouse model, they demonstrated that the selective adhesion of leukocytes to vascular cell adhesion molecule 1 (VCAM-1), a protein expressed on the surface of vascular endothelial cells in the brain and spinal cord,58 is a critical step to gain entry into the CNS across the BBB. Over 95% of this adhesion was significantly inhibited by antibodies against the integrin molecule α4β1 (very late-activation antigen 4 [VLA-4]), a glycoprotein surface molecule found on all leukocytes except neutrophils.57 Administration of the antibodies reduced the progression of inflammatory disease severity in MS patients (MRI lesions),59,60 and prevented the development of paralysis in animal studies.61 Hence, it was hypothesized that monoclonal antibodies against VLA-4 could help with treating autoimmune inflammatory diseases such as MS by blocking the VCAM-1/VLA-4 interaction, and preventing infiltration of leukocytes across the BBB.57,58 In early 2000, natalizumab (Antegren; Elan, Dublin, Ireland, and Biogen Idec Inc.), an α4-integrin humanized monoclonal antibody to VLA-4, was developed.62 Natalizumab specifically targets the T cells63 and inhibits the α4-integrin-mediated firm adhesion of T cells to the inflamed BBB by 70%, but does not interfere with the initial contact of T cells with the BBB, suggesting that the central mechanism of natalizumab action is prevention of T cell entry into the CNS.64

After establishment of the relative safety profile in a Phase I clinical trial,65 Phase II and Phase III double-blind placebo-controlled trials demonstrated its efficacy in reducing ARR, especially in patients with higher disease activity.60,66 The FDA subsequently approved natalizumab (Tysabri; Biogen Idec Inc.) as an MS monotherapy in 2004 (http://www.accessdata.fda.gov/drugsatfda_docs/appletter/2004/125104_0000_ltr.pdf). However, the emergence of three cases of progressive multifocal leukoencephalopathy (PML) led to its withdrawal from the market for a brief period between February 2005 and June 2006.67,68 In 2006, the results of the Natalizumab Safety and Efficacy in Relapsing Remitting Multiple Sclerosis (AFFIRM) trial,1 with a follow-up of over 2 years, elucidated the superiority of natalizumab in controlling progression of MS (Table 2), resulting in reinstatement of natalizumab as an MS therapeutic.1,69 Natalizumab therapy also significantly improves the overall quality of life of RRMS patients (Table 2).7074 The Safety and Efficacy of Natalizumab in Combination with Interferon Beta-1a in Patients with Relapsing Remitting Multiple Sclerosis (SENTINEL) study further reiterated the efficacy of natalizumab as a combination therapy with IFN-β1a (Table 2) than administration of IFN alone (Table 2).75

The principal limitation to natalizumab utilization has been PML, related to the mutation of the John Cunningham (JC) polyoma virus to a neurotrophic form. Postmarketing surveillance of natalizumab reported 377 incidences of PML76 across the world, and three principal factors have been identified to increase the risk of developing PML (therapy ≥24 months, history of immunosuppressant treatment, and JC-antibody positivity) in patients undergoing natalizumab therapy.71,76,77 Further, recent studies show that higher-titer levels of JC virus antibody predispose to development of PML.78 Less well defined is the possible contribution of excessively low CD62L (L-selectin) expression on CD4+ cell populations79 and the possible association between low body weight and increased PML risk, which are presently being studied extensively.80 Although its superior efficacy in modulating disease activity and progression has made natalizumab a reliable therapy for MS, the risk-to-benefit ratio of the drug varies dramatically among patients, and hence demands a more personalized approach to utilization.

Dimethyl fumarate

Dimethyl fumarate (DMF; BG-12, Tecfidera®; Biogen Idec Inc.) is a methyl ester of fumaric acid approved by the FDA as an oral MS therapy on March 27, 2013 (www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm345528.htm). A well-known pathway to MS is oxidative stress brought about by the effector molecule peroxynitrite (reactive nitrogen species [RNS]).81 Macrophages, upon localization in the glial cells, release proinflammatory cytokines and free radicals to aid with host immune protection.8284 The antimicrobial properties of Nitric Oxide (NO) are well established,85 but as a cytotoxic agent NO also leads to extensive host cellular damage. In inflamed regions, NO is released equivalently to the extent of inflammation: the more the inflammation, the more the NO released.86,87 NO reacts with other free radicals like superoxide to produce RNS,82,88 which in turn induces oxidative damage to the mitochondrial deoxyribonucleic acid, ultimately resulting in decreased adenosine triphosphate production.89 Thus axonal transport, a process that requires adenosine triphosphate, and cellular respiration are impaired, leading to axonal degeneration and irreversible cell apoptosis.89

Initial in vitro studies highlighted the detoxification and anti-inflammatory capabilities of DMF, which reduces with the production and release of inflammatory molecules, such as cytokines and NO, and elevates the production of detoxification enzymes such as reduced-form nicotinamide adenine dinucleotide phosphate quinone reductase 1 and/or glutathione.83,88,90 Additional studies in EAE mouse models showed a DMF dose-dependent decrease in inflammatory cell infiltrates (composed of macrophages, microglial cells, and proinflammatory cytokines).91 DMF also inhibits the expression of VCAM-192 and activates nuclear factor erythroid 2-related factor (Nrf2), a transcription factor with antioxidant properties. Nrf2-mediated antioxidative stress response reduces the free radicals, prevents synthesis of RNS, and thus protects the CNS from degeneration and axonal loss.93 Thus, DMF preserves myelin integrity via two pathways: by down-regulating oxidative stress and corresponding cellular injury, as well as by inhibiting proinflammatory cytokines.82,9395

The first exploratory study of oral fumaric acid esters was performed in ten patients with RRMS in 2006.96 Promising results from this study led to the expansion of clinical research to apply BG-12, a second-generation fumarate derivative as a potential oral therapeutic for RRMS.97 Kappos et al98 demonstrated the safety and efficacy of DMF, showing a 69% reduction in gadolinium-enhanced MRI (Gd-MRI) lesions and a 32% reduction in ARR when compared to placebo (Table 1). The Phase III trials DEFINE (Determination of the Efficacy and Safety of Oral Fumarate in Relapsing-Remitting MS)99 and CONFIRM (Comparator and an Oral Fumarate in Relapsing-Remitting Multiple Sclerosis)100 further elucidated the efficacy of DMF in reducing ARR by 53% and 44% compared to placebo or GA, respectively (Table 2), a decrease in Gd-MRI activity by 70%, and a decrease in disability progression of 38% (Table 2). In addition, the side effects of DMF were relatively benign, including gastrointestinal discomfort, flushing, decreased lymphocyte count, and elevated liver aminotransferase levels (Table 1).99101 Although some formulations and metabolites of fumaric acid esters are known to cause PML,102,103 DMF by itself has been suggested as a safe drug with relatively low side effects.104

Teriflunomide

Teriflunomide is the active metabolite of leflunomide, a chemical with known anti-inflammatory, anti-proliferative and immunosuppressive properties.105 The utility of oral teriflunomide in treating MS was realized in 2006 through a Phase II clinical study reported by O’Connor et al,106 which elucidated its immunomodulatory effects in decreasing MRI lesions and ARR in RRMS patients. Triggering of an immune response involves the proliferation of T cells and B cells to provide antigen-specific cell-mediated or humoral immunity, respectively. In order to activate the lymphocytes to undergo clonal expansion, adhesion of T cells to the antigen-presenting cells is a crucial step.107 Teriflunomide primarily acts by interfering with the lymphocyte cell cycle and inhibiting proliferation. Lymphocyte mitosis requires an eightfold increase in the level of pyrimidine ribonucleotides (eg, ribonucleotide uridine monophosphate) during the interphase of the cell cycle. A key enzyme, dihydroorotate dehydrogenase (DHODH), is necessary for the de novo synthesis of these pyrimidine ribonucleotides, which in turn fulfills the metabolic needs that are necessary for clonal expansion of lymphocytes.108,109 By preventing the synthesis of DHODH, teriflunomide actively reduces the pyrimidine ribonucleotide levels, stalls mitosis and further lymphocyte proliferation, and thus protects neurons from autoimmune damage. Teriflunomide also acts by inhibiting protein tyrosine kinases, leading to decreased T cell proliferation, and by shifting the cytokine profile to prevent inflammation (ie, inhibiting synthesis of proinflammatory cytokines and promoting anti-inflammatory cytokines).110 EAE animal models treated with teriflunomide showed a significant reduction in axonal damage by up to 96%, nonlatency or delay of motor-evoked potentials, and preservation of the anatomical integrity in both the ascending and descending tracts of the spinal cord, thus underscoring its direct effect on neuroprotection.111,112

The TEMSO (Teriflunomide Multiple Sclerosis Oral) trial68 illustrated the efficacy of Aubagio® (Genzyme, Boston, MA, USA) as an MS DMT (Table 2), demonstrating nearly 31% reduction in ARR, a longer time to first relapse, approximately 20% decrease in disability progression, and a decrease in Gd-MRI lesion activity when compared to placebo. Recent animal studies have shown that teriflunomide can significantly improve motor function and decrease the probability of debilitating paralysis, suggesting that it might become one of the early treatment drugs for MS.111 To this end, a Phase III clinical trial (TOPIC [Phase III Study with Teriflunomide Versus Placebo in Patients with First Clinical Symptom of Multiple Sclerosis]; ClinicalTrials.gov NCT00622700)113 is ongoing, with an expected study completion date of August 2015, that will inform the utility of teriflunomide in early clinical treatment.

Fingolimod

Levels of T cells and B cells are regulated through a circulatory mechanism between the blood and appropriate secondary lymphoid organs (SLOs), and the homing of T cells from the blood to sites of inflammation in the CNS is crucial for MS pathogenesis. The MBP-activated T cells breach the BBB, reach the site of inflammation, become encephalitogenic effector cells, and initiate demyelination within the CNS.114 An extracellular signaling molecule, sphingosine-1-phosphate (S1P), regulates the process of trafficking T cells and B cells from the lymph to the blood.115 The S1P receptors, when activated, induce egress of T cells (naïve) from peripheral blood and sequester them within the SLOs, thus decreasing T cell levels in the blood.106,116118

FTY720 (2-amino-[2-{4-octylphenyl}ethyl]-1, 3-propanediol hydrochloride), a synthetic S1P analogue, immunomodulates the S1P receptors and revises the T cells’ migratory pathway (ie, prevents emigration of activated T cells from lymph nodes and sequesters them within SLOs).118 This sequestration dramatically reduces the availability of T cells in the blood that can infiltrate the BBB and home in to the inflamed cells in the CNS. Thus, FTY720 (Fingolimod, Gilenya®; Novartis) effectively confers neuroprotection against demyelination.119121 In murine models, FTY720 dramatically reduced the expression of the proinflammatory Th1-type cytokines due to the absence of T cell migrants to promote further inflammation.122 In addition, FTY720 is also suggested to promote remyelination of neurons in the CNS via direct interaction with oligodendrocytes.123

The FREEDOMS (FTY720 Research Evaluating Effects of Daily Oral Therapy in Multiple Sclerosis) study124 tested two doses (0.5 mg or 1.25 mg) of fingolimod or placebo taken once daily for 24 months. Results showed that patients treated with fingolimod had an approximately 70% decrease in MS disease activity and a stable EDSS score (Table 2), and about 50% of patients had no change in T2-weighted MRI lesions.124 In a head-to-head comparison with IFN-β1a (TRANSFORMS [Trial Assessing Injectable Interferon Versus FTY720 Oral in Relapsing-Remitting Multiple Sclerosis]), RRMS patients on fingolimod on average had lower ARRs, significantly fewer new or enlarged T2-weighted MRI lesions, and a stable EDSS score (Table 2).125 Despite its promising efficacy, fingolimod’s safety profile has been challenging, with occurrence of bradyarrhythmias that have caused deaths and elicited prolonged cardiac monitoring for first dosings.126 Death of MS patients who were administered fingolimod in the presence of varicella zoster viral infection has also been reported.127 A first case of PML in the absence of prior natalizumab therapy has also been identified.128 Research reports on the safety and efficacy of fingolimod are still emerging, and additional data will further inform the risks versus benefits of this drug for MS therapy.

MS therapy: past, present, and future

Great strides have been made in the last 20 years in MS therapeutics, beginning with the initial INF-β-positive trials up through the recent approval of DMF. A number of newer agents are poised potentially to gain approval over the next few years, which we have briefly touched upon (Table 3). Incremental improvements in efficacy have been seen together with improved odds of disease stability with therapy and the potential for disease improvement with some agents.

Table 3.

Disease-modifying therapies (DMTs) currently under review for use as multiple sclerosis therapies

DMT (target) Study phase Study design Treatment Patients, n Study duration ARR reduction (% decrease) % disability progression (% decrease from placebo) % reduction in T2-weighted Gd-MRI from placebo
Alemtuzumab (CD52 – lymphocyte depletion) Phase III132 Randomized, double-blind, comparator arm SC-IFN 44 mg
Ale 12 mg
187
376
24 months 0.39
0.18 (54)
II
8 (30)

NS
Laquinimod (Th1 and Th2 – inhibits leukocyte migration into CNS) Phase III133 Randomized, double-blind, placebo-controlled Placebo
Laq 0.6 mg
556
550
24 months 0.39
0.30 (23)
16
11 (29)

30
Daclizumab (IL-2 signal modulator–inhibits innate lymphoid cells) Phase III134 Randomized, double-blind, placebo-controlled Placebo
Dac 150 mg
Dac 300 mg
204
208
209
52 weeks 0.46
0.21 (54)
0.23 (50)
13
6 (57)
8 (43)

70
79
Ocrelizumab (CD20–B cell depletion) Phase II135 Randomized, double-blind, placebo- and IFN-β1a-controlled Placebo
IFN-β1a
Ocr 600 mg
Ocr 2,000 mg
54
55
56
55
24–48 weeks 0.16
0.14
0.09
0.28
NA NS

Abbreviations: DMT, disease-modifying therapy; ARR, annualized relapse rate; NA, not available; NS, not significant; CNS, central nervous system; IFN-β1a, interferon-β1a; SC-IFN, subcutaneous interferon-β1a; IL, interkeukin; Ale, alemtuzumab; Laq, laquinimod; Dac, daclizumab; Ocr, ocrelizumab; CD, cluster of differentiation; Gd-MRI, gadolinium-enhanced magnetic resonance imaging; Th, helper T cell.

As discussed earlier, most agents modify the disease course primarily through anti-inflammatory pathways, although the newest entrant (DMF) may well be efficacious in utilizing a novel antioxidant pathway. However, all these agents carry adverse side effects of varying degrees, and hence a thorough evaluation of the risk–benefit ratio for the individual patient is imperative prior to drug administration (Figure 1). In MS, T cells are known to attack three different antigens: MBP, myelin oligodendrocyte glycoprotein, and proteolipid protein. The heterogeneity of MS disease is vast, and each patient’s antibody signature varies: the antigen epitopes that elicit antibody response differ among people, and so does the mechanism of “epitope spreading,” wherein autoreactive T cell activation is elicited by new epitopes secondary to the dominant epitope, either due to their physical proximity or molecular similarity to the dominant epitope, resulting in sequential self-damage. Currently, ways to personalize MS treatment by recognizing these individual epitopes and formulating corresponding antibodies are being explored. Remyelinating agents are actively under investigation, and may yield novel strategies to increase neuronal functionality and survival in the coming years. With preclinical animal trials showing superior efficacy in alleviating MS symptoms, ongoing human clinical trials are investigating the use of hematopoietic and mesenchymal stem cells for effective management of MS. While the goal of hematopoietic stem cell transplantation is refurbishing the aberrant T cell population with nonautoreactive T cells, mesenchymal stem cell transplantation can potentially promote neural restoration. MS therapeutics is now an area of rapid evolution, with broadening biological targets and ongoing improvement in efficacy.

Figure 1.

Figure 1

Optimizing the risk/efficacy balance of approved MS therapeutics: a physician perspective

Abbreviations: DMF, dimethyl fumarate; GA, glatiramer acetate; MS, Multiple Sclerosis; EDSS, Expanded Disability Status Scale; MR, magnetic resonance; CNS, central nervous system.

Footnotes

Disclosure

Dr Foley has received personal compensation for consulting, speaking and scientific advisory boards from Genzyme, Biogen Idec Inc. and Teva. Dr Foley has also received research support from Biogen Idec Inc., Genzyme, Teva and Avanir. The other authors report no conflicts of interest in this work.

References

  • 1.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. doi: 10.1056/NEJMoa044397. [DOI] [PubMed] [Google Scholar]
  • 2.Williams R, Rigby AS, Airey M, Robinson M, Ford H. Multiple sclerosis: its epidemiological, genetic, and health care impact. J Epidemiol Community Health. 1995;49(6):563–569. doi: 10.1136/jech.49.6.563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Compston A, Coles A. Multiple sclerosis. Lancet. 2002;359(9313):1221–1231. doi: 10.1016/S0140-6736(02)08220-X. [DOI] [PubMed] [Google Scholar]
  • 4.Compston A, Coles A. Multiple sclerosis. Lancet. 2008;372(9648):1502–1517. doi: 10.1016/S0140-6736(08)61620-7. [DOI] [PubMed] [Google Scholar]
  • 5.Keegan BM, Noseworthy JH. Multiple sclerosis. Ann Rev Medic. 2002;53:285–302. doi: 10.1146/annurev.med.53.082901.103909. [DOI] [PubMed] [Google Scholar]
  • 6.Koch-Henriksen N. The Danish Multiple Sclerosis Registry: a 50-year follow-up. Mult Scler. 1999;5(4):293–296. doi: 10.1177/135245859900500418. [DOI] [PubMed] [Google Scholar]
  • 7.Mayr WT, Pittock SJ, McClelland RL, Jorgensen NW, Noseworthy JH, Rodriguez M. Incidence and prevalence of multiple sclerosis in Olmsted County, Minnesota, 1985–2000. Neurology. 2003;61(10):1373–1377. doi: 10.1212/01.wnl.0000094316.90240.eb. [DOI] [PubMed] [Google Scholar]
  • 8.Hawkins SA, McDonnell GV. Benign multiple sclerosis? Clinical course, long term follow up, and assessment of prognostic factors. J Neurol Neurosurg Psychiatry. 1999;67(2):148–152. doi: 10.1136/jnnp.67.2.148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Whitacre CC, Reingold SC, O’Looney PA, et al. A gender gap in autoimmunity: Task Force on Gender, Multiple Sclerosis and Autoimmunity. Science. 1999;283(5406):1277–1278. doi: 10.1126/science.283.5406.1277. [DOI] [PubMed] [Google Scholar]
  • 10.Orton SM, Herrera BM, Yee IM, et al. Sex ratio of multiple sclerosis in Canada: a longitudinal study. Lancet Neurol. 2006;5(11):932–936. doi: 10.1016/S1474-4422(06)70581-6. [DOI] [PubMed] [Google Scholar]
  • 11.Kister I, Chamot E, Salter AR, Cutter GR, Bacon TE, Herbert J. Disability in multiple sclerosis: a reference for patients and clinicians. Neurology. 2013;80(11):1018–1024. doi: 10.1212/WNL.0b013e3182872855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Sadovnick AD, Ebers GC, Wilson RW, Paty DW. Life expectancy in patients attending multiple sclerosis clinics. Neurology. 1992;42(5):991–994. doi: 10.1212/wnl.42.5.991. [DOI] [PubMed] [Google Scholar]
  • 13.Charcot JM. Histologie de la sclerose en plaques. Gaz Hop Civ Mil Empire Ottoman. 1868;41:554–555. [Google Scholar]
  • 14.Polman CH, Reingold SC, Banwell B, et al. Diagnostic criteria for multiple sclerosis: 2010 revisions to the McDonald criteria. Ann Neurol. 2011;69(2):292–302. doi: 10.1002/ana.22366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Stangel M, Fredrikson S, Meinl E, Petzold A, Stuve O, Tumani H. The utility of cerebrospinal fluid analysis in patients with multiple sclerosis. Nat Rev Neurol. 2013;9(5):267–276. doi: 10.1038/nrneurol.2013.41. [DOI] [PubMed] [Google Scholar]
  • 16.Kuenz B, Deisenhammer F, Berger T, Reindl M. Diagnostic biomarkers in multiple sclerosis. Expert Opinion Medical Diagn. 2007;1(2):225–233. doi: 10.1517/17530059.1.2.225. [DOI] [PubMed] [Google Scholar]
  • 17.Katsavos S, Anagnostouli M. Biomarkers in multiple sclerosis: an up-to-date overview. Mult Scler Int. 2013;2013:340508. doi: 10.1155/2013/340508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Leone MA, Barizzone N, Esposito F, et al. Association of genetic markers with CSF oligoclonal bands in multiple sclerosis patients. PloS One. 2013;8(6):e64408. doi: 10.1371/journal.pone.0064408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Rudick RA. The elusive biomarker for personalized medicine in multiple sclerosis: the search continues. Neurology. 2012;79(6):498–499. doi: 10.1212/WNL.0b013e318259e13a. [DOI] [PubMed] [Google Scholar]
  • 20.Oberwahrenbrock T, Schippling S, Ringelstein M, et al. Retinal damage in multiple sclerosis disease subtypes measured by high-resolution optical coherence tomography. Mult Scler Int. 2012;2012:530305. doi: 10.1155/2012/530305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Vukusic S, Confavreux C. Primary and secondary progressive multiple sclerosis. J Neurol Sci. 2003;206(2):153–155. doi: 10.1016/s0022-510x(02)00427-6. [DOI] [PubMed] [Google Scholar]
  • 22.Hagman S, Raunio M, Rossi M, Dastidar P, Elovaara I. Disease-associated inflammatory biomarker profiles in blood in different subtypes of multiple sclerosis: prospective clinical and MRI follow-up study. J Neuroimmunol. 2011;234(1–2):141–147. doi: 10.1016/j.jneuroim.2011.02.009. [DOI] [PubMed] [Google Scholar]
  • 23.Freedman MS, Selchen D, Arnold DL, et al. Treatment optimization in MS: Canadian MS Working Group updated recommendations. Can J Neurol Sci. 2013;40(3):307–323. doi: 10.1017/s0317167100014244. [DOI] [PubMed] [Google Scholar]
  • 24.Kieseier BC, Wiendl H, Hemmer B, Hartung HP. Treatment and treatment trials in multiple sclerosis. Curr Opin Neurol. 2007;20(3):286–293. doi: 10.1097/WCO.0b013e3281568b80. [DOI] [PubMed] [Google Scholar]
  • 25.Spain RI, Cameron MH, Bourdette D. Recent developments in multiple sclerosis therapeutics. BMC Med. 2009;7:74. doi: 10.1186/1741-7015-7-74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Filippini G, Brusaferri F, Sibley WA, et al. Corticosteroids or ACTH for acute exacerbations in multiple sclerosis. Cochrane Database Syst Rev. 2000;(4):CD001331. doi: 10.1002/14651858.CD001331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Rudick RA, Cohen JA, Weinstock-Guttman B, Kinkel RP, Ransohoff RM. Management of multiple sclerosis. N Engl J Med. 1997;337(22):1604–1611. doi: 10.1056/NEJM199711273372207. [DOI] [PubMed] [Google Scholar]
  • 28.Berkovich R. Treatment of acute relapses in multiple sclerosis. Neurotherapeutics. 2013;10(1):97–105. doi: 10.1007/s13311-012-0160-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Burton JM, O’Connor PW, Hohol M, Beyene J. Oral versus intravenous steroids for treatment of relapses in multiple sclerosis. Cochrane Database Syst Rev. 2012;12:CD006921. doi: 10.1002/14651858.CD006921.pub3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Myhr KM, Mellgren SI. Corticosteroids in the treatment of multiple sclerosis. Acta Neurologica Scand Suppl. 2009;(189):73–80. doi: 10.1111/j.1600-0404.2009.01213.x. [DOI] [PubMed] [Google Scholar]
  • 31.Lassmann H. Pathology and disease mechanisms in different stages of multiple sclerosis. J Neurol Sci. 2013 Jun 2; doi: 10.1016/j.jns.2013.05.010. Epub. [DOI] [PubMed] [Google Scholar]
  • 32.Bø L, Esiri M, Evangelou N, Kuhlmann T. Demyelination and remyelination in multiple sclerosis. In: Duncan ID, Franklin RJM, editors. Myelin Repair and Neuroprotection in Multiple Sclerosis. New York: Springer; 2013. pp. 23–45. [Google Scholar]
  • 33.Crayton H, Heyman RA, Rossman HS. A multimodal approach to managing the symptoms of multiple sclerosis. Neurology. 2004;63(11 Suppl 5):S12–S18. doi: 10.1212/wnl.63.11_suppl_5.s12. [DOI] [PubMed] [Google Scholar]
  • 34.Kurtzke JF. Rating neurologic impairment in multiple sclerosis: an expanded disability status scale (EDSS) Neurology. 1983;33(11):1444–1452. doi: 10.1212/wnl.33.11.1444. [DOI] [PubMed] [Google Scholar]
  • 35.Weinstock-Guttman B, Ransohoff RM, Kinkel RP, Rudick RA. The interferons: biological effects, mechanisms of action, and use in multiple sclerosis. Ann Neurol. 1995;37(1):7–15. doi: 10.1002/ana.410370105. [DOI] [PubMed] [Google Scholar]
  • 36.Lin L. Betaseron. Dev Biol Stand. 1998;96:97–104. [PubMed] [Google Scholar]
  • 37.Weber F, Janovskaja J, Polak T, Poser S, Rieckmann P. Effect of interferon beta on human myelin basic protein-specific T-cell lines: comparison of IFNbeta-1a and IFNbeta-1b. Neurology. 1999;52(5):1069–1071. doi: 10.1212/wnl.52.5.1069. [DOI] [PubMed] [Google Scholar]
  • 38.Rudick RA, Ransohoff RM, Lee JC, et al. In vivo effects of interferon beta-1a on immunosuppressive cytokines in multiple sclerosis. Neurology. 1998;50(5):1294–1300. doi: 10.1212/wnl.50.5.1294. [DOI] [PubMed] [Google Scholar]
  • 39.Yong VW, Chabot S, Stuve O, Williams G. Interferon beta in the treatment of multiple sclerosis: mechanisms of action. Neurology. 1998;51(3):682–689. doi: 10.1212/wnl.51.3.682. [DOI] [PubMed] [Google Scholar]
  • 40.Kieseier BC. The mechanism of action of interferon-beta in relapsing multiple sclerosis. CNS Drugs. 2011;25(6):491–502. doi: 10.2165/11591110-000000000-00000. [DOI] [PubMed] [Google Scholar]
  • 41.Paty DW, Li DK. Interferon beta-1b is effective in relapsing-remitting multiple sclerosis. II. MRI analysis results of a multicenter, randomized, double-blind, placebo-controlled trial. UBC MS/MRI Study Group and the IFNB Multiple Sclerosis Study Group. Neurology. 1993;43(4):662–667. doi: 10.1212/wnl.43.4.662. [DOI] [PubMed] [Google Scholar]
  • 42.Interferon beta-1b is effective in relapsing-remitting multiple sclerosis. I. Clinical results of a multicenter, randomized, double-blind, placebo-controlled trial. The IFNB Multiple Sclerosis Study Group. Neurology. 1993;43(4):655–661. doi: 10.1212/wnl.43.4.655. [No authors listed] [DOI] [PubMed] [Google Scholar]
  • 43.Jacobs LD, Cookfair DL, Rudick RA, et al. Intramuscular interferon beta-1a for disease progression in relapsing multiple sclerosis. Ann Neurol. 1996;39(3):285–294. doi: 10.1002/ana.410390304. [DOI] [PubMed] [Google Scholar]
  • 44.Fernandez O, Antiquedad A, Arbizu T, et al. Treatment of relapsing-remitting multiple sclerosis with natural interferon beta: a multicenter, randomized clinical trial. Mult Scler. 1995;1(Suppl 1):S67–S69. [PubMed] [Google Scholar]
  • 45.Ponomarenko NA, Durova OM, Vorobiev II, et al. Autoantibodies to myelin basic protein catalyze site-specific degradation of their antigen. Proc Natl Acad Sci U S A. 2006;103(2):281–286. doi: 10.1073/pnas.0509849103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Ponomarenko NA, Durova OM, Vorobiev II, et al. Catalytic activity of autoantibodies toward myelin basic protein correlates with the scores on the multiple sclerosis expanded disability status scale. Immunol Lett. 2006;103(1):45–50. doi: 10.1016/j.imlet.2005.10.006. [DOI] [PubMed] [Google Scholar]
  • 47.Valli A, Sette A, Kappos L, et al. Binding of myelin basic protein peptides to human histocompatibility leukocyte antigen class II molecules and their recognition by T cells from multiple sclerosis patients. J Clin Invest. 1993;91(2):616–628. doi: 10.1172/JCI116242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Ota K, Matsui M, Milford EL, Mackin GA, Weiner HL, Hafler DA. T-cell recognition of an immunodominant myelin basic protein epitope in multiple sclerosis. Nature. 1990;346(6280):183–187. doi: 10.1038/346183a0. [DOI] [PubMed] [Google Scholar]
  • 49.Wolinsky JS. Copolymer 1: a most reasonable alternative therapy for early relapsing-remitting multiple sclerosis with mild disability. Neurology. 1995;45(7):1245–1247. doi: 10.1212/wnl.45.7.1245. [DOI] [PubMed] [Google Scholar]
  • 50.Fridkis-Hareli M, Teitelbaum D, Gurevich E, et al. Direct binding of myelin basic protein and synthetic copolymer 1 to class II major histocompatibility complex molecules on living antigen-presenting cells – specificity and promiscuity. Proc Natl Acad Sci U S A. 1994;91(11):4872–4876. doi: 10.1073/pnas.91.11.4872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Gran B, Tranquill LR, Chen M, et al. Mechanisms of immunomodulation by glatiramer acetate. Neurology. 2000;55(11):1704–1714. doi: 10.1212/wnl.55.11.1704. [DOI] [PubMed] [Google Scholar]
  • 52.Berger A. Th1 and Th2 responses: what are they? BMJ. 2000;321(7258):424. doi: 10.1136/bmj.321.7258.424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.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. doi: 10.1212/wnl.45.7.1268. [DOI] [PubMed] [Google Scholar]
  • 54.Korczyn AD, Nisipeanu P. Safety profile of copolymer 1: analysis of cumulative experience in the United States and Israel. J Neurol. 1996;243(4 Suppl 1):S23–S26. doi: 10.1007/BF00873698. [DOI] [PubMed] [Google Scholar]
  • 55.Munari L, Lovati R, Boiko A. Therapy with glatiramer acetate for multiple sclerosis. Cochrane Database Syst Rev. 2004;(1):CD004678. doi: 10.1002/14651858.CD004678. [DOI] [PubMed] [Google Scholar]
  • 56.La Mantia L, Munari LM, Lovati R. Glatiramer acetate for multiple sclerosis. Cochrane Database Syst Rev. 2010;(5):CD004678. doi: 10.1002/14651858.CD004678.pub2. [DOI] [PubMed] [Google Scholar]
  • 57.Yednock TA, Cannon C, Fritz LC, Sanchez-Madrid F, Steinman L, Karin N. Prevention of experimental autoimmune encephalomyelitis by antibodies against alpha 4 beta 1 integrin. Nature. 1992;356(6364):63–66. doi: 10.1038/356063a0. [DOI] [PubMed] [Google Scholar]
  • 58.Leger OJ, Yednock TA, Tanner L, et al. Humanization of a mouse antibody against human alpha-4 integrin: a potential therapeutic for the treatment of multiple sclerosis. Hum Antibodies. 1997;8(1):3–16. [PubMed] [Google Scholar]
  • 59.Kent SJ, Karlik SJ, Rice GP, Horner HC. A monoclonal antibody to alpha 4-integrin reverses the MR-detectable signs of experimental allergic encephalomyelitis in the guinea pig. J Magn Reson Imaging. 1995;5(5):535–540. doi: 10.1002/jmri.1880050510. [DOI] [PubMed] [Google Scholar]
  • 60.Miller DH, Khan OA, Sheremata WA, et al. A controlled trial of natalizumab for relapsing multiple sclerosis. N Engl J Med. 2003;348(1):15–23. doi: 10.1056/NEJMoa020696. [DOI] [PubMed] [Google Scholar]
  • 61.Kent SJ, Karlik SJ, Cannon C, et al. A monoclonal antibody to alpha 4 integrin suppresses and reverses active experimental allergic encephalomyelitis. J Neuroimmunol. 1995;58(1):1–10. doi: 10.1016/0165-5728(94)00165-k. [DOI] [PubMed] [Google Scholar]
  • 62.Elices MJ. Natalizumab. Elan/Biogen. Curr Opin Investig Drugs. 2003;4(11):1354–1362. [PubMed] [Google Scholar]
  • 63.Bauer M, Brakebusch C, Coisne C, et al. Beta1 integrins differentially control extravasation of inflammatory cell subsets into the CNS during autoimmunity. Proc Natl Acad Sci U S A. 2009;106(6):1920–1925. doi: 10.1073/pnas.0808909106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Coisne C, Mao W, Engelhardt B. Cutting edge: natalizumab blocks adhesion but not initial contact of human T cells to the blood-brain barrier in vivo in an animal model of multiple sclerosis. J Immunol. 2009;182(10):5909–5913. doi: 10.4049/jimmunol.0803418. [DOI] [PubMed] [Google Scholar]
  • 65.Sheremata WA, Vollmer TL, Stone LA, Willmer-Hulme AJ, Koller M. A safety and pharmacokinetic study of intravenous natalizumab in patients with MS. Neurology. 1999;52(5):1072–1074. doi: 10.1212/wnl.52.5.1072. [DOI] [PubMed] [Google Scholar]
  • 66.O’Connor P, Miller D, Riester K, et al. Relapse rates and enhancing lesions in a phase II trial of natalizumab in multiple sclerosis. Mult Scler. 2005;11(5):568–572. doi: 10.1191/1352458505ms1205oa. [DOI] [PubMed] [Google Scholar]
  • 67.Sheremata WA, Minagar A, Alexander JS, Vollmer T. The role of alpha-4 integrin in the aetiology of multiple sclerosis: current knowledge and therapeutic implications. CNS Drugs. 2005;19(11):909–922. doi: 10.2165/00023210-200519110-00002. [DOI] [PubMed] [Google Scholar]
  • 68.O’Connor P. Natalizumab and the role of alpha 4-integrin antagonism in the treatment of multiple sclerosis. Expert Opin Biol Ther. 2007;7(1):123–136. doi: 10.1517/14712598.7.1.123. [DOI] [PubMed] [Google Scholar]
  • 69.Miller DH, Soon D, Fernando KT, et al. MRI outcomes in a placebo-controlled trial of natalizumab in relapsing MS. Neurology. 2007;68(17):1390–1401. doi: 10.1212/01.wnl.0000260064.77700.fd. [DOI] [PubMed] [Google Scholar]
  • 70.Rudick RA, Miller D, Hass S, et al. Health-related quality of life in multiple sclerosis: effects of natalizumab. Ann Neurol. 2007;62(4):335–346. doi: 10.1002/ana.21163. [DOI] [PubMed] [Google Scholar]
  • 71.Balcer LJ, Galetta SL, Calabresi PA, et al. Natalizumab reduces visual loss in patients with relapsing multiple sclerosis. Neurology. 2007;68(16):1299–1304. doi: 10.1212/01.wnl.0000259521.14704.a8. [DOI] [PubMed] [Google Scholar]
  • 72.Phillips JT, Giovannoni G, Lublin FD, et al. Sustained improvement in Expanded Disability Status Scale as a new efficacy measure of neurological change in multiple sclerosis: treatment effects with natalizumab in patients with relapsing multiple sclerosis. Mult Scler. 2011;17(8):970–979. doi: 10.1177/1352458511399611. [DOI] [PubMed] [Google Scholar]
  • 73.Weinstock-Guttman B, Galetta SL, Giovannoni G, et al. Additional efficacy endpoints from pivotal natalizumab trials in relapsing-remitting MS. J Neurol. 2012;259(5):898–905. doi: 10.1007/s00415-011-6275-7. [DOI] [PubMed] [Google Scholar]
  • 74.Cadavid D, Jurgensen S, Lee S. Impact of natalizumab on ambulatory improvement in secondary progressive and disabled relapsing-remitting multiple sclerosis. PloS One. 2013;8(1):e53297. doi: 10.1371/journal.pone.0053297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Rudick RA, Stuart WH, Calabresi PA, et al. Natalizumab plus interferon beta-1a for relapsing multiple sclerosis. N Engl J Med. 2006 Mar 2;354(9):911–923. doi: 10.1056/NEJMoa044396. [DOI] [PubMed] [Google Scholar]
  • 76.Biogen Idec Tysabri (natalizumab): benefit/risk update and PML risk stratification. 2013. [Accessed September 17, 2013]. Available from: http://www.slideshare.net/gavingiovannoni/natalizuman.
  • 77.Bloomgren G, Richman S, Hotermans C, et al. Risk of natalizumab-associated progressive multifocal leukoencephalopathy. N Engl J Med. 2012;366(20):1870–1880. doi: 10.1056/NEJMoa1107829. [DOI] [PubMed] [Google Scholar]
  • 78.Plavina T, Subramanyam M, Bloomgren G, et al. Use of JC virus antibody index to stratify risk of progressive multifocal leukoencephalopathy in natalizumab-treated patients with multiple sclerosis; Poster presented at: 27th Annual Meeting of the CMSC and the Fifth Cooperative Meeting of the CMSC-ACTRIMS; May 29–June 1, 2013; Orlando, FL. [Google Scholar]
  • 79.Schwab N, Schneider-Hohendorf T, Posevitz V, et al. L-selectin is a possible biomarker for individual PML risk in natalizumab-treated MS patients. Neurology. 2013;81(10):865–871. doi: 10.1212/WNL.0b013e3182a351fb. [DOI] [PubMed] [Google Scholar]
  • 80.Foley JF. Natalizumab related PML: an evolving risk stratification paradigm; Poster presented at: American Academy of Neurology 2013 Annual Meeting; March 16–23, 2013; San Diego, CA. [Google Scholar]
  • 81.Gonsette RE. Neurodegeneration in multiple sclerosis: the role of oxidative stress and excitotoxicity. J Neurol Sci. 2008;274(1–2):48–53. doi: 10.1016/j.jns.2008.06.029. [DOI] [PubMed] [Google Scholar]
  • 82.Brosnan CF, Cannella B, Battistini L, Raine CS. Cytokine localization in multiple sclerosis lesions: correlation with adhesion molecule expression and reactive nitrogen species. Neurology. 1995;45(6 Suppl 6):S16–S21. doi: 10.1212/wnl.45.6_suppl_6.s16. [DOI] [PubMed] [Google Scholar]
  • 83.Wierinckx A, Brevé J, Mercier D, Schultzberg M, Drukarch B, Van Dam AM. Detoxication enzyme inducers modify cytokine production in rat mixed glial cells. J Neuroimmunol. 2005;166(1–2):132–143. doi: 10.1016/j.jneuroim.2005.05.013. [DOI] [PubMed] [Google Scholar]
  • 84.Miller E, Wachowicz B, Majsterek I. Advances in antioxidative therapy of multiple sclerosis. Current medicinal chemistry. 2013 Jun 25; doi: 10.2174/09298673113209990156. Epub. [DOI] [PubMed] [Google Scholar]
  • 85.Bogdan C. Nitric oxide and the immune response. Nat Immunol. 2001;2(10):907–916. doi: 10.1038/ni1001-907. [DOI] [PubMed] [Google Scholar]
  • 86.Nathan C, Shiloh MU. Reactive oxygen and nitrogen intermediates in the relationship between mammalian hosts and microbial pathogens. Proc Natl Acad Sci U S A. 2000;97(16):8841–8848. doi: 10.1073/pnas.97.16.8841. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Hibbs JB, Jr, Taintor RR, Vavrin Z, Rachlin EM. Nitric oxide: a cytotoxic activated macrophage effector molecule. Biochem Biophys Res Commun. 1988;157(1):87–94. doi: 10.1016/s0006-291x(88)80015-9. [DOI] [PubMed] [Google Scholar]
  • 88.Smith KJ, Kapoor R, Felts PA. Demyelination: the role of reactive oxygen and nitrogen species. Brain Pathol. 1999;9(1):69–92. doi: 10.1111/j.1750-3639.1999.tb00212.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Su KG, Banker G, Bourdette D, Forte M. Axonal degeneration in multiple sclerosis: the mitochondrial hypothesis. Curr Neurol Neurosci Rep. 2009;9(5):411–417. doi: 10.1007/s11910-009-0060-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Albrecht P, Bouchachia I, Goebels N, et al. Effects of dimethyl fumarate on neuroprotection and immunomodulation. J Neuroinflammation. 2012;9:163. doi: 10.1186/1742-2094-9-163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Schilling S, Goelz S, Linker R, Luehder F, Gold R. Fumaric acid esters are effective in chronic experimental autoimmune encephalomyelitis and suppress macrophage infiltration. Clin Exp Immunol. 2006;145(1):101–107. doi: 10.1111/j.1365-2249.2006.03094.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Vandermeeren M, Janssens S, Borgers M, Geysen J. Dimethylfumarate is an inhibitor of cytokine-induced E-selectin, VCAM-1, and ICAM-1 expression in human endothelial cells. Biochem Biophys Res Commun. 1997;234(1):19–23. doi: 10.1006/bbrc.1997.6570. [DOI] [PubMed] [Google Scholar]
  • 93.Linker RA, Lee DH, Ryan S, et al. Fumaric acid esters exert neuroprotective effects in neuroinflammation via activation of the Nrf2 antioxidant pathway. Brain. 2011;134(3):678–692. doi: 10.1093/brain/awq386. [DOI] [PubMed] [Google Scholar]
  • 94.Kohen R, Nyska A. Oxidation of biological systems: oxidative stress phenomena, antioxidants, redox reactions, and methods for their quantification. Toxicol Pathol. 2002;30(6):620–650. doi: 10.1080/01926230290166724. [DOI] [PubMed] [Google Scholar]
  • 95.Scannevin RH, Chollate S, Jung MY, et al. Fumarates promote cytoprotection of central nervous system cells against oxidative stress via the nuclear factor (erythroid-derived 2)-like 2 pathway. J Pharmacol Exp Ther. 2012;341(1):274–284. doi: 10.1124/jpet.111.190132. [DOI] [PubMed] [Google Scholar]
  • 96.Schimrigk S, Brune N, Hellwig K, et al. Oral fumaric acid esters for the treatment of active multiple sclerosis: an open-label, baseline-controlled pilot study. Eur J Neurol. 2006;13(6):604–610. doi: 10.1111/j.1468-1331.2006.01292.x. [DOI] [PubMed] [Google Scholar]
  • 97.Wakkee M, Thio HB. Drug evaluation: BG-12, an immunomodulatory dimethylfumarate. Curr Opinion Investig Drugs. 2007;8(11):955–962. [PubMed] [Google Scholar]
  • 98.Kappos L, Gold R, Miller DH, et al. Efficacy and safety of oral fumarate in patients with relapsing-remitting multiple sclerosis: a multicentre, randomised, double-blind, placebo-controlled phase IIb study. Lancet. 2008;372(9648):1463–1472. doi: 10.1016/S0140-6736(08)61619-0. [DOI] [PubMed] [Google Scholar]
  • 99.Gold R, Kappos L, Arnold DL, et al. Placebo-controlled phase 3 study of oral BG-12 for relapsing multiple sclerosis. N Engl J Med. 2012;367(12):1098–1107. doi: 10.1056/NEJMoa1114287. [DOI] [PubMed] [Google Scholar]
  • 100.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. doi: 10.1056/NEJMoa1206328. [DOI] [PubMed] [Google Scholar]
  • 101.Hutchinson M, Fox RJ, Miller DH, et al. Clinical efficacy of BG-12 (dimethyl fumarate) in patients with relapsing-remitting multiple sclerosis: subgroup analyses of the CONFIRM study. Journal of Neurology. 2013;260(9):2286–2296. doi: 10.1007/s00415-013-6968-1. [DOI] [PubMed] [Google Scholar]
  • 102.Ermis U, Weis J, Schulz JB. PML in a patient treated with fumaric acid. N Engl J Med. 2013;368(17):1657–1658. doi: 10.1056/NEJMc1211805. [DOI] [PubMed] [Google Scholar]
  • 103.van Oosten BW, Killestein J, Barkhof F, Polman CH, Wattjes MP. PML in a patient treated with dimethyl fumarate from a compounding pharmacy. N Engl J Med. 2013;368(17):1658–1659. doi: 10.1056/NEJMc1215357. [DOI] [PubMed] [Google Scholar]
  • 104.Sweetser MT, Dawson KT, Bozic C. Manufacturer’s response to case reports of PML. N Engl J Med. 2013;368(17):1659–1661. doi: 10.1056/NEJMc1300283. [DOI] [PubMed] [Google Scholar]
  • 105.Zeyda M, Poglitsch M, Geyeregger R, et al. Disruption of the interaction of T cells with antigen-presenting cells by the active leflunomide metabolite teriflunomide: involvement of impaired integrin activation and immunologic synapse formation. Arthritis Rheum. 2005;52(9):2730–2739. doi: 10.1002/art.21255. [DOI] [PubMed] [Google Scholar]
  • 106.O’Connor PW, Li D, Freedman MS, et al. A Phase II study of the safety and efficacy of teriflunomide in multiple sclerosis with relapses. Neurology. 2006;66(6):894–900. doi: 10.1212/01.wnl.0000203121.04509.31. [DOI] [PubMed] [Google Scholar]
  • 107.Warnke C, Meyer zu Hörste G, Hartung HP, Stüve O, Kieseier BC. Review of teriflunomide and its potential in the treatment of multiple sclerosis. Neuropsychiatr Dis Treat. 2009;5:333–340. [PMC free article] [PubMed] [Google Scholar]
  • 108.Fox RI, Herrmann ML, Frangou CG, et al. Mechanism of action for leflunomide in rheumatoid arthritis. Clin Immunol. 1999;93(3):198–208. doi: 10.1006/clim.1999.4777. [DOI] [PubMed] [Google Scholar]
  • 109.Herrmann ML, Schleyerbach R, Kirschbaum BJ. Leflunomide: an immunomodulatory drug for the treatment of rheumatoid arthritis and other autoimmune diseases. Immunopharmacology. 2000;47(2–3):273–289. doi: 10.1016/s0162-3109(00)00191-0. [DOI] [PubMed] [Google Scholar]
  • 110.Oh J, O’Connor PW. Teriflunomide for the treatment of multiple sclerosis. Semin Neurol. 2013;33(1):45–55. doi: 10.1055/s-0033-1343795. [DOI] [PubMed] [Google Scholar]
  • 111.Iglesias-Bregna D, Hanak S, Ji Z, et al. Effects of prophylactic and therapeutic teriflunomide in transcranial magnetic stimulation-induced motor-evoked potentials in the Dark Agouti rat model of experimental autoimmune encephalomyelitis. J Pharmacol Exp Ther. 26 doi: 10.1124/jpet.113.205146. Epub Jul. [DOI] [PubMed] [Google Scholar]
  • 112.Merrill JE, Hanak S, Pu SF, et al. Teriflunomide reduces behavioral, electrophysiological, and histopathological deficits in the Dark Agouti rat model of experimental autoimmune encephalomyelitis. J Neurol. 2009;256(1):89–103. doi: 10.1007/s00415-009-0075-3. [DOI] [PubMed] [Google Scholar]
  • 113.Sanofi Phase III Study With Teriflunomide Versus Placebo in Patients With First Clinical Symptom of Multiple Sclerosis (TOPIC) [Accessed September 27, 2013]. Available from: http://clinicaltrials.gov/show/NCT00622700. Identifier: NCT00622700.
  • 114.Olsson T, Zhi WW, Höjeberg B, et al. Autoreactive T lymphocytes in multiple sclerosis determined by antigen-induced secretion of interferon-gamma. J Clin Invest. 1990;86(3):981–985. doi: 10.1172/JCI114800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Mandala S, Hajdu R, Bergstrom J, et al. Alteration of lymphocyte trafficking by sphingosine-1-phosphate receptor agonists. Science. 2002;296(5566):346–349. doi: 10.1126/science.1070238. [DOI] [PubMed] [Google Scholar]
  • 116.Yopp AC, Randolph GJ, Bromberg JS. Leukotrienes, sphingolipids, and leukocyte trafficking. J Immunol. 2003;171(1):5–10. doi: 10.4049/jimmunol.171.1.5. [DOI] [PubMed] [Google Scholar]
  • 117.O’Sullivan C, Dev KK. The structure and function of the S1P1 receptor. Trends Pharmacol Sci. 2013;34(7):401–412. doi: 10.1016/j.tips.2013.05.002. [DOI] [PubMed] [Google Scholar]
  • 118.Xie JH, Nomura N, Koprak SL, Quackenbush EJ, Forrest MJ, Rosen H. Sphingosine-1-phosphate receptor agonism impairs the efficiency of the local immune response by altering trafficking of naive and antigen-activated CD4+ T cells. J Immunol. 2003;170(7):3662–3670. doi: 10.4049/jimmunol.170.7.3662. [DOI] [PubMed] [Google Scholar]
  • 119.Yanagawa Y, Masubuchi Y, Chiba K. FTY720, a novel immunosuppressant, induces sequestration of circulating mature lymphocytes by acceleration of lymphocyte homing in rats, III. Increase in frequency of CD62L-positive T cells in Peyer’s patches by FTY720-induced lymphocyte homing. Immunology. 1998;95(4):591–594. doi: 10.1046/j.1365-2567.1998.00639.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Yanagawa Y, Sugahara K, Kataoka H, Kawaguchi T, Masubuchi Y, Chiba K. FTY720, a novel immunosuppressant, induces sequestration of circulating mature lymphocytes by acceleration of lymphocyte homing in rats. II. FTY720 prolongs skin allograft survival by decreasing T cell infiltration into grafts but not cytokine production in vivo. J Immunol. 1998;160(11):5493–5499. [PubMed] [Google Scholar]
  • 121.Chiba K, Yanagawa Y, Masubuchi Y, et al. FTY720, a novel immuno-suppressant, induces sequestration of circulating mature lymphocytes by acceleration of lymphocyte homing in rats. I. FTY720 selectively decreases the number of circulating mature lymphocytes by acceleration of lymphocyte homing. J Immunol. 1998;160(10):5037–5044. [PubMed] [Google Scholar]
  • 122.Fujino M, Funeshima N, Kitazawa Y, et al. Amelioration of experimental autoimmune encephalomyelitis in Lewis rats by FTY720 treatment. J Pharmacol Exp Ther. 2003;305(1):70–77. doi: 10.1124/jpet.102.045658. [DOI] [PubMed] [Google Scholar]
  • 123.Miron VE, Ludwin SK, Darlington PJ, et al. Fingolimod (FTY720) enhances remyelination following demyelination of organotypic cerebellar slices. Am J Pathol. 2010;176(6):2682–2694. doi: 10.2353/ajpath.2010.091234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.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. doi: 10.1056/NEJMoa0909494. [DOI] [PubMed] [Google Scholar]
  • 125.Cohen JA, Barkhof F, Comi G, et al. Oral fingolimod or intramuscular interferon for relapsing multiple sclerosis. N Engl J Med. 2010;362(5):402–415. doi: 10.1056/NEJMoa0907839. [DOI] [PubMed] [Google Scholar]
  • 126.Lindsey JW, Haden-Pinneri K, Memon NB, Buja LM. Sudden unexpected death on fingolimod. Mult Scler. 2012;18(10):1507–1508. doi: 10.1177/1352458512438456. [DOI] [PubMed] [Google Scholar]
  • 127.Ratchford JN, Costello K, Reich DS, Calabresi PA. Varicella-zoster virus encephalitis and vasculopathy in a patient treated with fingolimod. Neurology. 2012;79(19):2002–2004. doi: 10.1212/WNL.0b013e3182735d00. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Rukovets O. FDA investigates PML in patient taking Gilenya. 2013. [Accessed September 17, 2013]. Available from: http://journals.lww.com/neurotodayonline/blog/breakingnews/pages/post.aspx?PostID=240.
  • 129.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. doi: 10.1002/ana.410390304. [DOI] [PubMed] [Google Scholar]
  • 130.Randomised double-blind placebo-controlled study of interferon beta-1a in relapsing/remitting multiple sclerosis. PRISMS (Prevention of Relapses and Disability by Interferon beta-1a Subcutaneously in Multiple Sclerosis) Study Group. Lancet. 1998;352(9139):1498–1504. [No authors listed] [PubMed] [Google Scholar]
  • 131.O’Connor P, Wolinsky JS, Confavreux C, et al. Randomized trial of oral teriflunomide for relapsing multiple sclerosis. N Engl J Med. 2011;365(14):1293–1303. doi: 10.1056/NEJMoa1014656. [DOI] [PubMed] [Google Scholar]
  • 132.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. doi: 10.1016/S0140-6736(12)61769-3. [DOI] [PubMed] [Google Scholar]
  • 133.Comi G, Jeffery D, Kappos L, et al. Placebo-controlled trial of oral laquinimod for multiple sclerosis. N Engl J Med. 2012;366(11):1000–1009. doi: 10.1056/NEJMoa1104318. [DOI] [PubMed] [Google Scholar]
  • 134.Gold R, Giovannoni G, Selmaj K, et al. Daclizumab high-yield process in relapsing-remitting multiple sclerosis (SELECT): a randomised, double-blind, placebo-controlled trial. Lancet. 2013;381(9884):2167–2175. doi: 10.1016/S0140-6736(12)62190-4. [DOI] [PubMed] [Google Scholar]
  • 135.Kappos L, Li D, Calabresi PA, et al. Ocrelizumab in relapsing-remitting multiple sclerosis: a phase 2, randomised, placebo- controlled, multicentre trial. Lancet. 2011;378(9805):1779–1787. doi: 10.1016/S0140-6736(11)61649-8. [DOI] [PubMed] [Google Scholar]

Articles from Biologics : Targets & Therapy are provided here courtesy of Dove Press

RESOURCES