Authors

  1. Costello, Kathleen

Abstract

ABSTRACT: Multiple sclerosis (MS) is a complex disease that affects the central nervous system. It is believed to be an immune mediated disease, and although the etiology remains unknown, it is believed to occur from a combination of genetic risk factors and environmental risk factors. There is no single diagnostic test for MS, and diagnostic criteria have been developed to aid the provider in making an accurate and timely diagnosis. Once a diagnosis of MS is made, treatments directed toward the inflammatory immune response should be initiated. Currently, there are 10 treatments for MS: four interferon beta products; one glatiramer acetate; one monoclonal antibody-natalizumab; three oral treatments-fingolimod, teriflunomide, and dimethyl fumarate; and one immunosuppressant agent-mitoxantrone. Each of these agents has a different administration and different risks and side effects. Numerous agents are in late stage development, and it is possible that several more agents, all with different mechanisms of action, will become available over the next several years.

 

Article Content

Multiple sclerosis (MS) is a chronic disease of the central nervous system (CNS) pathologically characterized by CNS inflammation, demyelination, and axonal loss (Compston & Coles, 2008). Clinically, the disease is most often characterized by early relapses and remissions of neurological symptoms referable to the CNS, a clinical course known as relapsing-remitting MS (RRMS). After early relapses with characteristic inflammation and demyelination, there is some functional reorganization and remyelination that occurs early in the disease and helps to restore function. Over time, RRMS often transitions to a disease course characterized by a progressive decline in function. Although relapses may still occur, they occur less often; this course of MS is known as secondary progressive MS. In individuals with secondary progressive MS, early repair mechanisms fail and more axonal loss occurs (Hagemeier, Bruck, & Kuhlmann, 2012). Approximately 10%-15% of all patients with MS will have a progressive course of the disease from onset, known as primary progressive MS (Lublin & Reingold, 1996).

 

The etiology of MS is unknown; however, it is thought to be because of a complex interplay of factors including genetic risk for autoimmunity, immune system dysregulation, and environmental factors (i.e., viral exposure, particularly Epstein Barr, vitamin D deficiency, and smoking; Ascherio & Munger, 2007; Giovannoni & Ebers, 2007; Shirani & Tremlett, 2010). Immunologically, the inflammatory process is thought to be mediated by T-cell and B-cell responses to CNS autoantigens. The inflammation that occurs within the CNS is also responsible, at least in part, for degenerative changes, including axonal loss and damage to oligodendrocytes that lead to irreversible damage in the brain and spinal cord.

 

Symptoms of MS vary widely in type and severity. Common symptoms include generalized and frequently overwhelming fatigue; depression; unilateral visual blurring or loss; diplopia; sensory symptoms, such as numbness, tingling, or pain; incoordination; spasticity; weakness; ataxia; and elimination dysfunction, such as urgency, frequency, and incontinence of bladder or bowel. Cognitive impairment, such as short-term memory loss and slowed processing, is also possible.

 

Diagnostics

Making the diagnosis of MS can be quite challenging because there is no specific diagnostic test. Traditionally, the diagnosis is made if there are at least two neurological events referable to the CNS, separated or disseminated in space (different areas of the CNS) and time (events occur at different points in time), with objective evidence of CNS involvement and all other possible causes of the symptoms are excluded. The diagnosis is mostly based on history of neurological symptoms and neurological examination findings. More recently, magnetic resonance imaging (MRI) findings and other paraclinical testing, such as spinal fluid analysis, have been used to establish dissemination in space and time when two clinical events have not occurred but MS is believed to be the most likely diagnosis.

 

Since the mid-1960s, there have been published diagnostic criteria developed by MS experts that have helped with the diagnosis of MS. In 2001, an international panel of MS experts convened and developed criteria that utilized both clinical and MRI findings, including the criteria of dissemination in space and time (McDonald et al., 2001). Since 2001, these criteria, known as the McDonald criteria, have been revised, both in 2005 (Polman et al., 2005) and in 2010 (Polman et al., 2011). In each of the revisions, the MRI criteria have been modified to reflect increased evidence of MRI changes consistent with MS.

 

One of the goals of these criteria is to expedite the diagnosis of MS. Therefore, the McDonald diagnostic criteria have traditionally been applied to those individuals who present with a clinically isolated syndrome (CIS). The CIS is a term that describes the first onset of neurological symptoms in an individual, which usually involve the optic nerve, brainstem, or spinal cord and are characteristic of MS (Miller et al., 2008). Symptoms should last a minimum of 24 hours in the absence of infection. Individuals who have symptoms and an MRI suggestive of inflammation and demyelination or spinal fluid with findings consistent with MS are considered at high risk to develop additional symptoms and would then be considered to have MS. The diagnostic criteria utilize paraclinical evidence of MRI and/or cerebrospinal fluid abnormalities to establish dissemination in space and/or time when only a single clinical event has occurred. Imaging criteria for the 2010 McDonald criteria revision were based on recent publications from the MAGNIMS research group (Montalban et al., 2010; Rovira et al., 2009; Swanton et al., 2007). Fluid-attenuated inversion recovery and T2-weighted sequences are the MRI techniques that are currently of greatest clinical interest in MS, along with postcontrast T1-weighted scans. These sequences provide information about active inflammatory and subclinical MS activity. Subclinical activity occurs much more frequently than do clinical symptoms (Filippi et al., 2011).

 

On the basis of the McDonald criteria, the criterion for dissemination in space can be met by the presence of an MRI lesion in at least two of four areas of the CNS, which include juxtacortical, periventricular, infratentorial, or spinal cord (Figure 1). The criterion of dissemination in time can be met with an additional T2 lesion and/or gadolinium (Gd)-enhancing lesion on a subsequent MRI, performed at any time after the initial MRI, or the simultaneous presence of asymptomatic Gd-enhancing lesions and nonenhancing lesions in an individual with CIS (Polman et al., 2011). If the criteria for dissemination in space and time are met in the absence of additional clinical symptoms and other causes of the symptoms have been completely excluded, the diagnosis of MS can be made (Polman et al., 2011). Other possible causes of neurological symptoms that must be excluded include other inflammatory conditions, CNS infections, metabolic conditions, vascular conditions, and inherited conditions. Table 1 lists some of the diagnostic mimics of MS.

  
Table 1 - Click to enlarge in new windowTABLE 1 MS Mimics (Cohen & Rensel, 2000)
 
Figure 1 - Click to enlarge in new windowFIGURE 1. MRI in Multiple Sclerosis

Although not part of the MRI diagnostic criteria nor part of the conventional imaging of clinical practice, nonconventional imaging techniques have been utilized to identify additional CNS pathology in MS. These techniques have added to the overall understanding of the disease. Although traditionally thought to be a white matter disease, gray matter abnormalities have been identified through the use of high-field-strength MRI (Stadelmann, Albert, Wegner, & Bruck, 2008). Calabrese and colleagues examined 59 patients with CIS and showed that gray matter atrophy in the superior frontal gyrus, thalamus, and cerebellum were independent predictors of conversion from CIS to MS (Calabrese et al., 2011). In addition, gray matter atrophy in patients with MS is associated with progression of MS (Fisniku et al., 2008).

 

Imaging techniques, such as brain parenchymal fraction and structural image evaluation using normalization of atrophy, provide different methods to quantify brain atrophy. These techniques have shown that brain atrophy measurements correlate better with disability than do conventional MRI techniques. It has been suggested that atrophy measurements may be a useful way of measuring the ability of disease-modifying treatments (DMTs) to provide neuroprotection (Barkhof, Calabresi, Miller, & Reingold, 2009).

 

Other MRI techniques utilized in the research arena are magnetic transfer imaging (MT-MRI) and MR spectroscopy. MT-MRI is a technique that measures the interaction of free water protons and bound protons and provides information about areas of MS damage as well as normal-appearing white matter (Filippi & Rocca, 2007). MR spectroscopy provides quantification of chemical changes found in lesions and normal-appearing white matter. Considered a marker of axonal integrity, N-acetyl aspartate can be measured using MR spectroscopy and provides evidence of the extent of axonal damage (Poloni, Minagar, Haacke, & Zivadinov, 2011).

 

Optical coherence tomography (OCT), a non-MRI imaging tool, is a noninvasive method to observe and quantify the retinal nerve fiber layer thickness. The retinal nerve is unmyelinated; thus, OCT is able to quantify axonal integrity and damage. Gordon-Lipkin and colleagues showed that OCT correlated with brain atrophy in MS (Gordon-Lipkin et al., 2007). This imaging tool may be an important outcome measurement in clinical trials of neuroprotection because it provides information about axonal integrity.

 

DMTs

Once the diagnosis of CIS or MS is made, treatment can be considered. DMTs, available since the early 1990s, are able to alter the natural history of RRMS because of their anti-inflammatory effect on the immune system. These agents have shown efficacy in the reduction of relapse frequency and reduction in CNS inflammation and have a variable ability to reduce the progression of disability. Emerging treatments in the research pipeline promise alternate mechanisms of action from the currently approved treatments as well as efficacy in relapse reduction, inflammatory events, and progression of disability.

 

Currently, there are 10 Food and Drug Administration (FDA)-approved treatments for MS, including four interferon beta preparations, glatiramer acetate, natalizumab, fingolimod, mitoxantrone, teriflunomide, and dimethyl fumarate (DMF; Table 2). The interferons, glatiramer acetate, natalizumab, teriflunomide, fingolimod, and DMF are indicated for RRMS. Mitoxantrone is indicated for secondary progressive MS, worsening relapsing MS, and progressive relapsing MS. In well-designed clinical trials, the interferon preparations and glatiramer acetate have been shown to delay the conversion of CIS to MS (Comi et al., 2009, 2012a; Jacobs et al., 2000; Kappos et al., 2006a). Two separate head-to-head trials showed superiority in reducing relapses and improving MRI outcomes for high-dose/high-frequency interferon beta compared with weekly interferon beta-1a (Durelli et al., 2002; Panitch et al., 2005). Two other separate head-to-head trials of interferon beta and glatiramer acetate showed a similar effect on relapse outcomes (Mikol et al., 2008; O'Connor et al., 2009).

  
Table 2 - Click to enlarge in new windowTABLE 2 FDA-Approved Disease-Modifying Treatments for MS (

Unfortunately, in secondary progressive MS and primary progressive MS, the clinical trial results of the available agents were not as positive. Only the European trial of interferon beta-1b showed a favorable effect on progression in secondary progressive MS (PRISMS [Prevention of Relapses and Disability by Interferon beta-1a Subcutaneously in Multiple Sclerosis] Study Group, 1998). The North American trial of interferon beta-1b in secondary progressive MS was unable to corroborate those results (Panitch et al., 2004). Upon further evaluation of the two trials, it was apparent that the populations studied in the two trials were slightly different, with the European cohort having more inflammatory activity (Derwenskus, 2011). A study of glatiramer acetate in primary progressive MS was stopped after the interim analysis showed that the drug failed to affect progression (Wolinsky et al., 2007). A trial in secondary progressive MS of interferon beta-1a subcutaneous injection was also negative with respect to effect on progression (Kappos, Polman, Pozzilli, Thompson, & Dahlke, 1998).

 

The interferon preparations are slightly different in that interferon beta-1b is produced in modified E. coli cells and interferon beta-1a is produced in mammalian cells. Both interferon beta-1b and interferon beta-1a work similarly by reducing peripheral antigen presentation and T-cell proliferation. Furthermore, interferon beta may downregulate adhesion molecules at the level of the blood brain barrier. All interferon preparations have shown efficacy in relapse reduction and disability outcomes. In addition, whether in phase 3 regulatory trials or subsequent trials, interferon preparations have shown efficacy for improving MRI outcome measures and have indicated favorable long-term safety (Herndon et al., 2005; Kappos et al., 2006b; Reder et al., 2010).

 

Glatiramer acetate is a synthetic polypeptide similar to myelin basic protein. It is thought to work by provoking the immune system to shift to a less inflammatory response. In several clinical trials, glatiramer acetate has shown a favorable effect on relapses and MRI outcomes. An open-label extension of the original phase 3 trial is ongoing and has established the long-term safety of glatiramer acetate (Ford et al., 2010).

 

More recent additions to the MS treatment armamentarium are natalizumab, fingolimod, teriflunomide, and DMF. Natalizumab is indicated for RRMS and is recommended in patients who have had a suboptimal response to injectable treatments or cannot tolerate other treatments. Natalizumab blocks VLA-4, an adhesion molecule found on activated lymphocytes. Without adhesion to the blood vessel wall, activated lymphocytes are unable to traffic into the CNS. Natalizumab was studied in two large clinical trials and, in both, was shown to have a statistically significant effect on relapses, disability progression, and MRI outcomes (Polman et al., 2006; Rudick et al., 2006). Natalizumab is associated with progressive multifocal leukoencephalopathy (PML), a rare infection of the CNS caused by the John Cunningham virus (JCV). JCV infects approximately one half of the adult population and, as a primary infection, is not associated with clinical symptoms. The virus becomes latent after the initial exposure and can become reactivated under certain conditions, such as untreated human immunodeficiency virus, immunosuppressant treatment, and treatment with certain monoclonal antibodies, including rituximab and natalizumab. Reactivated JCV can infect the CNS and causes widespread damage to myelin and oligodendrocytes, ultimately resulting in death if the causative agent is not promptly removed or eliminated (Monaco & Major, 2012). Over 300 individuals on natalizumab for MS have developed PML since 2006, and 22% of them have died. Previous exposure to JCV provokes the development of antibodies to JCV. Individuals with antibodies to JCV are more likely to develop PML. By 24 months of treatment, in antibody-positive patients with JCV, the risk of developing PML is approximately 1:200 (Monaco & Major, 2012). If an individual is JCV antibody-positive and has had previous immunosuppressant treatment, the risk of PML increases to approximately 1:100 (Monaco & Major, 2012).

 

Approved in 2010, fingolimod is the first FDA-approved oral agent for the treatment of RRMS. Fingolimod is a sphingosine 1-phosphate inhibitor that works by depriving naive and central memory T-cells of a signal to egress from secondary lymph organs. Results from two large phase 3 trials indicated that a 0.5-mg daily dose of fingolimod significantly reduced annualized relapse rate, MRI activity, and disability progression (Cohen et al., 2010; Kappos et al., 2010). Sphingosine 1-phosphate receptors are found on many different cells, and thus, unwanted side effects from fingolimod may occur. Fingolimod is associated with bradycardia, particularly on the first dose. Patients must be observed for 6 hours after the first dose for any heart rate or blood pressure issues (Ginsberg et al., 1995). An electrocardiogram is to be obtained before dosing and at the completion of the observation period. In May 2012, the FDA revised the labeling of fingolimod to include the following contraindications (Ginsberg et al., 1995):

 

* Patients who in the last 6 months experienced myocardial infarction, unstable angina, stroke, transient ischemic attack, decompensated heart failure requiring hospitalization, or Class III/IV heart failure.

 

* History or presence of Mobitz Type II second-degree or third-degree atrioventricular block or sick sinus syndrome, unless patient has a functioning pacemaker.

 

* Baseline QTc interval of >=500 milliseconds.

 

* Treatment with Class IA or Class III anti-arrhythmic drugs.

 

 

Lymphopenia and hepatic enzyme elevations are possible side effects, and laboratory monitoring is necessary. Herpes infections, including varicella zoster, were seen in clinical trials, and it is recommended that all patients have a varicella-zoster titer before the initiation of treatment and vaccination before treatment if no titer is detected. Macular edema has been observed in patients receiving fingolimod (Cohen et al., 2010; Kappos et al., 2010). Blood pressure elevations have also been observed with fingolimod use (Ginsberg et al., 1995).

 

Teriflunomide was approved for relapsing MS in September 2012. Teriflunomide is an oral treatment administered once daily. The approved doses are 7 mg once daily and 14 mg once daily. It is a derivative of leflunomide, which is FDA approved for the treatment of rheumatoid arthritis. This agent works by inhibiting dihydroorotate dehydrogenase, which is necessary for deoxyribonucleic acid replication. It reduces T-cell and B-cell activation, proliferation, and response to autoantigens. Two randomized, double-blind, placebo-controlled clinical trials met the relapse rate and disability end points (14-mg dose; Freedman et al., 2013; O'Connor et al., 2011). Both approved doses (7 and 14 mg) were shown to be superior compared with placebo for the MRI outcomes of total lesion volume and Gd enhancement (O'Connor et al., 2011). In a separate randomized, double-blind trial, teriflunomide (14 mg) was shown to be comparable with interferon beta-1a for the annualized relapse rate outcome (Vermersch et al., 2012). Adverse events reported with teriflunomide have included gastrointestinal upset, temporary hair thinning, hepatic enzyme elevation, mild neutropenia, and mild blood pressure elevations (O'Connor et al., 2011). Teriflunomide is contraindicated in pregnant patients and patients with severe hepatic impairment (Aubagio, 2013).

 

DMF, an oral treatment administered twice daily, was approved for relapsing MS in March 2013. DMF is thought to work in MS by several potential mechanisms. It has been found to induce T-cell apoptosis, potentially protect against oxidative stress, inhibit adhesion molecules, and potentially shift the immune response toward a Th-2 (helper T-cell) response (Gold, 2011; Kappos et al., 2008; Killestein et al., 2011). Results of a large, randomized, placebo-controlled, phase 3 trial of DMF in patients with RRMS showed that DMF (240 mg) was associated with significant reductions in the annualized relapse rate, a significant reduction in the probability of disability progression (based on the Expanded Disability Status Scale) and a reduction in MRI measures of disease progression (Abad, Gomez-Outes, Martinez-Gonzales, & Rocha, 2006). Side effects of DMF include flushing, bloating, diarrhea, lymphopenia, and eosinophilia (Kappos et al., 2008).

 

Emerging Therapies

There are numerous new treatments in late-stage development in the MS research pipeline. Those agents that have completed or are in phase 3 trials include laquinimod, alemtuzumab, and daclizumab. Laquinimod is an oral treatment, alemtuzumab is administered intravenously once yearly over several days, and daclizumab is administered subcutaneously every 2 or 4 weeks. Alemtuzumab is currently under FDA review for potential approval as a DMT for MS.

 

Laquinimod

Laquinimod is a small molecule related to linomide, which was tested in MS. Linomide appeared promising as an MS treatment; however, serious cardiac events resulted in the termination of clinical trials. Laquinimod, although structurally similar, has not been associated with cardiac events. The mechanism of action of laquinimod is not known, but it is believed to shift the immune response to a less inflammatory response (Gasperini & Ruggieri, 2011). Additional investigations have shown the inhibition of adhesion molecules at the level of the blood brain barrier with laquinimod (Wegner et al., 2010). Laquinimod may also affect the CNS. Laquinimod was evaluated in two large phase 3 trials of RRMS (Comi et al., 2012b; Vollmer, 2011). The first phase 3 trial, known as the ALLEGRO trial, showed a reduction in the annualized relapse rate of 23% in the laquinimod group relative to placebo (p < .0024). In addition, there was a 36% decrease in the risk for disability progression, as measured by the Expanded Disability Status Scale compared with placebo (p = .0122) and a 32.8% reduction in brain volume loss (p < .0001; Comi et al., 2012b).

 

The second phase 3 trial of laquinimod, known as the BRAVO trial, included 1,106 patients with RRMS. That trial consisted of a laquinimod arm, a placebo arm, and a rater-blinded comparator arm of interferon beta-1a. The trial failed to meet the primary end point of relapse rate reduction. It was determined that the baseline MRI characteristics of the treated and placebo groups were different, which may have accounted for the missed end point. After adjusting for the differences, the results were statistically significant, with a 21% reduction in relapses (p = .26), a 33.5% decrease in risk of disability progression (p = .044), and a 27.5% reduction in brain volume loss (p < .0001; Vollmer, 2011).

 

Side effects of laquinimod were mild and included mild liver function test elevations, mild arthralgia, elevated erythrocyte sedimentation rate, and increased rates of infection in the treated groups. There were no cardiac events reported with laquinimod treatment (Comi et al., 2010; Gasperini & Ruggieri, 2011). At this time, laquinimod will not be submitted to the FDA for approval. However, plans are underway for regulatory approval in Europe.

 

Alemtuzumab

Alemtuzumab is a humanized anti-CD52 monoclonal antibody. It has been used and is FDA approved for the treatment of chronic lymphoid leukemia. CD-52 is expressed on a variety of cell types, including T and B lymphocytes, monocytes, and macrophages; alemtuzumab depletes these cells rapidly after infusion. Alemtuzumab is dosed once yearly by intravenous infusion daily for 5 days. In the second year of clinical trials, the dosing was daily for 3 consecutive days. Alemtuzumab was evaluated in two large phase 3 trials (Comi et al., 2010; Jeffrey, 2011). Of note, both trials compared alemtuzumab with interferon beta-1a three times weekly, and there was no placebo arm in either trial. The first trial included 581 DMT-naive patients, and the results showed a 55% reduction in relapses in the alemtuzumab group relative to the interferon group (p < .0001). The disability end point was not met in that trial (Comi et al., 2010). In the second phase 3 trial, 840 patients who had breakthrough disease were eligible for participation, and again, the comparator arm was interferon beta-1a by subcutaneous injection three times a week. In this trial, there was a 49% reduction in the annualized relapse rate in the alemtuzumab arm compared with interferon beta-1a (p < .0001). The disability end point was met with a 43% reduction in sustained disability (p = .008; Jeffrey, 2011). Adverse events, including idiopathic thrombocytopenia purpura, autoimmune thyroid disease, infusion-related reactions, and increased risk for infections, were observed in the clinical trials (Jeffrey, 2011).

 

Daclizumab

Daclizumab is a monoclonal antibody against the interleukin-2 receptor that binds to CD-25. It has been found to decrease abnormal T-cell activation and may also increase the population of regulatory natural killer cells. Daclizumab has been administered by intravenous infusion and, more recently, by subcutaneous injection. Two phase 2 trials have been completed that indicate efficacy in relapse reduction and MRI outcomes (Giovannoni et al., 2011; Wynn et al., 2010). The SELECT trial was a 1-year, randomized, double-blind trial that compared 150 and 300 mg of monthly subcutaneous daclizumab to placebo. Both treatment groups reduced annual relapse rate by approximately 50% (p = .0002). New and enlarging T2 MRI lesions were reduced by 70%-78% (p < .0001; Giovannoni et al., 2011). A second phase 2 trial, the CHOICE trial, was a 24-week study of 230 patients with RRMS who had at least one relapse or Gd-enhancing lesion in the brain or spinal cord while on stable interferon beta therapy. Patients were randomized to receive high-dose daclizumab (2 mg/kg every 2 weeks subcutaneously plus interferon beta), low-dose daclizumab (1 mg/kg every 4 weeks subcutaneously plus interferon beta), or interferon beta plus placebo. The primary outcome was new or enhancing MRI lesions, and a significant 72% reduction was observed in the number of new or enlarging T2 MRI lesions in the high-dose daclizumab plus interferon beta group compared with the interferon beta plus placebo group (p = .004). Adverse events included rash and infections consisting mainly of urinary tract and respiratory tract infections (Wynn et al., 2010). Mild lymphopenia and mild elevations in hepatic enzymes have also been observed with daclizumab treatment (Martin, 2012). A phase 3 trial of daclizumab is currently in progress.

 

Summary

MS is a complex disease that causes widespread demyelination and axonal damage of white and gray matter, leading to often irreversible damage and disabling neurological symptoms. Diagnosis of MS is often difficult and requires clinical and paraclinical information, along with the exclusion of other possible causes of symptoms. An international panel has developed and revised diagnostic criteria for MS to expedite the diagnosis and to improve diagnostic accuracy.

 

Since 1993, 10 medications have received FDA approval for MS: nine for RRMS and mitoxantrone for worsening relapsing and secondary progressive MS. Interferon beta-1a weekly injections and interferon beta-1b subcutaneous injections, as well as glatiramer acetate, are approved for CIS and RRMS. These injectable treatments have shown long-term efficacy and long-term safety. The more recent treatments natalizumab, fingolimod, teriflunomide, and DMF have shown efficacy in RRMS, but their use is or may be associated with greater potential risks. The MS treatment pipeline has several treatments that have been recently submitted to the FDA for approval and in clinical development.

 

The treatment landscape for MS has expanded over the past 20 years and will likely undergo further expansion over the next several years. Although new treatments have shown efficacy in relapse reduction and MRI outcomes, treatment side effects and potential risks associated with these treatments will become important points of discussion with patients. Treatments continue to be directed toward the inflammatory response seen in MS, with outcomes of relapse reduction and reduction in new MRI activity. Some also show a positive effect on progression of disability and may have neuroprotective properties.

 

Nurses will need to understand the immunological basis of MS as well as the diagnostic criteria to help patients understand their disease and diagnosis. In addition, with the ever-growing treatment options, nurses will need to understand the mechanisms of action of MS treatments; their effects on relapses, MRI, and progression; and the risks and tolerability of these treatments to help patients make informed decisions. Nurses will also play a role in the long-term monitoring of the safety and efficacy of MS treatments to allow for the identification of patients with worsening MS who may need potential adjustments to their treatment plan.

 

Acknowledgments

Editorial support for the writing of this manuscript was provided by Megan Knagge, PhD, of MedErgy and was funded by Sanofi-Aventis. The author retained full editorial control over the content of this manuscript.

 

References

 

Abad J. I., Gomez-Outes A., Martinez-Gonzalez J., Rocha E. (2006). A prospective observational study on the effectiveness and safety of bemiparin, first dose administered 6 h after knee or hip replacement surgery. Archives of Orthopaedic and Trauma Surgery, 127, 665-670. [Context Link]

 

Ascherio A., Munger K. L. (2007). Environmental risk factors for multiple sclerosis. Part II: Noninfectious factors. Annals of Neurology, 61, 504-513. [Context Link]

 

Aubagio. (2013). Aubagio (teriflunomide) tablets for oral administration. Initial US approval: 2012. Available at http://products.sanofi.us/aubagio/aubagio.pdf[Context Link]

 

Barkhof F., Calabresi P. A., Miller D. H., Reingold S. C. (2009). Imaging outcomes for neuroprotection and repair in multiple sclerosis trials. Nature Reviews Neurology, 5, 256-266. [Context Link]

 

Calabrese M., Rinaldi F., Mattisi I., Bernardi V., Favaretto A., Perini P., Gallo P. (2011). The predictive value of gray matter atrophy in clinically isolated syndromes. Neurology, 77, 257-263. [Context Link]

 

Cohen J. A., Barkhof F., Comi G., Hartung H. P., Khatri B. O., Montalban X., Kappos L. (2010). Oral fingolimod or intramuscular interferon for relapsing multiple sclerosis. New England Journal of Medicine, 362, 402-415. [Context Link]

 

Cohen J., Rensel M. (2000). The differential diagnosis and clues to misdiagnosis. In J. S. Burks & K. P. Johnson (Eds.), Multiple sclerosis: Diagnosis, medical management and rehabilitation (pp. 127-138). New York: Demos Medical Publishing.

 

Comi G., Abramsky O., Arbizu T., Boyko A., Gold R., Havrdova E., Filippi M. (2010). Oral laquinimod in patients with relapsing-remitting multiple sclerosis: 36-week double-blind active extension of the multi-centre, randomized, double-blind, parallel-group placebo-controlled study. Multiple Sclerosis, 16, 1360-1366. [Context Link]

 

Comi G., De S. N., Freedman M. S., Barkhof F., Polman C. H., Uitdehaag B. M., Kappos L. (2012a). Comparison of two dosing frequencies of subcutaneous interferon beta-1a in patients with a first clinical demyelinating event suggestive of multiple sclerosis (REFLEX): A phase 3 randomised controlled trial. Lancet Neurology, 11, 33-41. [Context Link]

 

Comi G., Jeffery D., Kappos L., Montalban X., Boyko A., Rocca M. A., Filippi M. (2012b). Placebo-controlled trial of oral laquinimod for multiple sclerosis. New England Journal of Medicine, 366, 1000-1009. [Context Link]

 

Comi G., Martinelli V., Rodegher M., Moiola L., Bajenaru O., Carra A., Filippi M. (2009). Effect of glatiramer acetate on conversion to clinically definite multiple sclerosis in patients with clinically isolated syndrome (PreCISe study): A randomised, double-blind, placebo-controlled trial. Lancet, 374, 1503-1511. [Context Link]

 

Compston A., Coles A. (2008). Multiple sclerosis. Lancet, 372, 1502-1517. [Context Link]

 

Derwenskus J. (2011). Current disease-modifying treatment of multiple sclerosis. Mount Sinai Journal of Medicine, 78, 161-175. [Context Link]

 

Durelli L., Verdun E., Barbero P., Bergui M., Versino E., Ghezzi A., Zaffaroni M. (2002). Every-other-day interferon beta-1b versus once-weekly interferon beta-1a for multiple sclerosis: Results of a 2-year prospective randomised multicentre study (INCOMIN). Lancet, 359, 1453-1460. [Context Link]

 

Filippi M., Rocca M. A. (2007). Magnetization transfer magnetic resonance imaging of the brain, spinal cord, and optic nerve. Neurotherapeutics, 4, 401-413. [Context Link]

 

Filippi M., Rocca M. A., De S. N., Enzinger C., Fisher E., Horsfield M. A., Comi G. (2011). Magnetic resonance techniques in multiple sclerosis: The present and the future. Archives of Neurology, 68, 1514-1520. [Context Link]

 

Fisniku L. K., Chard D. T., Jackson J. S., Anderson V. M., Altmann D. R., Miszkiel K. A., Miller D. H. (2008). Gray matter atrophy is related to long-term disability in multiple sclerosis. Annals of Neurology, 64, 247-254. [Context Link]

 

Ford C., Goodman A. D., Johnson K., Kachuck N., Lindsey J. W., Lisak R., Wolinsky J. (2010). Continuous long-term immunomodulatory therapy in relapsing multiple sclerosis: Results from the 15-year analysis of the US prospective open-label study of glatiramer acetate. Multiple Sclerosis, 16, 342-350. [Context Link]

 

Freedman M., Confavreaux C., Olsson T., Comi G., Miller A., Wolinsky J., O'Connor P. (2013). Teriflunomide efficacy and safety analyses: Results from TEMSO and TOWER. Presented at: the 5th Annual Cooperative Meeting of CMSC and ACTRIMS; May 29-June 1, 2013; Orlando, FL. [Context Link]

 

Gasperini C., Ruggieri S. (2011). Emerging oral drugs for relapsing-remitting multiple sclerosis. Expert Opinion on Emerging Drugs, 16, 697-712. [Context Link]

 

Ginsberg B., Gil K. M., Muir M., Sullivan F., Williams D. A., Glass P. S. (1995). The influence of lockout intervals and drug selection on patient-controlled analgesia following gynecological surgery. Pain, 62, 95-100. [Context Link]

 

Giovannoni G., Ebers G. (2007). Multiple sclerosis: The environment and causation. Current Opinion in Neurology, 20, 261-268. [Context Link]

 

Giovannoni G., Gold R., Selmaj K., Havrdova E., Montalban X., Radue E. W., O'Neill G. (2011). A randomized, double-blind, placebo-controlled study to evaluate the safety and efficacy of daclizumab HYP monotherapy in relapsing-remitting multiple sclerosis: Primary results of the SELECT trial. Presented at the 5th Triennial Joint Meeting of the European and Americas Committees for Treatment and Research in Multiple Sclerosis; October 19-22, 2011; Amsterdam, Netherlands. Abstract 149. [Context Link]

 

Gold R. (2011). Oral therapies for multiple sclerosis: A review of agents in phase III development or recently approved. CNS Drugs, 25, 37-52. [Context Link]

 

Gordon-Lipkin E., Chodkowski B., Reich D. S., Smith S. A., Pulicken M., Balcer L. J., Calabresi P. A. (2007). Retinal nerve fiber layer is associated with brain atrophy in multiple sclerosis. Neurology, 69, 1603-1609. [Context Link]

 

Hagemeier K., Bruck W., Kuhlmann T. (2012). Multiple sclerosis-Remyelination failure as a cause of disease progression. Histology and Histopathology, 27, 277-287. [Context Link]

 

Hartung H. P., Gonsette R., Konig N., Kwiecinski H., Guseo A., Morrissey S. P., Zwingers T. (2002). Mitoxantrone in progressive multiple sclerosis: A placebo-controlled, double-blind, randomised, multicentre trial. Lancet, 360, 2018-2025. [Context Link]

 

Herndon R. M., Rudick R. A., Munschauer F. E. III., Mass M. K., Salazar A. M., Coats M. E., Riester K. (2005). Eight-year immunogenicity and safety of interferon beta-1a-Avonex treatment in patients with multiple sclerosis. Multiple Sclerosis, 11, 409-419. [Context Link]

 

Jacobs L. D., Beck R. W., Simon J. H., Kinkel R. P., Brownscheidle C. M., Murray T. J., Sandrock A. W.; the CHAMPS Study Group. (2000). Intramuscular interferon beta-1a therapy initiated during a first demyelinating event in multiple sclerosis. CHAMPS Study Group. New England Journal of Medicine, 343, 898-904. [Context Link]

 

Jacobs L. D., Cookfair D. L., Rudick R. A., Herndon R. M., Richert J. R., Salazar A. M., Whitham R. H. (1996). Intramuscular interferon beta-1a for disease progression in relapsing multiple sclerosis. The Multiple Sclerosis Collaborative Research Group (MSCRG). Annals of Neurology, 39, 285-294. [Context Link]

 

Jeffrey S. (2011). Second phase 3 trial positive with alemtuzumab in MS. Medscape Medical News. Available at: http://www.medscape.com[Context Link]

 

Johnson K. P., Brooks B. R., Cohen J. A., Ford C. C., Goldstein J., Lisak R. P., Schiffer R. B. (1995). 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, 45, 1268-1276. [Context Link]

 

Kappos L., Gold R., Miller D. H., MacManus D. G., Havrdova E., Limmroth V., O'Neill G. N. (2008). Efficacy and safety of oral fumarate in patients with relapsing-remitting multiple sclerosis: A multicentre, randomised, double-blind, placebo-controlled phase IIb study. Lancet, 372, 1463-1472. [Context Link]

 

Kappos L., Polman C., Pozzilli C., Thompson A., Dahlke F. (1998). Placebo-controlled multicentre randomised trial of interferon beta-1b in treatment of secondary progressive multiple sclerosis. European Study Group on Interferon beta-1b in Secondary Progressive MS. Lancet, 352, 1491-1497. [Context Link]

 

Kappos L., Polman C. H., Freedman M. S., Edan G., Hartung H. P., Miller D. H., Sandbrink R. (2006a). Treatment with interferon beta-1b delays conversion to clinically definite and McDonald MS in patients with clinically isolated syndromes. Neurology, 67, 1242-1249. [Context Link]

 

Kappos L., Radue E. W., O'Connor P., Polman C., Hohlfeld R., Calabresi P., Burtin P. (2010). A placebo-controlled trial of oral fingolimod in relapsing multiple sclerosis. New England Journal of Medicine, 362, 387-401. [Context Link]

 

Kappos L., Traboulsee A., Constantinescu C., Eralinna J. P., Forrestal F., Jongen P., Li D. (2006b). Long-term subcutaneous interferon beta-1a therapy in patients with relapsing-remitting MS. Neurology, 67, 944-953. [Context Link]

 

Killestein J., Rudick R. A., Polman C. H. (2011). Oral treatment for multiple sclerosis. Lancet Neurology, 10, 1026-1034. [Context Link]

 

Lublin F. D., Reingold S. C. (1996). Defining the clinical course of multiple sclerosis: Results of an international survey. National Multiple Sclerosis Society (USA) Advisory Committee on Clinical Trials of New Agents in Multiple Sclerosis. Neurology, 46, 907-911. [Context Link]

 

Martin R. (2012). Anti-CD25 (daclizumab) monoclonal antibody therapy in relapsing-remitting multiple sclerosis. Clinical Immunology, 142, 9-14. [Context Link]

 

McDonald W. I., Compston A., Edan G., Goodkin D., Hartung H. P., Lublin F. D., Wolinsky J. S. (2001). Recommended diagnostic criteria for multiple sclerosis: Guidelines from the International Panel on the diagnosis of multiple sclerosis. Annals of Neurology, 50, 121-127. [Context Link]

 

Mikol D. D., Barkhof F., Chang P., Coyle P. K., Jeffery D. R., Schwid S. R., Uitdehaag B. M. (2008). 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 Neurology, 7, 903-914. [Context Link]

 

Miller D. H., Weinshenker B. G., Filippi M., Banwell B. L., Cohen J. A., Freedman M. S., Polman C. H. (2008). Differential diagnosis of suspected multiple sclerosis: A consensus approach. Multiple Sclerosis, 14, 1157-1174. [Context Link]

 

Monaco M. C., Major E. O. (2012). The link between VLA-4 and JC virus reactivation. Expert Review of Clinical Immunology, 8, 63-72. [Context Link]

 

Montalban X., Tintore M., Swanton J., Barkhof F., Fazekas F., Filippi M., Miller D. H. (2010). MRI criteria for MS in patients with clinically isolated syndromes. Neurology, 74, 427-434. [Context Link]

 

O'Connor P., Filippi M., Arnason B., Comi G., Cook S., Goodin D., Comi G. (2009). 250 microg or 500 microg interferon beta-1b versus 20 mg glatiramer acetate in relapsing-remitting multiple sclerosis: A prospective, randomised, multicentre study. Lancet Neurology, 8, 889-897. [Context Link]

 

O'Connor P., Wolinksy J. S., Confavreux C., Comi G., Kappos L., Olsson T. P., Freedman M. S. (2011). Randomized trial of oral teriflunomide for relapsing multiple sclerosis. New England Journal of Medicine, 365, 1293-1303. [Context Link]

 

Panitch H., Goodin D., Francis G., Chang P., Coyle P., O'Connor P., Weinshenker B. (2005). Benefits of high-dose, high-frequency interferon beta-1a in relapsing-remitting multiple sclerosis are sustained to 16 months: Final comparative results of the EVIDENCE trial. Journal of the Neurological Sciences, 239, 67-74. [Context Link]

 

Panitch H., Miller A., Paty D., Weinshenker B. (2004). Interferon beta-1b in secondary progressive MS: Results from a 3-year controlled study. Neurology, 63, 1788-1795. [Context Link]

 

Polman C. H., O'Connor P. W., Havrdova E., Hutchinson M., Kappos L., Miller D. H., Sandrock A. W. (2006). A randomized, placebo-controlled trial of natalizumab for relapsing multiple sclerosis. New England Journal of Medicine, 354, 899-910. [Context Link]

 

Polman C. H., Reingold S. C., Banwell B., Clanet M., Cohen J. A., Filippi M., Wolinsky J. S. (2011). Diagnostic criteria for multiple sclerosis: 2010 revisions to the McDonald criteria. Annals of Neurology, 69, 292-302. [Context Link]

 

Polman C. H., Reingold S. C., Edan G., Filippi M., Hartung H. P., Kappos L., Wolinsky J. S. (2005). Diagnostic criteria for multiple sclerosis: 2005 revisions to the "McDonald Criteria." Annals of Neurology, 58, 840-846. [Context Link]

 

Poloni G., Minagar A., Haacke E. M., Zivadinov R. (2011). Recent developments in imaging of multiple sclerosis. Neurologist, 17, 185-204. [Context Link]

 

PRISMS (Prevention of Relapses and Disability by Interferon beta-1a Subcutaneously in Multiple Sclerosis) Study Group. (1998). Randomised double-blind placebo-controlled study of interferon beta-1a in relapsing/remitting multiple sclerosis. Lancet, 352, 1498-1504.

 

Reder A. T., Ebers G. C., Traboulsee A., Li D., Langdon D., Goodin D. S., Konieczny A. (2010). Cross-sectional study assessing long-term safety of interferon-beta-1b for relapsing-remitting MS. Neurology, 74, 1877-1885. [Context Link]

 

Rovira A., Swanton J., Tintore M., Huerga E., Barkhof F., Filippi M., Montalban X. (2009). A single, early magnetic resonance imaging study in the diagnosis of multiple sclerosis. Archives of Neurology, 66, 587-592. [Context Link]

 

Rudick R. A., Stuart W. H., Calabresi P. A., Confavreux C., Galetta S. L., Radue E. W., Sandrock A. W. (2006). Natalizumab plus interferon beta-1a for relapsing multiple sclerosis. New England Journal of Medicine, 354, 911-923. [Context Link]

 

Shirani A., Tremlett H. (2010). The effect of smoking on the symptoms and progression of multiple sclerosis: A review. Journal of Inflammation Research, 3, 115-126. [Context Link]

 

Stadelmann C., Albert M., Wegner C., Bruck W. (2008). Cortical pathology in multiple sclerosis. Current Opinion in Neurology, 21, 229-234. [Context Link]

 

Swanton J. K., Rovira A., Tintore M., Altmann D. R., Barkhof F., Filippi M., Miller D. H. (2007). MRI criteria for multiple sclerosis in patients presenting with clinically isolated syndromes: A multicentre retrospective study. Lancet Neurology, 6, 677-686. [Context Link]

 

The IFNB Multiple Sclerosis Study Group and The University of British Columbia MS/MRI Analysis Group. (1995). Interferon beta-1b in the treatment of multiple sclerosis: Final outcome of the randomized controlled trial. The IFNB Multiple Sclerosis Study Group and the University of British Columbia MS/MRI Analysis Group. Neurology, 45, 1277-1285. [Context Link]

 

Vermersch P., Czlonkowska A., Grimaldi L., Confavreux C., Comi G., Kappos L., O'Connor P.for the TENERE Trial Group. (2012). Evaluation of patient satisfaction from the TENERE study: A comparison of teriflunomide and subcutaneous interferon beta-1a in patients with relapsing multiple sclerosis. Presented at: the 22nd Meeting of the European Neurological Society; June 9-12, 2012; Prague, Czechoslovakia. [Context Link]

 

Vollmer T., Soelberg Sorensen P., Arnold D. L. (2011). A placebo-controlled and active comparator phase III trial (BRAVO) for relapsing-remitting multiple sclerosis. Multiple Sclerosis, 17, S507-S508. Abstract 148; Presented at the 5th Triennial Congress of the European and Americas Committees for Treatment and Research in Multiple Sclerosis; October 19-21, 2011; Amsterdam, Netherlands. [Context Link]

 

Wegner C., Stadelmann C., Pfortner R., Raymond E., Feigelson S., Alon R., Bruck W. (2010). Laquinimod interferes with migratory capacity of T cells and reduces IL-17 levels, inflammatory demyelination and acute axonal damage in mice with experimental autoimmune encephalomyelitis. Journal of Neuroimmunology, 227, 133-143. [Context Link]

 

Wolinsky J. S., Narayana P. A., O'Connor P., Coyle P. K., Ford C., Johnson K., Ladkani D. (2007). Glatiramer acetate in primary progressive multiple sclerosis: Results of a multinational, multicenter, double-blind, placebo-controlled trial. Annals of Neurology, 61, 14-24. [Context Link]

 

Wynn D., Kaufman M., Montalban X., Vollmer T., Simon J., Elkins J., Rose J. W. (2010). Daclizumab in active relapsing multiple sclerosis (CHOICE study): A phase 2, randomised, double-blind, placebo-controlled, add-on trial with interferon beta. Lancet Neurology, 9, 381-390. [Context Link]

 

Keywords: alemtuzumab; daclizumab; dimethyl fumarate; disease modifying treatment; fingolimod; glatiramer acetate; interferon beta; laquinimod; MCDONALD Criteria; multiple sclerosis; natalizumab; teriflunomide