11,322
Views
57
CrossRef citations to date
0
Altmetric
Neurology

Systematic literature review and network meta-analysis of cladribine tablets versus alternative disease-modifying treatments for relapsing–remitting multiple sclerosis

, , , , &
Pages 1361-1371 | Received 26 Oct 2017, Accepted 16 Nov 2017, Published online: 28 Nov 2017

Abstract

Objective: To assess the comparative efficacy and safety of cladribine tablets versus alternative disease modifying treatments (DMTs) in patients with active relapsing–remitting multiple sclerosis (RRMS), and in a subgroup with high disease activity (HRA + DAT), using systematic literature review (SLR) and network meta-analysis (NMA).

Methods: MEDLINE, Embase, MEDLINE In-Process and CENTRAL databases were systematically searched to identify English-language publications of relevant studies of approved DMTs for RRMS. Searches were conducted from database inception to January 2017. Conference websites and trial registries were also searched. NMA considered the effects of DMTs on annualized relapse rate (ARR), confirmed disease progression (CDP), no evidence of disease activity (NEDA) and safety.

Results: Of 10,825 articles retrieved and screened, 44 studies assessing 12 DMTs contributed to the NMA. In patients with active RRMS, cladribine tablets were associated with a significant 58% reduction in ARR versus placebo (p < .05); cladribine tablets were similar or significantly better than other DMT regimens and ranked fourth among DMTs, behind alemtuzumab, natalizumab and ocrelizumab. For CDP for 6 months and NEDA, improvements with cladribine tablets were significantly greater than those of placebo (p < .05), with no comparator DMT demonstrating significantly better results. Similar findings were reported in the HRA + DAT population. Overall adverse event risk for cladribine tablets did not differ significantly from that of placebo and most alternative DMTs.

Conclusion: In this first NMA to consider cladribine tablets, ocrelizumab and daclizumab for treatment of RRMS, cladribine tablets are a comparatively effective and safe alternative to other DMTs in both active RRMS and HRA + DAT populations.

Introduction

Multiple sclerosis (MS) is a chronic, inflammatory, neuro-degenerative, autoimmune disorder of the central nervous system, in which multifocal lymphocytic infiltration causes demyelination and axonal damage. Approximately 85% of patients with MS present with the relapsing–remitting form of the disease (RRMS), which is characterized by periods of remission attenuated by sporadic relapsesCitation1,Citation2. Relapses are unpredictable events characterized by acute symptoms including vision changes, loss of co-ordination, and changes in bowel and bladder function that may stabilize over days or weeks before partially or completely resolvingCitation1.

RRMS can be treated with disease-modifying treatments (DMTs) to suppress autoimmune responseCitation1–3. Treatment options include intramuscular (interferon beta 1a [Avonex]) subcutaneous (interferon betas, glatiramer acetate, and daclizumab), infused (ocrelizumab, alemtuzumab [two 5 and 3 day cycles one year apart] and natalizumab), and oral (e.g. fingolimod, dimethyl fumarate and teriflunomide) treatments. The benefit and harms of DMTs have been studied across a large number of randomized controlled trials (RCTs) that included placebo or interferon beta as a comparator. Whilst these studies provide valuable insight into the use of DMTs in RRMS, alone they fail to address the question of the relative efficacy and safety of different DMTs. With the lack of head-to-head trials between DMTs, indirect comparisons and network meta-analyses (NMAs) have been performed to bridge this evidence gap and help inform clinical decision makingCitation4–7. Over time, these analyses require updating as new evidence emerges, new treatment options gain approval and new outcome measures gain relevance.

In August 2017, the European Medicines Agency (EMA) approved cladribine tablets, an oral, monotherapy, fixed-course regimen comprising two short courses given over two years, as a treatment for adult patients with RMS with highly active disease as defined by clinical or imaging featuresCitation8,Citation9. Cladribine tablets were granted approval based on the results of two RCTs that compared cladribine tablets versus placebo (CLARITY)Citation10 and cladribine tablets plus interferon versus placebo plus interferon (ONWARD)Citation11. In subgroup analyses of the CLARITY data, cladribine tablets showed consistent efficacy in patients with high disease activity (HRA + DAT), defined as patients with ≥2 relapses in the previous year, whether on Disease modifying drug (DMD) treatment or not (high relapse activity; HRA) plus patients with ≥1 relapse and ≥1 T1 Gd + lesion or ≥9 T2 lesions in the previous year while on therapy with other DMDs (disease activity on treatment, DAT)Citation9. As with other DMTs, the benefits and harms associated with use of cladribine tablets as an alternative to other treatments for RRMS requires evaluation.

In a RCT setting, the most common primary efficacy endpoint is annualized relapse rate (ARR)Citation2. Reduction in relapses alone is, however, not sufficient in daily practice, and in both RCTs and clinical settings, other measures of effect are considered, including brain lesion load from imaging data, and disease progression on the Expanded Disability Status Scale (EDSS). A combined endpoint representing freedom from relapse, progression and lesion load, termed “no evidence of disease activity” (NEDA), has also emerged providing a measure of the efficacy of DMT in stopping all forms of disease activity.

The aim of this analysis was to assess the comparative efficacy and safety of cladribine tablets versus alternative DMTs in patients with RRMS. A systematic literature review (SLR) and NMA were performed using methods similar to those of previous studies. The NMA focuses on the effects of DMTs on ARR and disability progression (confirmed disease progression [CDP]), and also includes the newer endpoint, NEDA, and safety.

Methods

Literature search

A SLR was performed following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses statement (PRISMA), and the requirements of health technology appraisal organizations in the United States, United Kingdom, Germany and France. The search strategy and review protocol are provided in Supplementary Tables 1 and 2, respectively.

MEDLINE, Embase, MEDLINE In-Process and CENTRAL databases were systematically searched using a combination of medical subject headings (MeSH) and free-text terms to identify English-language publications of relevant studies of approved DMTs for RRMS. Searches were conducted from database inception to January 2017 (date of search). Conference abstracts were included and identified via Embase or hand-searches of relevant conference websites. Trial registries (e.g. ClinicalTrials.gov and European Clinical Trial Registry [EUCTR]), the United States Food and Drug Administration (FDA) and EMA websites were also searched. Data for cladribine tablets was extracted from available clinical trial reports, and additional post-hoc analyses of the PRISMS study were performed to fill evidence gaps.

Study selection and data extraction

Citations retrieved through the literature search were screened for inclusion in the review based on the following prospectively defined eligibility criteria: RCTs of DMTs approved by United States or European regulatory agencies in adult patients with RRMS, or a patient population with subgroup of ≥80% RRMS patients. Screening was performed by two independent reviewers in two stages, including an initial abstract and title review followed by full text screening. Two independent reviewers extracted relevant data from the included studies. Any discrepancies between reviewers in screening or data extraction were reconciled by a third independent reviewer.

Assessment of risk of bias and heterogeneity of included trials

The risk of bias was assessed using National Institute for Health and Care Excellence (NICE)Citation12, German Institut für Qualität und Wirtschaftlichkeit im Gesundheitswesen (IQWiG)Citation13, and French Haute Autorité de Santé (HAS)Citation14 checklists, and by study grade (for adequacy of concealment of allocation) and Jadad score (for study quality and study reporting)Citation15. Sources of clinical heterogeneity were summarized and assessed.

Network meta-analysis

The comparative efficacy and safety of cladribine tablets versus alternative DMTs was assessed via a series of Bayesian network meta-analyses (NMA) based on the methods of Dias et al.Citation16, and performed in WinBUGS version 1.4.3.

The analysis included both random and fixed-effect models, with results reported for the best fitting model according to residual deviance (closest total residual deviance to the number of data points) and deviance information criterion (DIC) (lowest being best)Citation16. Each analysis used three independent Monte Carlo Markov chains, each containing a sample of 100,000 simulations after a burn-in of 20,000. Vague prior distributions were assigned to study-level treatment effects, baseline outcomes and between-study heterogeneity parameters. The posterior of the between-trial standard deviation was used to assess between-study heterogeneity. The level of consistency between direct and indirect evidence was tested using the loop-specific approach reported by Veroniki et al.Citation17.

Separate NMAs were performed on the efficacy endpoints of ARR, CDP sustained for 6 months at 24 months (CDP6M), CDP sustained for 3 months at 24 months (CDP3M), proportion relapse-free at 24 months and NEDA at 24 months. For brevity, results for relapse-free and CDP3M analyses are reported in Supplementary file 1. The safety analysis was performed on the proportion of patients with any adverse events (AEs) as reported in each RCT. Treatment effect estimates comprised relapse rate ratio (RR) for ARR, hazard ratios (HR) for CDP6M and CDP3M, and odds ratios (OR) for NEDA and safety endpoints. The number needed to treat (NNT) and number needed to harm (NNH) comparing DMT versus placebo were also reported. Summaries included posterior effect size, 95% credible intervals, Bayesian p-values, and median rank in line with ISPOR guidelinesCitation18. A Bayesian p-value of <.05 was used to determine statistical significance.

Where feasible, the efficacy analyses were performed on the intent-to-treat (ITT) populations of included studies, and on the subgroup with HRA + DAT. The ITT population was commonly defined as all patients with ≥1 relapse in the previous year or ≥2 relapses in the previous two years (active RRMS). Where data from patients with HRA + DAT was not reported, available subgroup data for patients with HRA or patients with DAT were used and generalized to the full HRA + DAT population. Safety analyses were performed on ITT populations only, as safety data specific to subgroups are rarely reported.

A comprehensive set of sensitivity analyses were performed to assess the impact on results of excluding outlier studies, studies that utilized Poser diagnostic criteria or had an unclear diagnostic criteria, studies published prior to 2000, open-label studies or studies for which blinding status was unclear, and phase II studies (Supplementary file 2). These analyses were supplemented by a meta-regression on baseline risk to explore the impact of between-study heterogeneity on treatment effect. Differences in patient characteristics among trials are likely to reflect heterogeneity in baseline risk, and hence act as a surrogate for underlying factors. Due to data demands, meta-regression analyses were restricted to the active RRMS efficacy analysis.

Results

Systematic literature review

A total of 10,825 articles were retrieved and screened following a search run on 4 January 2017. Forty-nine trials, from 778 publications and two clinical trial reports, met the inclusion criteria and were included in the review. Of these, 44 studies assessing 12 DMTs contributed to the NMA ()Citation19–61. Design details of these studies are summarized in Supplementary Tables 3 and 4.

Figure 1. PRISMA diagram showing the flow of studies through the systematic review process. Citation1Five of the 49 studies were excluded because they evaluated unlicensed treatments (n = 2), did not include patients with active RRMS (n = 2) or did not report any outcomes of interest (n = 1). Abbreviations. CSR, clinical study report; HRA + DAT, high disease activity, defined as patients with ≥2 relapses in the previous year, whether on DMD treatment or not (high relapse activity; HRA) plus patients with ≥1 relapse and ≥1 T1 Gd + lesion or ≥9 T2 lesions in the previous year while on therapy with other DMDs (disease activity on treatment, DAT); ITT, intent-to-treat; DMD: disease modifying drug; SGA: Sub-group analysis.

Figure 1. PRISMA diagram showing the flow of studies through the systematic review process. Citation1Five of the 49 studies were excluded because they evaluated unlicensed treatments (n = 2), did not include patients with active RRMS (n = 2) or did not report any outcomes of interest (n = 1). Abbreviations. CSR, clinical study report; HRA + DAT, high disease activity, defined as patients with ≥2 relapses in the previous year, whether on DMD treatment or not (high relapse activity; HRA) plus patients with ≥1 relapse and ≥1 T1 Gd + lesion or ≥9 T2 lesions in the previous year while on therapy with other DMDs (disease activity on treatment, DAT); ITT, intent-to-treat; DMD: disease modifying drug; SGA: Sub-group analysis.

Study and patient characteristics

The 44 studies included in the review were published between 1987 and 2017 and showed variability in diagnostic criteria, blinding, study phase, sample size (31 to 2244), year of publication (Supplementary Table 3; Supplementary Figure 1(a)), and the definitions of “relapse” and disability progression. The majority of studies defined relapse as new or worsening symptoms that lasted at least 24 hours and occurred in the absence of fever or infection. Disability progression was commonly defined as an increase in EDSS of ≥1 point, or ≥0.5 point for baseline EDSS ≥5.5, confirmed during two subsequent neurological examinations separated by an interval of at least 3–6 months free of relapses. The duration of the randomized phase of each study ranged from 12 to 260 weeks; 24 of the 44 studies reported a randomized phase of ≥100 weeks’ duration (Supplementary Table 3). Treatment regimens evaluated are summarized in , which also reports the regimen abbreviations used in this paper.

Table 1. Treatment regimens evaluated in the network meta-analysis.

The majority of included RCTs enrolled adult patients with RRMS who had a baseline EDSS score of ≤6 and had experienced at least one relapse in the preceding year or two relapses in the previous 2 years (Supplementary Table 3). Baseline characteristics were generally comparable across included studies with the notable exception of baseline disease duration, which ranged from 1.2 to 9.1 years (Supplementary Figures 1(b) and (c)).

Risk of bias in the included studies

Across included studies, the risk of bias was generally low according to NICE, IQWiG and HAS checklists (Supplementary File 3). Exceptions were the open-label study of IFN beta-1a 44 mcg versus IFN beta-1bCitation49 and studies evaluating alemtuzumabCitation20–22, which were all single-, assessor-blinded and considered at higher risk of bias. Included studies were generally of good quality in terms of reporting, despite a notable absence of reporting on the clinical significance of study findings.

Network meta-analysis – efficacy

The evidence networks showing the DMT comparisons contributing to the efficacy analyses in patients with active RRMS are presented in Supplementary Figure 2. In total, 41 studies reported data for patients with active RRMSCitation19–21,Citation23–29,Citation31–52,Citation54–61 and 11 studies reported data for patients with HRA + DATCitation20–22,Citation28,Citation29,Citation38,Citation39,Citation43,Citation56,Citation58,Citation59. Of the 11 HRA + DAT studies, only two reported data for the full HRA + DAT populationCitation43,Citation59. Eight of the nine remaining studies reported data from patients with HRA only and one study reported data from patients with DAT only. Overall results of the NMA for clinical endpoints are summarized in .

Table 2. Summary of efficacy results between cladribine tablets and comparators.

Annualized relapse rate

Forty-one studies contributed to the active RRMS NMA for ARR (the random-effects model was best fit), and 11 studies contributed to the HRA + DAT NMA for ARR (fixed-effects model).

In the active RRMS population, cladribine tablets were associated with a statistically significant reduction (p < .05) in ARR of 58% versus placebo, which translated to a significant reduction of 36–48% when compared with both regimens of teriflunomide, all IFN beta regimens and both GA regimens (; ). There were no statistically significant difference in ARR between cladribine tablets and DMF, fingolimod, daclizumab HYP, alemtuzumab, natalizumab or ocrelizumab. The top four DMTs based on rank were alemtuzumab, natalizumab, ocrelizumab and cladribine tablets (). Results of the NNT analysis were consistent with those of the RR analysis (). The median NNT to avoid one relapse event (versus placebo) over a 2 year period was 2 for alemtuzumab and natalizumab, 3 for cladribine tablets, ocrelizumab, daclizumab HYP, fingolimod and DMF, and ranged from 5 to 8 for all other DMTs ().

Figure 2. Summary plot of efficacy risk ratios for cladribine tablets versus comparators. (a) Annualized relapse rate. (b) Confirmed disease progression for 6 months at 24 months.

Figure 2. Summary plot of efficacy risk ratios for cladribine tablets versus comparators. (a) Annualized relapse rate. (b) Confirmed disease progression for 6 months at 24 months.

Table 3. Summary of annualized relapse rate results in the ITT population.

In the HRA + DAT population, cladribine tablets were associated with a significant (p < .05) reduction in ARR of 65% versus placebo, which translated to significant reductions in ARR of 49–56% versus teriflunomide 7 mg, IFN beta-1a 30 mcg, IFN beta-1a 44 mcg and GA 20 mg. There was no significant difference in ARR between cladribine tablets and alemtuzumab, and fingolimod, and non-significant but numerical reductions of 34% and 37% versus DMF and teriflunomide 14 mg, respectively. No comparisons were possible versus ocrelizumab and daclizumab HYP due to lack of published subgroup data. Overall, cladribine tablets were ranked second behind natalizumab. The NNT to avoid one relapse event in patients with HRA + DAT were consistent with results in the active RRMS across all DMTs.

Confirmed disease progression sustained for 6 months at 24 months

Twenty studies contributed to the active RRMS NMA for CDP6M at 24 months (random-effects model), and four studies contributed to the HRA + DAT NMA (fixed-effects model).

In patients with active RRMS, cladribine tablets demonstrated improvements in CDP6M that were significantly greater than placebo (HR = 0.54, p < .05) and statistically comparable to other DMTs (; ). Cladribine tablets was ranked highest of all other oral DMTs including DMF, fingolimod and both teriflunomide regimens and was ranked amongst the top 50% of all licensed MS therapies alongside alemtuzumab, ocrelizumab, natalizumab and daclizumab HYP. It should be noted that IFN beta-1b was also ranked amongst the top 50% of all licensed MS DMTs; however, data informing this analysis is derived from the INCOMIN study and this result is discussed later. The NNT to avoid one progression event versus placebo over a 2 year period was 11 for cladribine tablets, and ranged from 8 to 22 for other DMTs ().

Table 4. Summary of confirmed disease progression sustained for 6 months at 24 months results in the ITT population.

In patients with HRA + DAT, cladribine tablets were associated with a statistically significant 82% reduction in the rate of CDP6M versus placebo, translating to a 61–68% reduction in CDP6M versus alemtuzumab and IFN-beta 1a 44 mcg (p < .05). Cladribine tablets were ranked first ahead of alemtuzumab and IFN-beta 1a 44 mcg. No other DMT comparisons were possible in patients with HRA + DAT due to a lack of published subgroup data.

No evidence of disease activity at 24 months

Five studies contributed to the active RRMS NMA of NEDA at 24 months (fixed-effects model). No NMA was performed for patients with HRA + DAT as only the CLARITY study reported NEDA in this population (cladribine tablets versus placebo).

In patients with active RRMS, cladribine tablets were associated with significantly increased odds of NEDA versus placebo (OR = 4.69) that translated to significant benefits for cladribine tablets versus teriflunomide 14 mg/7 mg (OR = 2.00/2.84), DMF (OR = 2.72) and GA 20 mg (3.39) (). There was a non-significant but numerically increased odds of NEDA with natalizumab versus cladribine tablets (OR = 0.64). No other DMT comparisons were possible due to lack of published data. Cladribine tablets were ranked second behind natalizumab. The NNT for an additional NEDA versus placebo was 2 for natalizumab, 3 for cladribine tablets, and ranged from 6 to 19 for other DMTs ().

Table 5. Summary of no evidence of disease activity at 24 months results in the ITT population.

Sensitivity analyses

Results of the sensitivity analyses are presented in Supplementary file 2. There was no change in the direction of relative treatment differences between cladribine tablets and comparators, although in some instances the significance level of findings changed. The results of the meta-regression on baseline risk were in line with the NMA suggesting that variations in baseline risk had a limited overall effect on results. Sensitivity analyses were not conducted for NEDA at 24 months because insufficient studies were included in the network.

Consistency between direct and indirect evidence was assessed for ARR and CDP6M, with results suggesting that evidence were generally consistent within each network. The exception being the ARR comparisons of placebo versus IFN beta-1a 44 mcg versus teriflunomide 7 mg. Consistency testing was not feasible for NEDA at 24 months due to a three arm trial contribution in the closed loop.

Network meta-analysis – safety, all active relapsing–remitting multiple sclerosis populations

Twenty-five studies contributed to the NMA (random) for any AE (Supplementary Figure 3). The definition of any AE varied across studies with differences in cut-off limits (e.g. >10% of population) and the inclusion of relapse due to multiple sclerosis as an AE. A summary of the results of the safety NMA is provided in .

Table 6. Summary of adverse events results in the ITT population.

The results indicate no statistically significant difference in overall AE risk for cladribine tablets versus placebo and most alternative DMTs. The overall AE risk of cladribine tablets was statistically comparable to other oral DMTs, all IFN-beta regimens and GA 40 mg. There was a numerical but non-significant increased overall AE risk with alemtuzumab (OR = 0.27), peg IFN-beta (OR = 0.51), and ocrelizumab versus cladribine tablets (OR = 0.67). The overall AE risk for cladribine tablets was numerically increased versus natalizumab (OR = 2.70) and GA 20 mg (OR = 2.23). When ranked according to lowest overall AE risk, cladribine tablets were eighth and ahead of alemtuzumab (sixteenth). The NNH for DMTs versus placebo ranged from 9 (alemtuzumab) to 29 (teriflunomide 7 mg and fingolimod), with natalizumab and GA 20 mg being associated with a reduced AE risk versus placebo and NNT to avoid any AE of 11 and 18 respectively. The NNH for cladribine tablets was 19, compared with 20 for DMF, and 29 for fingolimod and teriflunomide 7 mg.

Discussion

The results of the SLR and NMA indicate that cladribine tablets are a relatively effective and safe alternative to other DMTs in both active RRMS and HRA + DAT populations. The active RRMS analysis suggests cladribine tablets are amongst the most effective oral DMTs and have an overall AE risk profile that is not significantly different from alternative DMTs. In this population, infusion therapies including alemtuzumab and ocrelizumab were associated with numerical but non-significant benefits on ARR and CDP6M versus cladribine tablets, with a higher overall AE risk. In patients with HRA + DAT, the benefits of cladribine tablets versus placebo from CLARITY translated to an increased effect versus all other DMTs, with cladribine tablets having efficacy greater than or comparable to alemtuzumab and natalizumab. These analyses demonstrate cladribine tablets as an alternative to other DMTs in an active RRMS population, and further highlight the value of this treatment in those with high unmet need, such as the HRA + DAT population. These results are representative of the clinical trial evidence for cladribine tablets.

The choice of DMT depends on numerous factors including tolerability and safety profile, patient preference, ease of administration and efficacy. Cladribine tablets are the first oral therapy to be given as a fixed course treatment, providing persons with RRMS an alternative treatment approach without daily therapy or infusions. This is the first review and NMA that considers cladribine tablets in comparison with other DMTs, and it combines both efficacy and safety endpoints to inform clinical decisions on the use of this new treatment option. A novel aspect of this analysis was the consideration of NNT and NNH as a way of comparing the harms and benefits of different DMTs. A positive benefit–risk profile is characterized by a lower NNT than NNH, with greater differences in NNT and NNH indicating improved benefit–risk profileCitation62. As expected, the NNT for relapse and progression for all DMTs including cladribine tablets was consistently lower than or equal to its corresponding NNH, supporting a positive benefit–risk profile for DMT. In general, the benefit–risk profile of cladribine tablets was comparable or favorable to alternative DMTs. For disease progression, the benefit risk of cladribine tablets (NNT/NNH: 11/19) in patients with active RRMS appeared to be more favorable than the numerically more effective strategies of alemtuzumab (NNT/NNH: 9/9) and ocrelizumab (NNT/NNH: 9/12). The AE risk profile differs across DMTs and hence consideration of the individual risk is an important aspect of treatment decision-making.

This is the first NMA to consider the endpoint of NEDA, which represents no activity on EDSS, relapse or lesion imaging. This endpoint has not been routinely included as part of RCTs and hence there is limited data currently available. Of the available evidence, cladribine tablets were significantly more effective than other oral DMTs (DMF and teriflunomide) and of comparable efficacy to natalizumab. These analyses should be revisited when further evidence on the effect of DMTs such as alemtuzumab, fingolimod, ocrelizumab and daclizumab on NEDA becomes available. This is particularly relevant as the goal of treatment in RRMS increasingly moves towards inducing no disease activity, and endpoints such as NEDA become increasingly relevant benchmarks of treatment effect.

The results of our NMA are comparable to a previous analysisCitation6 in demonstrating that, on average, the most effective treatments according to CDP and ARR are infusion-based therapies, followed by oral treatments and injectable therapies. No previous NMA have included cladribine tablets, and none considered recently published evidence for ocrelizumab and daclizumab. The present NMA also includes unpublished post-hoc analysis of CDP6M for interferon beta-1a versus placebo from PRISMS. This analysis was performed to improve the level of evidence available for CDP6M in both active RRMS and HRA + DAT populations, and to improve the evidence connecting alemtuzumab and ocrelizumab, both of which were studied versus interferon beta-1a, to the rest of the network.

As in Fogarty et al.Citation6, IFN beta-1b was found to be numerically superior to all other DMTs for CDP6M at 24 months, while the results for CDP3M indicated that cladribine tablets were better than IFN beta-1b (Supplementary file 1). The variation in our results might be due to inclusion in the CDP6M analyses of the INCOMIN trialCitation33, which reported a statistically significantly improvement in CDP for IFN beta-1b compared with IFN beta-1a (RR [95% CI]: 0.44 [0.25–0.80]). The improvement in CDP6M for IFN beta-1b reported in a single study by Durelli et al.Citation33 is in contrast to results seen for improvements in CDP3M for IFN beta-1b reported in two other studies that show comparable efficacy for IFN beta-1b versus placebo and glatiramer acetateCitation40,Citation45.

There are limitations associated with this current review. As with all NMA, these analyses relied on the similarity of studies to generate exchangeable treatment effects. The trials included in this NMA were conducted over a 30 year period (1987 to 2017), during which time the treatment algorithm, as well as the diagnosis criteria, for RRMS has changed; this will impact the generalizability of our resultsCitation63. The trials also differed in terms of study characteristics (diagnostic criteria, study phase and blinding), the patient populations recruited (mean relapses in prior year, disease duration, treatment history – previously treated versus treatment naïve) and definitions of outcomes. The NMA for patients with HRA + DAT is also limited by variations in the definition of HRA + DAT across studies, a lack of subgroup-specific baseline characteristic data to assess potential for effect modifiers and uncertainty surrounding effect sizes, as highlighted in Huisman et al.Citation7.

The safety findings must be interpreted with caution. The intention was to provide an overall AE risk profile using evidence available from RCTs. The reporting of any AE varied across studies leading to heterogeneity and potential bias in the analysis. This included the consideration of relapse of multiple sclerosis as an AE. In the AFFIRM studyCitation29, this resulted in a higher overall risk of AE with placebo than active treatment, leading to natalizumab appearing safer than other DMTs in the NMA. Additionally, not all relevant AEs are captured in RCTs, such as the cases of progressive multifocal leukoencephalopathy (PML) with natalizumab, DMF and fingolimod reported from post-surveillance monitoring. Further, the overall “any AE” risk category does not differentiate among the individual AEs associated with each DMT, nor consider the effect of different AEs on overall patient wellbeing. Hence, when considering the potential for harm with DMTs, further information on the risk profile of each DMT should be taken into account alongside its overall risk profile.

Conclusions

This is the first NMA to consider recently approved treatments for RRMS, cladribine tablets, ocrelizumab and daclizumab. The results of this analysis show that cladribine tablets are a comparatively effective and safe alternative to other DMTs in both the active RRMS and HRA + DAT populations.

Transparency

Declaration of funding

This work was supported by Merck KGaA, Darmstadt, Germany.

Author contributions: All authors were involved in the conception and design of the study. All authors provided analysis and interpretation of the data, critically revised the manuscript for intellectual content and provided final approval of the submitted version.

Declaration of financial/other relationships

M.K.S. has disclosed that she/he is an employee of Parexel International, Chandigarh, India. I.S.K. has disclosed that she/he is an employee of Parexel International, Chandigarh, India. S.B. has disclosed that she/he is an employee of Parexel International, London, UK. R.H. has disclosed that she/he has no significant relationships with or financial interests in any commercial companies related to this study or article. G.H. has disclosed that she/he is an employee of Merck KGaA, Middlesex, UK. S.L.W. has disclosed that she/he is an employee of EMD Serono Ltd., Billerica, MA, USA and is also a stock shareholder of EMD Serono, a business of Merck KGaA, Darmstadt, Germany.

CMRO peer reviewers on this manuscript have received an honorarium from CMRO for their review work, but have no relevant financial or other relationships to disclose.

Supplemental material

Supplemental Material (Figure 3)

Download MS Word (76.3 KB)

Supplemental Material (Figure 2)

Download MS Word (182.3 KB)

Supplemental Material (Figure 1)

Download MS Word (258 KB)

Supplemental Material (Table 4)

Download MS Word (43.3 KB)

Supplemental Material (Table 3)

Download MS Word (93.4 KB)

Supplemental Material (Table 2)

Download MS Word (23 KB)

Supplemental Material (Table 1)

Download MS Word (26.9 KB)

Supplemental Material (File 3)

Download MS Word (56.9 KB)

Supplemental Material (File 2)

Download MS Word (625.7 KB)

Supplemental Material (File 1)

Download MS Word (150.8 KB)

Acknowledgements

Medical writing support was provided by Mark OConnor and Caroline Spencer of inScience Communications, Springer Healthcare, London, UK, and was supported by Merck KGaA, Darmstadt, Germany.

References

  • Loma I, Heyman R. Multiple sclerosis: pathogenesis and treatment. Curr Neuropharmacol 2011;9:409-16
  • Guideline on Clinical Investigation of Medicinal Products for the Treatment of Multiple Sclerosis. Doc. Ref. EMA/CHMP/771815/2011, Rev. 2. London, UK: European Medicines Agency (EMA) Committee for Medicinal Products for Human Use (CHMP), 26 March 2015. Available at: http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2015/03/WC500185161.pdf [Last accessed 14 June 2017]
  • Compston A, Coles A. Multiple Sclerosis. Lancet 2008;372:1502-17
  • Mendes D, Alves C, Batel-Marques F. Benefit–risk of therapies for relapsing–remitting multiple sclerosis: testing the number needed to treat to benefit (NNTB), number needed to treat to harm (NNTH) and the likelihood to be helped or harmed (LHH): a systematic review and meta-analysis. CNS Drugs 2016;30:909-29
  • Tolley K, Hutchinson M, You X, et al. A network meta-analysis of efficacy and evaluation of safety of subcutaneous pegylated interferon beta-1a versus other injectable therapies for the treatment of relapsing–remitting multiple sclerosis. PLoS One 2015;10:e0127960
  • Fogarty E, Schmitz S, Tubridy N, et al. Comparative efficacy of disease-modifying therapies for patients with relapsing remitting multiple sclerosis: Systematic review and network meta-analysis. Mult Scler Relat Disord 2016;9:23-30
  • Huisman E, Papadimitropoulou K, Jarrett J, et al. Systematic literature review and network meta-analysis in highly active relapsing-remitting multiple sclerosis and rapidly evolving severe multiple sclerosis. BMJ Open 2017;7:e013430
  • European Medicines Agency. EPAR summary for the public. Mavenclad. London, UK. Available at: http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Summary_for_the_public/human/004230/WC500234564.pdf [Last accessed 13 October 2017]
  • European Medicines Agency. London, UK: Merck Serono Europe Limited. Mavenclad Summary of Product Characteristics. Available at: http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Product_Information/human/004230/WC500234561.pdf [Last accessed 2 October 2017]
  • Giovannoni G, Comi G, Cook S, et al. A placebo-controlled trial of oral cladribine for relapsing multiple sclerosis. New Engl J Med 2010;362:416-26
  • Montalban X, Cohen B, Leist T, et al. Efficacy of cladribine tablets as add-on to IFN-beta therapy in patients with active relapsing MS: final results from the phase II ONWARD study (P3.029). Neurology 2016;85(16 Suppl):P3.029
  • Specification for manufacturer/sponsor submission of evidence. NICE STA. June 2012. Available at: http://www.nice.org.uk/aboutnice/howwework/devnicetech/SpecificationForManufacturerSponsorSubmissionOfEvidence.jsp?domedia = 1&mid = 97E2DC44-19B9-E0B5-D4F73F1DB153AA86 [Last accessed 22 May 2017]
  • The benefit assessment of pharmaceuticals in accordance with the German Social Code, Book Five (SGB V), section 35a. G-BA; 2011. Available at: http://www.g-ba.de/institution/themenschwerpunkte/arzneimittel/nutzenbewertung35a/anlagen/#abschnitt-3. German [Last accessed 22 May 2017]
  • Analysis of the literature and recommendations grading guide [in French]. HAS, 2000. Available at: http://www.has-sante.fr/portail/jcms/c_266796/fr/analyse-litterature-medicale-guide-methodologique-2000pdf. [Last accessed 22 May 2017]
  • Jadad AR, Moore RA, Carroll D, et al. Assessing the quality of reports of randomized clinical trials: is blinding necessary? Control Clin Trials 1996;17:1-12
  • Dias S, Sutton A, Ades A, et al. Evidence synthesis for decision making 2: a generalized linear modeling framework for pairwise and network meta-analysis of randomized controlled trials. Med Decis Making 2013;33:607-17
  • Veroniki AA, Mavridis D, Higgins JP, et al. Characteristics of a loop of evidence that affect detection and estimation of inconsistency: a simulation study. BMC Med Res Methodol 2014;14:106
  • Jansen JP, Fleurence R, Devine B, et al. Interpreting indirect treatment comparisons and network meta-analysis for health-care decision making: report of the ISPOR Task Force on Indirect Treatment Comparisons Good Research Practices: part 1. Value Health 2011;14:417-28
  • Calabresi PA, Radue EW, Goodin D, et al. Safety and efficacy of fingolimod in patients with relapsing-remitting multiple sclerosis (FREEDOMS II): a double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Neurol 2014;13:545-56
  • Cohen JA, Coles AJ, Arnold DL, et al. Alemtuzumab versus interferon beta 1a as first-line treatment for patients with relapsing-remitting multiple sclerosis: a randomised controlled phase 3 trial. Lancet 2012;380:1819-28
  • Coles AJ, Twyman CL, Arnold DL, et al. Alemtuzumab for patients with relapsing multiple sclerosis after disease-modifying therapy: a randomised controlled phase 3 trial. Lancet 2012;380:1829-39
  • Coles AJ, Compston DA, Selmaj KW, et al. Alemtuzumab vs. interferon beta-1a in early multiple sclerosis. N Engl J Med 2008;359:1786-801
  • Khan O, Rieckmann P, Boyko A, et al. Three times weekly glatiramer acetate in relapsing–remitting multiple sclerosis. Ann Neurol 2013;73:705-13
  • Cohen J, Belova A, Selmaj K, et al. Equivalence of generic glatiramer acetate in multiple sclerosis: a randomized clinical trial. JAMA Neurol 2015;72:1433-41
  • Cadavid D, Wolansky LJ, Skurnick J, et al. Efficacy of treatment of MS with IFNbeta-1b or glatiramer acetate by monthly brain MRI in the BECOME study. Neurology 2009;72:1976-83
  • 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;45:1268-76
  • Lublin FD, Cofield SS, Cutter GR, et al. Randomized study combining interferon and glatiramer acetate in multiple sclerosis. Ann Neurol 2013;73:327-40
  • 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;367:1098-107
  • Polman CH, O’Connor PW, Havrdova E, et al. A randomized, placebo-controlled trial of natalizumab for relapsing multiple sclerosis. N Engl J Med 2006;354:899-910
  • Mokhber N, Azarpazhooh A, Orouji E, et al. Cognitive dysfunction in patients with multiple sclerosis treated with different types of interferon beta: a randomized clinical trial. J Neurol Sci 2014;342:16-20
  • Calabrese M, Bernardi V, Atzori M, et al. Effect of disease-modifying drugs on cortical lesions and atrophy in relapsing–remitting multiple sclerosis. Mult Scler 2012;18:418-24
  • Comi G, Filippi M, Wolinsky JS. European/Canadian multicenter, double-blind, randomized, placebo-controlled study of the effects of glatiramer acetate on magnetic resonance imaging – measured disease activity and burden in patients with relapsing multiple sclerosis. European/Canadian Glatiramer Acetate Study Group. Ann Neurol 2001;49:290-7
  • Durelli L, Verdun E, Barbero P, et al. Every-other-day interferon beta-1b versus once-weekly interferon beta-1a for multiple sclerosis: results of a 2-year prospective randomised multicentre study (INCOMIN). Lancet 2002;359:1453-60
  • Knobler RL, Greenstein JI, Johnson KP, et al. Systemic recombinant human interferon-beta treatment of relapsing–remitting multiple sclerosis: pilot study analysis and six-year follow-up. J Interferon Res 1993;13:333-40
  • Schwid SR, Panitch HS. Full results of the Evidence of Interferon Dose-Response-European North American Comparative Efficacy (EVIDENCE) study: a multicenter, randomized, assessor-blinded comparison of low-dose weekly versus high-dose, high-frequency interferon beta-1a for relapsing multiple sclerosis. Clin Ther 2007;29:2031-48
  • Mikol DD, Barkhof F, Chang P, et al. Comparison of subcutaneous interferon beta-1a with glatiramer acetate in patients with relapsing multiple sclerosis (the REbif vs Glatiramer Acetate in Relapsing MS Disease [REGARD] study): a multicentre, randomised, parallel, open-label trial. Lancet Neurol 2008;7:903-14
  • O’Connor P, Wolinsky JS, Confavreux C, et al. Randomized trial of oral teriflunomide for relapsing multiple sclerosis. N Engl J Med 2011;365:1293-303
  • 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;367:1087-97
  • 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;362:387-401
  • O’Connor P, Filippi M, Arnason B, et al. 250 microg or 500 microg interferon beta-1b versus 20 mg glatiramer acetate in relapsing–remitting multiple sclerosis: a prospective, randomised, multicentre study. Lancet Neurol 2009;8:889-97
  • Calabresi PA, Kieseier BC, Arnold DL, et al. Pegylated interferon β-1a for relapsing–remitting multiple sclerosis (ADVANCE): a randomised, phase 3, double-blind study. Lancet Neurol 2014;13:657-65
  • Kappos L, Li D, Calabresi PA, et al. Ocrelizumab in relapsing–remitting multiple sclerosis: a phase 2, randomised, placebo-controlled, multicentre trial. Lancet 2011;378:1779-87
  • Ebers GC, Rice G, Lesaux J, et al. 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;352:1498-504
  • 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;39:285-94
  • Duquette P, Girard M, Despault L, et al. 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;43:655-61
  • De Stefano N, Sormani MP, Stubinski B, et al. Efficacy and safety of subcutaneous interferon β-1a in relapsing-remitting multiple sclerosis: further outcomes from the IMPROVE study. J Neurol Sci 2012;312:97-101
  • 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;381:2167-75
  • Kappos L, Wiendl H, Selmaj K, et al. Daclizumab HYP versus interferon beta-1a in relapsing multiple sclerosis. N Engl J Med 2015;373:1418-28
  • Singer B, Bandari D, Cascione M, et al. Comparative injection-site pain and tolerability of subcutaneous serum-free formulation of interferonβ-1a versus subcutaneous interferonβ-1b: results of the randomized, multicenter, phase IIIb REFORMS study. BMC Neurol 2012;12:154
  • Bornstein MB, Miller A, Slagle S, et al. A pilot trial of Cop 1 in exacerbating-remitting multiple sclerosis. N Engl J Med 1987;317:408-14
  • Etemadifar M, Janghorbani M, Shaygannejad V. Comparison of Betaferon, Avonex, and Rebif in treatment of relapsing–remitting multiple sclerosis. Acta Neurol Scand 2006;113:283-7
  • 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;66:894-900
  • Wroe SJ. Effects of dose titration on tolerability and efficacy of interferon beta-1b in people with multiple sclerosis. J Int Med Res 2005;33:309-18
  • Saida T, Kikuchi S, Itoyama Y, et al. A randomized, controlled trial of fingolimod (FTY720) in Japanese patients with multiple sclerosis. Mult Scler 2012;18:1269-77
  • Vollmer TL, Sorensen PS, Selmaj K, et al. A randomized placebo-controlled phase III trial of oral laquinimod for multiple sclerosis. J Neurol 2014;261:773-83
  • Confavreux C, O’Connor P, Comi G, et al. Oral teriflunomide for patients with relapsing multiple sclerosis (TOWER): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Neurol 2014;13:247-56
  • Vermersch P, Czlonkowska A, Grimaldi LM, et al. Teriflunomide versus subcutaneous interferon beta-1a in patients with relapsing multiple sclerosis: a randomised, controlled phase 3 trial. Mult Scler 2014;20:705-16
  • Cohen JA, Barkhof F, Comi G, et al. Oral fingolimod or intramuscular interferon for relapsing multiple sclerosis. N Engl J Med 2010;362:402-15
  • Merck Serono. CLARITY clinical trial report 2010 [Last accessed 22 May 2017]
  • Hauser SL, Bar-Or A, Comi G, et al. Ocrelizumab versus interferon beta-1a in relapsing multiple sclerosis. N Engl J Med 2017;376:221-34
  • Saida T, Kira JI, Kishida S, et al. Efficacy, safety, and pharmacokinetics of natalizumab in Japanese multiple sclerosis patients: a double-blind, randomized controlled trial and open-label pharmacokinetic study. Mult Scler Relat Disord 2017;11:25-31
  • Guo JJ, Pandey S, Doyle J, et al. A review of quantitative risk–benefit methodologies for assessing drug safety and efficacy – report of the ISPOR risk–benefit management working group. Value Health 2010;13:657-66
  • Klawiter EC, Cross AH, Naismith RT. The present efficacy of multiple sclerosis therapeutics: is the new 66% just the old 33%? Neurology 2009;73:984-90

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.