Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Multiple Sclerosis and Related Disorders 9 (2016) 23–30

Contents lists available at ScienceDirect

Multiple Sclerosis and Related Disorders


journal homepage: www.elsevier.com/locate/msard

Comparative efficacy of disease-modifying therapies for patients


with relapsing remitting multiple sclerosis: Systematic review
and network meta-analysis
Emer Fogarty a,n, Susanne Schmitz b, Niall Tubridy c, Cathal Walsh d, Michael Barry e
a
National Centre for Pharmacoeconomics, Dublin, Ireland
b
Health Economics and Evidence Synthesis Research Unit, Department of Population Health, Luxembourg Institute of Health, Luxembourg
c
Department of Neurology, St. Vincent's University Hospital, Dublin, Ireland
d
Department of Mathematics and Statistics, University of Limerick, Ireland
e
National Centre for Pharmacoeconomics, Dublin, Ireland

art ic l e i nf o a b s t r a c t

Article history: Introduction: Randomised studies have demonstrated efficacy of disease-modifying therapies in relap-
Received 24 April 2015 sing remitting multiple sclerosis (RRMS). However it is unclear how the magnitude of treatment efficacy
Received in revised form varies across all currently available therapies.
29 April 2016
Objective: To perform a systematic review and network meta-analysis to evaluate the comparative ef-
Accepted 7 June 2016
ficacy of available therapies in reducing relapses and disability progression in RRMS.
Methods: A systematic review identified 28 randomised, placebo-controlled and direct comparative
Keywords: trials. A network meta-analysis was conducted within a Bayesian framework to estimate comparative
Network meta-analysis annualised relapse rates (ARR) and risks of disability progression (defined by both a 3-month, and
Systematic review
6-month confirmation interval). Potential sources of treatment-effect modification from study-level
Disease-modifying therapy
covariates and baseline risk were evaluated through meta-regression methods. The Surface Under the
Multiple sclerosis
Disability progression Cumulative RAnking curve (SUCRA) method was used to provide a ranking of treatments for each out-
Relapse come.
Results: The magnitude of ARR reduction varied between 15–36% for all interferon-beta products, gla-
tiramer acetate and teriflunomide, and from 50 to 69% for alemtuzumab, dimethyl fumarate, fingolimod
and natalizumab. The risk of disability progression (3-month) was reduced by 19–28% with interferon-
beta products, glatiramer acetate, fingolimod and teriflunomide, by 38–45% for pegylated interferon-
beta, dimethyl fumarate and natalizumab and by 68% with alemtuzumab. Broadly similar estimates for
the risk of disability progression (6-month), with the exception of interferon-beta-1b 250 mcg which was
much more efficacious based on this definition. Alemtuzumab and natalizumab had the highest SUCRA
scores (97% and 95% respectively) for ARR, while ranking for disability progression varied depending on
the definition of the outcome. Interferon-beta-1b 250 mcg ranked among the most efficacious treat-
ments for disability progression confirmed after six months (92%) and among the least efficacious when
the outcome was confirmed after three months (30%). No significant modification of relative treatment
effects was identified from study-level covariates or baseline risk.
Conclusion: Compared with placebo, clear reductions in ARR with disease-modifying therapies were
accompanied by more uncertain changes in disability progression. The magnitude of the reduction and
the uncertainty associated with treatment effects varied between DMTs. While natalizumab and alem-
tuzumab demonstrated consistently high ranking across outcomes, with older interferon-beta and gla-
tiramer acetate products ranking lowest, variation in disability progression definitions lead to variation
in the relative ranking of treatments. Rigorously conducted comparative studies are required to fully
evaluate the comparative treatment effects of disease modifying therapies for RRMS.
& 2016 Elsevier B.V. All rights reserved.

n
Corresponding author.
E-mail address: efogarty@stjames.ie (E. Fogarty).

http://dx.doi.org/10.1016/j.msard.2016.06.001
2211-0348/& 2016 Elsevier B.V. All rights reserved.
24 E. Fogarty et al. / Multiple Sclerosis and Related Disorders 9 (2016) 23–30

1. Introduction baseline EDSS was Z5.5, confirmed during two subsequent neu-
rological examinations separated by an interval of at least three to
Multiple sclerosis (MS) is a chronic, autoimmune, disabling six months free of relapses (Kurtzke, 1983). Disability progression
disease of the central nervous system. Relapses and disability confirmed after three months and after six months were included
progression are the clinical hallmarks of MS and the two most in the analysis as two separate outcomes.
commonly assessed clinical endpoints used to evaluate ther-
apeutic interventions in clinical trials (European Medicines Agency 2.3. Data extraction and quality assessment
(EMA) Committee for Medicinal Products for Human Use (CHMP),
2006). Relapses represent focal, acute, recurrent inflammation, Total number of treated patients, duration of follow-up, inter-
and are characterised by a gradual onset of symptoms which sta- vention and control details, ARR, proportion of patients free of
bilise over days or weeks and resolve gradually, either completely disability progression (confirmed after three months and/or six
or partially (Coles, 2009). Progression refers to the steady and ir- months) at the end of the study-period, and key population
reversible worsening of symptoms and signs independent of the baseline characteristics (age, gender, baseline EDSS score, duration
occurrence of relapses resulting from diffuse, early, chronic, pro- of disease, pre-trial relapse rate, and proportion of patients pre-
gressive neurodegeneration (Confavreux and Vukusic, 2006a). The viously treated with DMT) were extracted from all studies, where
efficacy of disease-modifying therapies (DMTs) on these clinical reported. FDA and EMA reports were used as the primary data
endpoints has been demonstrated in clinical trials over the last sources, where available. Where ARR and/or patient years of fol-
two decades, however few trials have assessed the comparative low-up were not reported, rates were derived from total number
effectiveness of DMTs. We conducted a systematic review and of patients recruited, total number of relapses, mean number of
Bayesian network meta-analysis considering all placebo-con- relapses per person and total patient years of follow-up, where
trolled and comparative trials of DMTs, to evaluate their com- reported. The quality of included RCTs was assessed using Co-
parative efficacy in reducing the risk of relapse and disability chrane Collaboration's Risk of bias tool focussing on sequence
progression in MS. generation, allocation concealment, blinding of participants and
personnel, blinding of outcome assessment, incomplete outcome
data, selective outcome reporting and ‘other issues’ (Higgins and
2. Methods Green, 2011). The overall risk of bias associated with each study
was classified as low, medium or high. High risk of bias was at-
2.1. Literature search and study selection tributed to trials with lack of blinding among participants, per-
sonnel or outcome assessors, open treatment allocation, sig-
A systematic search of the published literature was conducted nificant attrition bias due to incomplete outcome reporting.
from inception to March 2016 to identify eligible studies using Medium risk of bias was attributed to trials with unclear methods
EMBASE, MEDLINE (via PubMed) and CENTRAL (via Cochrane Li- of random sequence generation, and moderate attrition bias due to
brary) databases. In addition, the Food and Drug Administration incomplete outcome reporting. Low risk of bias was attributed to
(FDA) and European Medicines Agency (EMA) websites were trials without any bias detected in any of the seven domains, and
searched for reviews and assessment reports on interventions of in trials with unclear methods of allocation concealment which
interest. For inclusion in the analysis, randomised controlled trials were otherwise judged to have been randomised appropriately.
(RCTs) were required to include adult patients with relapsing re-
mitting MS (RRMS), report at least one of the primary outcome 2.4. Data analysis
measures of interest, and compare DMTs which are authorised by
US and/or European regulatory agencies for the treatment of A network meta-analysis was conducted combining data from
RRMS. Eleven DMTs met this criterion including interferon beta-1b trials comparing DMTs with placebo, as well as direct comparative
(IFN β-1b) subcutaneous (SC) 250 mcg, IFN β-1a SC 22 mcg and trials. Combining direct and indirect (via a common comparator)
IFN β-1a SC 44 mcg, IFN β-1a intramuscular (IM) 30 mcg, pegy- evidence in this way increases the precision of relative effect es-
lated IFN β-1a SC 125 mcg, glatiramer acetate 20 mg, glatiramer timates and allows treatments to be ranked for each of the out-
acetate 40 mg, natalizumab, alemtuzumab, fingolimod, teri- comes studied. The network meta-analysis was conducted using
flunomide, and dimethyl fumarate. The Preferred Reporting Items Bayesian Markov Chain Monte Carlo methods and fitted in R to
for Systematic Reviews and Meta-Analyses (PRISMA) statement WinBUGS (Sturtz et al., 2005; Lunn et al., 2000). Vague or non-
was followed for our network-meta analysis protocol (Moher et al., informative priors were used. Convergence and lack of auto-
2009). The full search strategy, inclusion and exclusion criteria are correlation were confirmed with autocorrelation plots after a
detailed in Additional File 1. 150,000-simulation burn-in phase. Posterior means were based on
an additional 400,000 simulation phase. The model assumed fixed
2.2. Outcome measures effects as the network largely consisted of single-study connec-
tions. A non-informative vague prior was used for means and a
Annualised relapse rate (ARR) and confirmed disability pro- uniform prior on the standard deviation parameter was assumed
gression were selected as the most commonly reported clinical for the baseline effects (Gelman, 2006). For all pairwise compar-
outcomes in RRMS trials. ARR is defined as the mean number of isons the model estimates relative hazard rates (HR) of disability
confirmed relapses per patient adjusted for the duration of follow- progression (assuming progression-free survival follows an ex-
up. Definitions of relapse and disability progression were as per ponential distribution) and relative ARR (assuming a poisson dis-
the individual trial(s), and are detailed in Additional File 2. The tribution for the number of relapses within one study arm). The
definition of ‘relapse’ was subject to slight variation across trials probabilistic analysis does not rely on statistical significance; ra-
but it was commonly defined as new or worsening symptoms that ther comparative effectiveness is based on probabilities of being
last 24 h, occurring in the absence of fever or infection (Schu- among the best treatments for each outcome. The Surface under
macher et al., 1965). Definitions of disability progression varied the cumulative ranking curve (SUCRA) is used to provide a hier-
between trials, but it was commonly defined as at least 1 point archy of treatments for each outcome, accounting for both the
increase on the Expanded Disability Status Scale (EDSS, an am- location and the variance of all relative treatment effects (Salanti
bulation-centred scale from 0 to 10), or a 0.5 point increase if the et al., 2011). Values are expressed as a percentage and show the
E. Fogarty et al. / Multiple Sclerosis and Related Disorders 9 (2016) 23–30 25

cumulative probability of a treatment being among the top r Gold et al., 2012; Fox et al., 2012; Comi et al., 2001; Lublin et al.,
treatments. The larger the SUCRA value the better the treatment 2013; Coles et al., 2008, 2012; Vollmer et al., 2014; Polman et al.,
(Salanti et al., 2011). A ranking plot may be used to illustrate the 2006; Khan et al., 2013). Trials were published between 1993 and
treatments’ hierarchy and the differences in SUCRA values (Chai- 2014, were of 1.75 years mean duration and comprised a total of
mani et al., 2013). Potential sources of treatment-effect modifica- 17,040 patients. The evidence network is illustrated in Fig. 2. ARR
tion from baseline population differences were investigated by outcomes were obtained from all 28 trials, while data on disability
extending the model to a meta-regression including age, gender, progression confirmed after three months and six months were
baseline EDSS, duration of disease, number of relapses in the available from 16 trials. Overall the included studies were broadly
previous two years, and proportion of patients previously treated comparable in terms of mean age (31.8–40.4 years), percentage
with DMT. Treatment effects were estimated at the study-mean female (63–79%), and mean baseline EDSS score (1.9–3.2). Greater
covariate level. By modelling relative effect differences, the model variation was observed in the mean number of relapses in the
accounts for random variation in baseline risks. However, to ex- previous two years (1.7–3.5), mean duration of disease prior to
plain a potential systematic impact of baseline risk on response, recruitment (1.2–10.5 years), and in the proportion of patients who
baseline risk i.e. outcome in the placebo arm, was included had received prior treatment with a DMT (0–100%) (see Additional
alongside other potential confounders as a covariate. Only trials file 3 for full details of the baseline characteristics of the study po-
which include a placebo arm contribute to the estimation of this pulations). The overall risk of bias within included studies was
regression parameter. Inconsistency between direct and indirect judged to be low in 14 studies (50%), medium in one study (4%)
evidence for each mean treatment effect was examined by com- and high in 13 studies (46%). High risk of bias was predominantly
paring the model to a model relaxing the consistency assumption. due to the single-blind nature of many trials. All but one study
The Deviance Information Criterion was used to assess model fit. employed outcome-assessor blinding (Durelli et al., 2002) but
participants were not blinded to treatment allocation in a further
12 RCTs (Vermersch et al., 2013; Mikol et al., 2008; Kappos et al.,
3. Results 2011; Durelli et al., 2002; Panitch et al., 2002; Etemadifar et al.,
2006; Fox et al., 2012; Coles et al., 2008, 2012; Cohen et al., 2012a;
3.1. Evidence base Vollmer et al., 2014; O’Connor et al., 2009). Incomplete outcome
data due to loss to follow-up or imbalance in discontinuations
The systematic search identified 6086 potentially relevant across treatment groups was identified in three studies, (Jacobs
publications, of which 28 trials met the inclusion criteria (Fig. 1) et al., 1996; Coles et al., 2008, 2012) with high attrition bias
(Cohen et al., 2010, 2012a; Confavreux et al., 2014; Vermersch identified in one study (Jacobs et al., 1996) (see Additional file 4 for
et al., 2013; O’Connor et al., 2009, 2011; Saida et al., 2012; Mikol full details of quality assessment).
et al., 2008; PRISMS (Prevention of Relapses and Disability by In-
terferon beta-1a Subcutaneously in Multiple Sclerosis) Study 3.2. Network meta-analysis
Group, 1998; Jacobs et al., 1996; Kappos et al., 2010, 2011; Johnson
et al., 1995; Durelli et al., 2002; Duquette et al., 1993; Calabresi Fig. 3a–c presents the network meta-analysis results for each
et al., 2014a, 2014b; Panitch et al., 2002; Etemadifar et al., 2006; DMT versus placebo for the three outcomes analysed. Each of the

Fig. 1. PRISMA flowchart – Identification and selection of randomised controlled trials of disease-modifying therapies in patients with relapsing remitting multiple sclerosis.
26 E. Fogarty et al. / Multiple Sclerosis and Related Disorders 9 (2016) 23–30

Fig. 2. Network diagram of trials – Each node in the network represents a disease-modifying therapy. Links between nodes represent pairwise treatment comparisons
extracted from randomised controlled trials. Nodes are weighted according to the number of studies included for respective therapies. IFN β ¼interferon beta, IM -
¼ intramuscular, SC ¼ subcutaneous.

DMTs reduced ARR versus placebo. The magnitude of ARR reduc- previously treated with DMT), and one covariate represented
tion varied between 15–36% for all IFN β products, glatiramer baseline risk. The different meta-regression analyses suggested
acetate and teriflunomide, and from 50 to 69% for alemtuzumab, negligible covariate effects from each of the included covariates
dimethyl fumarate, fingolimod and natalizumab. The risk of dis- (See additional file 6). Adjustment of the models for baseline risk
ability progression confirmed after three months was reduced by (based on trial-specific placebo arms in each trial) similarly had
19–28% with IFN β products, glatiramer acetate, fingolimod and negligible impact on the results and the overall ranking of thera-
teriflunomide, by 38–45% for pegylated IFN β, dimethyl fumarate pies (See additional file 7).
and natalizumab and by 68% with alemtuzumab. Superiority over DIC statistics were lower for the consistency model than the
placebo was less certain for IFN β  1a 30 mcg, IFN β  1a 22 mcg, inconsistency model for all outcomes indicating a better fit to the
IFN β  1b 250 mcg and glatiramer acetate 20 mg compared with data and no evidence of inconsistency between the direct and
other therapies. Results for disability progression confirmed after indirect data (See Additional File 5).
six months varied slightly from disability progression confirmed
after three months for each DMT, with the exception of IFN β  1b
250 mcg which was much more efficacious when the outcome was 4. Discussion
defined by a six-month confirmation interval (HR 0.31, 95% CI
0.15–0.62 versus HR 0.83, 95% CI 0.62–1.12). A cumulative ranking The last decade has seen major breakthroughs in the devel-
analysis (Fig. 3d–f) revealed that alemtuzumab and natalizumab opment of new therapeutic strategies for RRMS. However, the
had the highest SUCRA scores for ARR and disability progression extent to which they present efficacy advantages compared to
confirmed after three months, while IFN β 1a 30 mcg ranked established treatments has been unclear, as many trials are pla-
lowest and second-lowest among active treatments for these cebo-controlled, or don’t include all comparators of interest. The
outcomes (SUCRA scores of 9% and 32% respectively). Ranking of Bayesian network meta-analysis framework allows the estimation
treatments was affected by the definition of disability progression of relative treatment effects through the combination of placebo-
largely due to the conflicting results of IFN β  1b 250 mcg, ranking controlled and direct comparative studies while preserving with-
as the most efficacious treatment for disability progression con- in-trial randomised treatment comparisons (Lunn et al., 2000).
firmed after six months (92%) and as the least efficacious for dis- The objective of this study was to compare the efficacy of in-
ability progression confirmed after three months (30%). Alemtu- dividual DMTs for patients with RRMS in terms of the most
zumab and natalizumab both scored relatively highly for disability commonly used clinical trial outcomes, relapse and disability
progression confirmed after six months. Notable variation in progression. Both relapses and progression are the clinical hall-
ranking across outcomes was observed for fingolimod (81% for marks of the MS disease process. However evidence from natural
ARR, 39–46% for the disability progression outcomes). history studies has shown dissociation between relapses and dis-
ability progression pathologies (Confavreux and Vukusic, 2006b).
3.3. Covariate analysis This dissociation has been borne out in individual clinical trials
and in this network meta-analysis where clear reductions in ARR
Meta-regression models including seven study-level covariates are accompanied by more uncertain changes in disability pro-
were used to evaluate the impact of potential treatment effect gression. For many patients relapses are the initial defining feature
modifiers. Six covariates represented population baseline char- of their disease. Relapses can have a significant physical, psycho-
acteristics (age, gender, baseline EDSS, duration of disease, number logical and social impact on patients, and place a substantial cost
of relapses in the previous two years, and proportion of patients burden on patients, their families and healthcare systems (O’Brien
E. Fogarty et al. / Multiple Sclerosis and Related Disorders 9 (2016) 23–30 27

Fig. 3. a-c. Forest plots of treatments versus placebo for a) Annualised relapse rate, b) Disability progression confirmed at three months c) Disability progression confirmed at
six months. Relative treatment effects (rate ratios for annualised relapse rate, hazard ratios for disability progression) are represented by coloured nodes, and corresponding
95% credible intervals are represented by solid lines. Bars to the left of the central vertical line of no difference indicate superiority of the treatment over placebo. Hollow
nodes represent pairwise comparisons where the 95% credible interval spans the central vertical line of no difference. d-f. Network ranking plots for d) Annualised relapse
rate, e) Disability progression confirmed at three months c) Disability progression confirmed at six months. Treatments are ranked according to the surface under the
cumulative ranking curve (SUCRA). SUCRA values provide the hierarchy for the treatments and placebo, and show the cumulative probability (expressed as a percentage) of a
treatment being among the best options. The y-axis shows the possible ranks from r ¼ 1 up to r ¼ 11 and the x-axis shows the cumulative probabilities that the corresponding
treatment is among the top r treatments. The larger the SUCRA value the better the treatment. IFN β ¼interferon beta; IM ¼ intramuscular; SC ¼ subcutaneous; RR ¼rate ratio;
HR ¼hazard ratio; CI ¼credible interval.
28 E. Fogarty et al. / Multiple Sclerosis and Related Disorders 9 (2016) 23–30

et al., 2003; Kalb, 2007; Halper, 2007). However, it is the accu- highlighted, and it is not clear whether either of the definitions of
mulation of disability which has the greatest long-term clinical, progression assessed in this analysis truly represents irreversible
social and economic impact on patients and society (Fogarty et al., sustained disability progression and reliably reflect long-term
2013, 2014). changes in disease progression (Cohen et al., 2012b). Despite its
Generally, DMTs were superior to placebo in reducing MS re- limitations, at the moment the EDSS remains the only validated
lapse rates and disability progression. However the magnitude of outcome measurement to determine disability (determined by the
the reduction and the uncertainty associated with treatment ef- European Medicines Agency) and its use is recommended to
fects varied between DMTs, and between the different outcomes continue in the future alongside improved measures of disability
included in the analysis, leading to variation in the relative ranking progression to facilitate comparison with other studies. Research is
of treatments. The monoclonal antibody therapies alemtuzumab ongoing into novel approaches to demonstrating efficacy. RCTs are
and natalizumab were generally among the highest ranked treat- increasingly incorporating adjunctive imaging outcomes and
ments for all outcomes. Among the oral therapies, fingolimod and composite measures such as disease-activity-free-status (Bevan
dimethyl fumarate ranked higher than other therapies for ARR, and Cree, 2014). These and other novel outcomes will provide
while there was little difference between teriflunomide and other additional evidence on the comparative efficacy of treatments but
first-line DMTs for this outcome. Dimethyl fumarate, pegylated IFN are not developed sufficiently to serve as substitutes for the out-
β and IFN β 44 mcg occupied higher rankings than other DMTs for comes investigated in this analysis.
disability progression confirmed after three months and there was The long-term relevance of treatment effects estimated on the
little to distinguish between the rankings of other treatments. basis of short-term trials (6–33 months follow-up) may be limited.
European guidelines on the clinical investigation of medicinal RRMS may require treatment with DMT for many years, but there
products for the treatment of MS recommend a six month con- is little evidence on the long-term efficacy of these agents. The
firmation interval for an accurate and reliable definition of sus- development of neutralising antibodies can complicate prolonged
tained worsening as studies have shown that improvement in IFN β therapy (Giovannoni et al., 2002). Results from some long-
EDSS scores after relapse can continue beyond three months term open-label extension studies suggest maintained efficacy of
(European Medicines Agency (EMA) Committee for Medicinal DMTs on relapse rate over extended periods of use, however the
Products for Human Use (CHMP), 2006; Cohen et al., 2012b). De- lack of a comparator arm limits the conclusions on clinical efficacy
spite this, disability progression is often defined in RCTs on the which can be drawn from these studies (Khatri et al., 2011; Con-
basis of a three-month confirmation interval rather than six favreux et al., 2012; Kappos et al., 2012a, 2012b; Gold et al., 2014;
months. For this reason, disability progression defined on the basis Ford et al., 2010). Conflicting results have been reported for the
of both three and six month confirmation intervals were included long-term efficacy of first line DMTs on reducing disability pro-
as two separate outcomes in the analysis. With the exception of gression (Goodin et al., 2011; Shirani et al., 2012).
IFN β  1b 250 mcg SC, point estimates for the efficacy of DMTs The study has a number of limitations. As with traditional
versus placebo in reducing disability progression were similar pairwise meta-analysis, the model requires that individual studies
between both definitions with credible intervals for the two out- are sufficiently similar, so as to generate exchangeable treatment
comes overlapping in all cases. A significant improvement in ef- effects. The trials included in the analysis were conducted over a
ficacy was observed for IFN β 1b 250 mcg SC when disability period of over 25 years. Slight variation in outcome definitions
progression was defined on the basis of a six-month confirmation between studies and changes in patient populations were evident
interval, ranking highest for this outcome compared with being in duration of disease, proportion of trial populations who had
among the lowest ranked treatments on the basis of a three- received prior DMT, and also in the downward trend in relapse
month confirmation interval. Just one study, the INCOMIN study, rates over time. We explored potential sources of heterogeneity
contributed evidence for the efficacy of IFN β  1b 250 mcg SC on from several known covariates to adjust for baseline imbalance in
disability progression confirmed after six months, finding it to be underlying risk across studies. No significant association between
superior to IFN β  1a 30 mcg IM for this outcome (Durelli et al., efficacy and baseline risk or other covariates were identified. Al-
2002). This is in contrast to two studies, one placebo controlled though no significant confounding of treatment effect was iden-
and one versus glatiramer acetate which did not show significant tified, meta-regression based on aggregate study-level data is
improvements with IFN β  1b 250 mcg SC in disability progres- prone to ecological bias and the risk of unknown imbalances in
sion confirmed after three months (Duquette et al., 1993; O’Connor treatment effect modifiers cannot be excluded (Cooper et al.,
et al., 2009). It has been suggested that differences in baseline 2009). Consistency between the direct and indirect evidence is
disease characteristics indicate that patients treated with IFN dependent on the distribution of effect modifiers and where both
β  1a 30 mcg IM in the INCOMIN study may have had a poorer direct and indirect evidence was available no evidence of incon-
prognosis than IFN β  1b 250 mcg SC treated patients (Vartanian, sistency was identified. There was variation in study quality
2003). Bias may also have been introduced to the INCOMIN study among the included trials. In particular, concerns regarding the
by the lack of blinding as this was the only study included in the open-label design may affect our level of confidence in the esti-
analysis which was not assessor-blinded. mates of effect from some trials, as the potential for bias cannot be
The relevance of the clinical outcomes investigated in this excluded. In this study we did not investigate the relative ac-
study in the setting of a chronic, heterogeneous condition which ceptability or tolerability of the various treatment strategies.
evolves over time, is uncertain. While the mechanisms that un- Consideration of safety outcomes alongside treatment efficacy is
derlie relapses and gradual progression may be distinct, they may central to decision-making in healthcare and has been addressed
sometimes present concurrently (Cohen et al., 2012b). Improve- elsewhere (Saidha et al., 2012; Smith et al., 2010). Cost-effective-
ment in EDSS scores after relapses may continue beyond three to ness, likewise, was not considered. This is increasingly a critical
six months and variable recovery from relapses may lead to the determinant of reimbursement in many countries, due to the high
accrual of relapse-related disability. Relapses decrease over time acquisition cost of many of these treatments.
and disappear in many patients with the onset of secondary pro-
gression (Tremlett et al., 2008). The EDSS is an ordinal rather than 5. Conclusion
linear measure and differences between each level (e.g. a one
point change between 1.5 and 2.5, and between 3.5 and 4.5 may The network meta-analysis approach is not a substitute for
not comparable. Other inadequacies of the EDSS scale have been large, well-designed RCTs comparing the comparators of interest.
E. Fogarty et al. / Multiple Sclerosis and Related Disorders 9 (2016) 23–30 29

However, given the current limitations in the evidence base, this Calabresi, P.A., Radue, E.-W., Goodin, D., Jeffery, D., Rammohan, K.W., Reder, A.T.,
study provides an alternative framework within which compara- et al., 2014a. Safety and efficacy of fingolimod in patients with relapsing-re-
mitting multiple sclerosis (FREEDOMS II): a double-blind, randomised, placebo-
tive effectiveness can be estimated. This analysis integrated evi- controlled, phase 3 trial. Lancet Neurol. 13 (6), 545–556.
dence from a comprehensive network of trials of DMT in RRMS, Calabresi, P.A., Kieseier, B.C., Arnold, D.L., Balcer, L.J., Boyko, A., Pelletier, J., et al.,
using both direct and indirect data to simultaneously evaluate 2014b. Pegylated interferon beta-1a for relapsing-remitting multiple sclerosis
(ADVANCE): a randomised, phase 3, double-blind study. Lancet Neurol. 13 (7),
treatment effects of interventions not previously studied in head- 657–665.
to-head trials. Treatment rankings have been presented for sepa- Chaimani, A., Higgins, J.P.T., Mavridis, D., Spyridonos, P., Salanti, G., 2013. Graphical
rate outcomes, based on cumulative probabilities of being among tools for network meta-analysis in STATA. PLoS One 8 (10), e76654.
Cohen, J.A., Coles, A.J., Arnold, D.L., Confavreux, C., Fox, E.J., Hartung, H.P., et al.,
the most effective treatments. There is little difference (indicating
2012a. Alemtuzumab versus interferon beta 1a as first-line treatment for pa-
similarity in treatment effects) between these probabilities for tients with relapsing-remitting multiple sclerosis: a randomised controlled
many treatments, with newer agents natalizumab and alemtuzu- phase 3 trial. Lancet 380 (9856), 1819–1828.
Cohen, J.A., Reingold, S.C., Polman, C.H., Wolinsky, J.S., 2012b. Disability outcome
mab demonstrating consistently high ranking across outcomes.
measures in multiple sclerosis clinical trials: Current status and future pro-
Health policy and resource allocation decision-making frequently spects. Lancet Neurol. 11 (5), 467–476.
requires quantification of comparative efficacy. Evidence synthesis Cohen, J.A., Barkhof, F., Comi, G., Hartung, H.P., Khatri, B.O., Montalban, X., et al.,
within this network meta-analysis framework is a powerful ap- 2010. Oral fingolimod or intramuscular interferon for relapsing multiple
sclerosis. New Engl. J. Med. 362 (5), 402–415.
proach to dealing with complex evidence structures to facilitate Coles, A., 2009. Multiple sclerosis: the bare essentials. Pract. Neurol. 9 (2), 118–126.
coherent, evidence-based decision-making. Coles, A.J., Compston, D.A.S., Selmaj, K.W., Lake, S.L., Moran, S., Margolin, D.H., et al.,
2008. Alemtuzumab vs. interferon beta-1a in early multiple sclerosis. New Engl.
J. Med. 359 (17), 1786–1801.
Coles, A.J., Twyman, C.L., Arnold, D.L., Cohen, J.A., Confavreux, C., Fox, E.J., et al.,
Author contributions 2012. Alemtuzumab for patients with relapsing multiple sclerosis after disease-
modifying therapy: a randomised controlled phase 3 trial. Lancet 380 (9856),
1829–1839.
EF conceived and designed the study, identified and acquired Comi, G., Filippi, M., Wolinsky, J.S., 2001. European/Canadian multicenter, double-
trials, extracted data, contributed to data analysis, interpreted the blind, randomized, placebo-controlled study of the effects of glatiramer acetate
results drafted the manuscript and is guarantor. SS identified trials, on magnetic resonance imaging–measured disease activity and burden in pa-
tients with relapsing multiple sclerosis. European/Canadian Glatiramer Acetate
extracted data, performed statistical analysis and critically revised
Study Group. Ann. Neurol. 49 (3), 290–297.
the manuscript for important intellectual content. CW provided Confavreux, C., Vukusic, S., 2006a. Natural history of multiple sclerosis: a unifying
statistical advice and critically revised the manuscript for im- concept. Brain 129 (3), 606–616.
portant intellectual content. NT and MB critically revised the Confavreux, C., Vukusic, S., 2006b. Accumulation of irreversible disability in mul-
tiple sclerosis: from epidemiology to treatment. Clin. Neurol. Neurosurg. 108
manuscript for important intellectual content. All authors ap- (3), 327–332.
proved the final version of the manuscript. Confavreux, C., Li, D.K., Freedman, M.S., Truffinet, P., Benzerdjeb, H., Wang, D., et al.,
2012. Long-term follow-up of a phase 2 study of oral teriflunomide in relapsing
multiple sclerosis: safety and efficacy results up to 8.5 years. Mult. Scler. 18 (9),
1278–1289.
Conflict of interest statement Confavreux, C., O’Connor, P., Comi, G., Freedman, M.S., Miller, A.E., Olsson, T.P., et al.,
2014. Oral teriflunomide for patients with relapsing multiple sclerosis (TOW-
ER): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet
EF, SS, CW and MB report no conflict of interest. Prof. Tubridy Neurol. 13 (3), 247–256.
has received unrestricted educational and research grants on be- Cooper, N.J., Sutton, A.J., Morris, D., Ades, A.E., Welton, N.J., 2009. Addressing be-
half of the Department Of Neurology at St. Vincent’s University tween-study heterogeneity and inconsistency in mixed treatment compar-
isons: application to stroke prevention treatments in individuals with non-
Hospital from Bayer Schering, Biogen Idec and Novartis.
rheumatic atrial fibrillation. Stat. Med. 28 (14), 1861–1881.
Duquette, P., Girard, M., Despault, L., DuBois, R., Knobler, R.L., Lublin, F.D., et al.,
1993. Interferon beta-1b is effective in relapsing-remitting multiple sclerosis. I.
Clinical results of a multicenter, randomized, double-blind, placebo- controlled
Ethical approval
trial. Neurology 43 (4 I), 655–661.
Durelli, L., Verdun, E., Barbero, P., Bergui, M., Versino, E., Ghezzi, A., et al., 2002.
Not required for this study. 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 (9316), 1453–1460.
Etemadifar, M., Janghorbani, M., Shaygannejad, V., 2006. Comparison of Betaferon,
Funding Avonex, and Rebif in treatment of relapsing-remitting multiple sclerosis. Acta
Neurol. Scand. 113 (5), 283–287.
European Medicines Agency (EMA) Committee for Medicinal Products for Human
No funding was received for this study. Use (CHMP). Guideline on Clinical Investigation of Medicinal Products for the
Treatment of) Multiple Sclerosis. London, 16 November 2006 Doc. Ref. CPMP/
EWP/561/98 Rev. 1.
Fogarty, E., Walsh, C., McGuigan, C., Tubridy, N., Barry, M., 2014. Direct and indirect
Data sharing economic consequences of multiple sclerosis in Ireland. Appl. Health Econ.
Health Policy, 1–11.
Statistical code is available from the corresponding author. Fogarty, E., Walsh, C., Adams, R., McGuigan, C., Barry, M., Tubridy, N., 2013. Relating
health-related quality of Life to disability progression in multiple sclerosis,
using the 5-level EQ-5D. Mult. Scler. 19 (9), 1190–1196.
Ford, C., Goodman, A.D., Johnson, K., Kachuck, N., Lindsey, J.W., Lisak, R., et al., 2010.
Appendix A. Supplementary material 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. Mult. Scler. 16 (3), 342–350.
Supplementary data associated with this article can be found in Fox, R.J., Miller, D.H., Phillips, J.T., Hutchinson, M., Havrdova, E., Kita, M., et al., 2012.
the online version at http://dx.doi.org/10.1016/j.msard.2016.06. Placebo-controlled phase 3 study of oral BG-12 or glatiramer in multiple
001. sclerosis. New Engl. J. Med. 367 (12), 1087–1097.
Gelman, A., 2006. Prior distributions for variance parameters in hierarchical
models. Bayesian Anal. 1 (3) 151–33.
Giovannoni, G., Munschauer, F.E., Deisenhammer, F., 2002. Neutralising antibodies
References to interferon beta during the treatment of multiple sclerosis. J. Neurol., Neu-
rosurg. Psychiatry 73 (5), 465–469.
Gold, R., Kappos, L., Arnold, D.L., Bar-Or, A., Giovannoni, G., Selmaj, K., et al., 2012.
Bevan, C.J., Cree, B.C., 2014. Disease activity free status: a new end point for a new Placebo-controlled phase 3 study of oral BG-12 for relapsing multiple sclerosis.
era in multiple sclerosis clinical research? JAMA Neurol. 71 (3), 269–270. New Engl. J. Med. 367 (12), 1098–1107.
30 E. Fogarty et al. / Multiple Sclerosis and Related Disorders 9 (2016) 23–30

Gold, R., Phillips, J.T., Bar-Or, A., Hutchinson, M., Kappos, L., Rana, J., et al., 2014. 3- O’Brien J.A., Ward, A.J., Patrick, A.R., Caro, J., 2003. Cost of managing an episode of
year follow-up of oral BG-12 (dimethyl fumarate) for relapsing-remitting relapse in multiple sclerosis in the United States. BMC Health Services Research
multiple sclerosis (RRM)S: Integrated clinical outcomes from the define, con- 3(O’Brien J.A., jobrien@caroresearch.com; Ward A.J., alexward@caroresearch.
firm, and endorse studies. Mult. Scler. 20 (7), 932. com; Patrick A.R., apatrick@hsph.harvard.edu; Caro J., jcaro@caroresearch.com).
Goodin, D.S., Jones, J., Li, D., Traboulsee, A., Reder, A.T., Beckmann, K., et al., 2011. Caro Research Institute, Concord, MA 01742, United States, pp. 1–12.
Establishing long-term efficacy in chronic disease: use of recursive partitioning O’Connor, P., Filippi, M., Arnason, B., Comi, G., Cook, S., Goodin, D., et al., 2009. 250
and propensity score adjustment to estimate outcome in MS. PLoS One 6 (11), (mu)g or 500 (mu)g interferon beta-1b versus 20 mg glatiramer acetate in re-
e22444. lapsing-remitting multiple sclerosis: a prospective, randomised, multicentre
Halper, J., 2007. The psychosocial effect of multiple sclerosis: the impact of relapses. study. Lancet Neurol. 8 (10), 889–897.
J. Neurol. Sci. 256 (SUPPL. 1), S34–S38. O’Connor, P., Wolinsky, J.S., Confavreux, C., Comi, G., Kappos, L., Olsson, T.P., et al.,
Higgins, J. Green S, (editors). Cochrane Handbook for Systematic Reviews of Inter- 2011. Randomized trial of oral teriflunomide for relapsing multiple sclerosis.
ventions. Version 5.1.0. the Cochrane Collaboration,. 2011. New Engl. J. Med. 365 (14), 1293–1303.
Jacobs, L.D., Cookfair, D.L., Rudick, R.A., Herndon, R.M., Richert, J.R., Salazar, A.M., Panitch, H., Goodin, D.S., Francis, G., Chang, P., Coyle, P.K., O’Connor, P., et al., 2002.
et al., 1996. Intramuscular interferon beta  1a for disease progression in re- Randomized, comparative study of interferon (beta)-1a treatment regimens in
lapsing multiple sclerosis. Ann. Neurol. 39 (3), 285–294. MS: the evidence trial. Neurology 59 (10), 1496–1506.
Johnson, K.P., Brooks, B.R., Cohen, J.A., Ford, C.C., Goldstein, J., Lisak, R.P., et al., 1995. Polman, C.H., O’Connor, P.W., Havrdova, E., Hutchinson, M., Kappos, L., Miller, D.H.,
Copolymer 1 reduces relapse rate and improves disability in relapsing- remit- et al., 2006. A randomized, placebo-controlled trial of natalizumab for relapsing
ting multiple sclerosis: results of a phase III multicenter, double- blind, place- multiple sclerosis. New Engl. J. Med. 354 (9), 899–910.
bo-controlled trial. Neurology 45 (7), 1268–1276. PRISMS (Prevention of Relapses and Disability by Interferon beta-1a Sub-
Kalb, R., 2007. The emotional and psychological impact of multiple sclerosis re- cutaneously in Multiple Sclerosis) Study Group, 1998. Randomised double-
lapses. J. Neurol. Sci. 256 (SUPPL. 1), S29–S33. blind placebo-controlled study of interferon beta-1a in relapsing/remitting
Kappos, L., Radue, E.W., O’Connor, P., Polman, C., Hohlfeld, R., Calabresi, P., et al., multiple sclerosis. Lancet 352 (9139), 1498–1504.
2010. A placebo-controlled trial of oral fingolimod in relapsing multiple
Saida, T., Kikuchi, S., Itoyama, Y., Hao, Q., Kurosawa, T., Nagato, K., et al., 2012. A
sclerosis. New Engl. J. Med. 362 (5), 387–401.
randomized, controlled trial of fingolimod (FTY720) in Japanese patients with
Kappos, L., Li, D., Calabresi, P.A., O’Connor, P., Bar-Or, A., Barkhof, F., et al., 2011.
multiple sclerosis. Mult. Scler. 18 (9), 1269–1277.
Ocrelizumab in relapsing-remitting multiple sclerosis: a phase 2, randomised,
Saidha, S., Eckstein, C., Calabresi, P.A., 2012. New and emerging disease modifying
placebo-controlled, multicentre trial. Lancet 378 (9805), 1779–1787.
therapies for multiple sclerosis. Ann. N. Y. Acad. Sci. 1247 (1), 117–137.
Kappos, L., Radue, E.W., O’Connor, P., Polman, C., Hohlfeld, R., Calabresi, P., et al.,
Salanti, G., Ades, A.E., Ioannidis, J.P.A., 2011. Graphical methods and numerical
2012a. Long-term efficacy and safety of fingolimod (FTY720) in relapsing-re-
summaries for presenting results from multiple-treatment meta-analysis: an
mitting multiple sclerosis (RRMS): results from the extension of the phase III
overview and tutorial. J. Clin. Epidemiol. 64 (2), 163–171.
FREEDOMS study. Neurology 78, 1.
Schumacher, G.A., Beebe, G.W., Kibler, R.F., Kurland, L.T., Kurtzke, J.F., Fea, Mc.
Kappos, L., Belachew, S., Butzkueven, H., Pellegrini, F., Trojano, M., Wiendl, H., et al.,
Dowell, 1965. Problems of experimental trials of therapy in multiple sclerosis:
2012b. Long-term safety and efficacy and association between baseline treat-
report by the panel on the evaluation of experimental trials of therapy in
ment history and postbaseline relapses in multiple sclerosis patients treated
with natalizumab in the tysabri(registered trademark) observational program multiple sclerosis. Ann. N. Y. Acad. Sci. 122, 552–568.
(top). Neurology 78 (1). Shirani, A., Zhao, Y., Karim, M.E., Evans, C., Kingwell, E., Van Der Kop, M.L., et al.,
Khan, O., Rieckmann, P., Boyko, A., Selmaj, K., Zivadinov, R., Group, G.S., 2013. Three 2012. Association between use of interferon beta and progression of disability
times weekly glatiramer acetate in relapsing-remitting multiple sclerosis. Ann. in patients with relapsing-remitting multiple sclerosis. JAMA: J. Am. Med. As-
Neurol. 73 (6), 705–713. soc. 308 (3), 247–256.
Khatri, B., Barkhof, F., Comi, G., Hartung, H.-P., Kappos, L., Montalban, X., et al., 2011. Smith, B., Carson, S., Fu, R., et al. Drug class review: disease-modifying drugs for
Comparison of fingolimod with interferon beta-1a in relapsing-remitting multiple sclerosis: Final Update 1 Report [Internet]. Portland (OR): Oregon
multiple sclerosis: a randomised extension of the TRANSFORMS study. Lancet Health & Science University; 2010 Aug. Available from: 〈http://www.ncbi.nlm.
Neurol. 10 (6), 520–529. nih.gov/books/NBK50570/〉.
Kurtzke, J.F., 1983. Rating neurologic impairment in multiple sclerosis: an expanded Sturtz, S., Ligges, U., Gelman, A., 2005. R2WinBUGS: a package for running Win-
disability status scale (EDSS). Neurology 33 (11), 1444–1452. BUGS from R. J. Stat. Softw. 12 (3), 1–16.
Lublin, F.D., Cofield, S.S., Cutter, G.R., Conwit, R., Narayana, P.A., Nelson, F., et al., Tremlett, H., Zhao, Y., Joseph, J., Devonshire, V., Adams, D., Craig, D., et al., 2008.
2013. Randomized study combining interferon and glatiramer acetate in mul- Relapses in multiple sclerosis are age- and time-dependent. J. Neurol. Neuro-
tiple sclerosis. Ann. Neurol. 73 (3), 327–340. surg. Psychiatry 79 (12), 1368–1374.
Lunn, D.J., Thomas, A., Best, N., Spiegelhalter, D., 2000. WinBUGS – a Bayesian Vartanian, T., 2003. An examination of the results of the EVIDENCE, INCOMIN, and
modelling framework: concepts, structure, and extensibility. Stat. Comput. 10, phase III studies of interferon beta products in the treatment of multiple
325–337. sclerosis. Clin. Ther. 25 (1), 105–118.
Mikol, D.D., Barkhof, F., Chang, P., Coyle, P.K., Jeffery, D.R., Schwid, S.R., et al., 2008. Vermersch, P., Czlonkowska, A., Grimaldi, L.M., Confavreux, C., Comi, G., Kappos, L.,
Comparison of subcutaneous interferon beta-1a with glatiramer acetate in 2013. Teriflunomide versus subcutaneous interferon beta-1a in patients with
patients with relapsing multiple sclerosis (the REbif vs Glatiramer Acetate in relapsing multiple sclerosis: a randomised, controlled phase 3 trial. Mult. Scler.
Relapsing MS Disease [REGAR)D] (stud)y): (a multicentre, randomised, parallel, J.
open-label trial). Lancet Neurol. 7 (10), 903–914. Vollmer, T.L., Sorensen, P.S., Selmaj, K., Zipp, F., Havrdova, E., Cohen, J.A., et al., 2014.
Moher, D., Liberati, A., Tetzlaff, J., Altman, D.G., 2009. Preferred reporting items for A randomized placebo-controlled phase III trial of oral laquinimod for multiple
systematic reviews and meta-analyses: the PRISMA statement. BMJ, 339. sclerosis. J. Neurol. 261 (4), 773–783.

You might also like