Drug
Molecule
Mechanism of action
Main trials
Side effects
Daclizumab
Humanized monoclonal antibody of CD25 subunit of IL-2 receptor
Increase of the CD56 NK cell levels
1. SELECT
Phase II
2. SELECTION
Phase II extension
3. DECIDE
Phase III
ARR reduction 45 %
Infections
Cutaneous reactions
LFT elevation
Rituximab
Human-mouse chimeric anti-CD20 monoclonal antibody
B-cell depletion
1. HERMES
Phase II RRMS
91 % reduction in total T1 Gd + lesions
2. OLYMPUS
Phase II/III PPMS
Delayed time to CDP in younger individuals
Infusion-associated reactions (fever, chills, hypotension, rigors)
UTI
Sinusitis
Ocrelizumab
Recombinant humanized anti-CD20 monoclonal antibody
1. Phase II trial
600 mg and 2000 mg
89 % and 96 % reduction in total T1 Gd + lesions
ARR reduction 80 and 73 %
2. OPERA I and II
Phase III: ARR reduction 46 % vs. interferon, 95 % reduction in T1 Gd + lesions
Infusion-associated reactions
Ofatumumab
Fully human anti-CD20 antibody
1. Phase II trial
>99 % reduction in total T1 Gd + lesions
Bronchospasm
Rash
Pharyngeal edema
Erythema
5.2.1 Daclizumab
Daclizumab, a humanized monoclonal antibody (Ab) targeting the CD25 subunit of the interleukin (IL)-2 receptor, is currently approved by the FDA for use in rheumatoid arthritis and other autoimmune diseases. Initially thought to decrease T-cell expansion via a reduction in IL-2 signaling, daclizumab was subsequently found to increase the levels of circulating CD56bright natural killer (NK) cells. The contact dependent inhibitory effect of NK cells on T-cell survival is the proposed mechanism of action in relapsing MS [1]. It has been studied as a once-monthly subcutaneous injection.
After a successful Phase II study (SELECT) [2] and its 1 year extension (SELECTION) [3], the efficacy of daclizumab (monthly 150 mg subcutaneous injection) in RRMS was further assessed in a Phase III, randomized, multicenter, double-blind study, using the weekly intramuscular interferon (IFN) β-1a as an active comparator (DECIDE trial). The DECIDE trial enrolled over 1800 patients and successfully met its primary outcome by demonstrating a 45 % annualized relapse rate (ARR) reduction compared to weekly IFN β-1a (P < 0.0001). Secondary endpoints included the number of new or newly enlarging T2 lesions and the proportion of relapse-free patients. The majority of secondary endpoints were also met with statistically significant 54 % reduction in the number of new/enlarging T2 MRI lesions at week 96. The second-ordered secondary endpoint of disability progression confirmed at 12 weeks was not statistically different between the two groups. At 144 weeks, 16 % in the daclizumab high-yield process (HYP) group and 20 % in the IFN β-1a group had progressed. Although 67 % of patients were relapse-free in the daclizumab treatment arm (compared to 51 % in IFN arm) at 144 weeks, translating into relative reduction of 41 %, this was not considered significant on the basis of the prespecified hierarchical testing plan.
The adverse events and side effects reported in the DECIDE study included serious infections, which were reported in 4 % of patients (compared to 2 % in IFN arm). Patients in the daclizumab arm also had a higher incidence of cutaneous adverse events (37 % vs. 19 %) and serious cutaneous reactions (2 % vs. 1 %). Significant elevation of liver enzymes was observed in 6 % of daclizumab-treated patients (3 % in the IFN arm) [4]. It remains to be seen what type of monitoring, including liver function tests, will be recommended. The Biologics License Application requesting marketing approval of daclizumab for RRMS is currently in review process by the Food and Drug Administration (FDA) in the United States and has now been approved by the European Medicines Agency (EMA).
5.2.2 Ocrelizumab
Ocrelizumab is a B-cell depleting monoclonal Ab with binding affinity towards a specific epitope of the common B-cell surface marker CD20. As a recombinant humanized Ab, ocrelizumab is less immunogenic than the human-mouse chimeric rituximab with repeated infusions, resulting in a potentially lower rate of infusion- associated adverse reactions. Compared to rituximab, in vitro studies also suggested that ocrelizumab had greater B-cell depleting capacity. This agent is given by intravenous infusion, with cycles given every 6 months.
The Phase II, multicenter, randomized, double-blind, dose-finding study assessed efficacy and safety of ocrelizumab in 220 patients with RRMS. Two ocrelizumab dosing regimens (600 mg and 2000 mg) were compared to placebo and the study also included an active, open label, rater-masked, control weekly IFN β-1a treatment arm. Both ocrelizumab doses had statistically significant impact on the total number of gadolinium-enhancing T1 lesions, the primary outcome of this study, showing relative reduction by 89 % at 600 mg and 96 % at 2000 mg dose. Compared to placebo and IFN treatment arms, a higher proportion of participants in both ocrelizumab treatment arms remained free of gadolinium-enhancing lesions (77 % and 88 %). The ARR at 24 weeks was 80 and 73 % lower in 600 and 2000 mg ocrelizumab arms vs. placebo.
With regard to adverse events, serious infection rates were similar across all four arms. Most infusion-associated reactions were mild to moderate and occurred with greater frequency in both ocrelizumab arms (35 % in 600 mg, 44 % at 2000 mg) than in placebo (9 %), but this difference was observed only during the first infusion [5]. One patient in the ocrelizumab 2000 mg group died of a systemic inflammatory response of unknown etiology.
The OPERA I and II Phase III trials, were multicenter, randomized double-blind, double-dummy studies comparing 600 mg dose of ocrelizumab, administered intravenously every 6 months to three-times weekly IFN β-1a in patients with RRMS. Both studies successfully met the primary outcome and showed 46 % and 47 % reduction of the ARR over a 2-year period when compared to IFN β-1a. The majority of the secondary outcomes were also met; specifically, the 43 % and 37 % reduction in confirmed disability progression at 24 months and reduction in total number of gadolinium T1-enhancing lesions (94 and 95 %), as well as reduction in total number of new and/or enlarging T2H lesions (77 and 83 %).
The incidence of serious adverse events including infections did not differ between the active and comparative treatment arms (6.9 % vs. 8.7 %) and the most frequent adverse events were mild-to-moderate infusion-associated reactions (34.3 % vs. 9.7 %). The data has been submitted for review to US and EU regulatory authorities and ocrelizumab has been designated a “breakthrough therapy” by the FDA [6].
5.3 Remyelination and Repair
5.3.1 Anti-LINGO-1
While existing immunomodulatory treatment agents reduce disease activity in patients with RRMS, they are not able to facilitate repair mechanisms. Anti-LINGO-1 Ab is the first agent directed towards the repair of the existing damage in MS rather than preventing new injury. The oligodendrocytic leucine and rich repeat (LRR) and immunoglobulin-like (Ig) domain-containing neurite outgrowth inhibitor (Nogo) receptor interacting protein (LINGO-1) negatively regulates oligodendrocyte differentiation and myelination [7]. In animal studies, application of an Ab against LINGO-1 resulted in remyelination. The encouraging pre-clinical data supported further advancement into Phase II and later Phase II human studies RENEW and SYNERGY.
The RENEW study assessed the remyelination potential of anti-LINGO-1 treatment in acute optic neuritis (ON). This randomized, double-blind, placebo-controlled trial enrolled 82 patients with a first unilateral episode of ON. After completing treatment with high-dose steroids, participants in the active arm received 100 mg/kg of anti-LINGO-1 Ab intravenously every 4 weeks for six doses total. The trial met its primary outcome and demonstrated 34 % improvement in the recovery of optic nerve latency as measured by full-field visual evoked potential relative to placebo in the per protocol population (P = 0.0504) [8]. Severity and incidence of adverse events were comparable across the treatment arms. The serious adverse event profile included hypersensitivity reactions close to the time of infusion (two patients) and an asymptomatic elevation of LFTs (one patient) [8].
Another Phase II dose-finding, efficacy, and safety study of anti-LINGO-1 in patients with active RRMS and secondary progressive MS (SPMS) treated also with IFN β-1a (SYNERGY trial) is currently underway. A total of 396 patients with active RRMS or SPMS are being randomized to the active arm with intravenous anti-LINGO-1 treatment or placebo for 72 weeks as an add-on to weekly intramuscular IFN β-1a. The trial aims to evaluate sustained improvement in neurophysical and/or cognitive function for 3 months as the primary outcome; sustained worsening in function for 3 months is the key secondary outcome. Conventional and nonconventional MRI outcomes are exploratory efficacy imaging endpoints. Results of this study will inform decisions on further clinical development of anti-LINGO-1 for CNS remyelination and/or neuroaxonal protection [9]. If investigations of anti-LINGO-1 prove successful, this agent may be utilized in combination with existing disease-modifying agents for relapsing MS, to both prevent new disease activity and foster myelin repair.
5.4 Stem Cell Therapeutics
Stem cell transplantation-based therapies in MS either seek to remove disease-causing immune cells and induce a reset of the immune system, or use multipotent stem cells with neuroprotective and restorative capabilities. After encouraging results from early studies, three main concepts of stem cell therapies are currently under investigation: hematopoietic stem cell transplantation (HSCT), mesenchymal stem cell transplantation, and glial progenitor cell transplantation (Table 5.2).
Table 5.2
Stem cell therapeutics