Effects of teriflunomide treatment on cognitive performance and brain volume in patients with relapsing multiple sclerosis: Post hoc analysis of the TEMSO core and extension studies

Till Sprenger, Ludwig Kappos, Maria Pia Sormani, Aaron E Miller, Elizabeth M Poole, Steven Cavalier, Jens Wuerfel, Till Sprenger, Ludwig Kappos, Maria Pia Sormani, Aaron E Miller, Elizabeth M Poole, Steven Cavalier, Jens Wuerfel

Abstract

Background: In post hoc analyses of Teriflunomide Multiple Sclerosis Oral study (TEMSO; NCT00134563), teriflunomide 14 mg significantly reduced brain volume loss (BVL) versus placebo in patients with relapsing multiple sclerosis (MS).

Objective: In this post hoc analysis of TEMSO and its long-term extension (NCT00803049), we examined the relationship between teriflunomide's effects on BVL and cognition.

Methods: We analyzed data from 709 patients who received teriflunomide 14 mg in TEMSO or its extension. The change in cognitive performance, assessed using the Paced Auditory Serial Addition Test 3 (PASAT-3), was measured in subgroups stratified by BVL over 2 years (least BVL: ⩽ 0.52%; intermediate BVL: >0.52%-2.18%; most BVL: >2.18%). BVL, MRI lesions, and relapses over 2 years were evaluated as potential mediators of the effect of teriflunomide on cognition.

Results: Teriflunomide 14 mg significantly improved PASAT-3 Z-scores versus placebo through year 2. In the least- and intermediate-BVL groups, significant improvements in PASAT-3 Z-score were demonstrated versus the most-BVL group over 3 years in the extension. According to the mediation analysis, 44% of the teriflunomide effect on cognition was due to effects on BVL at year 2.

Conclusion: Teriflunomide improves cognition largely through its effects on BVL. Accelerated BVL earlier in the disease course may predict cognitive outcomes.

Clinicaltrials.gov identifier: NCT00134563, NCT00803049.

Keywords: TEMSO; Teriflunomide; brain volume loss; cognition; multiple sclerosis.

Conflict of interest statement

Declaration of conflicting interests: The author(s) declared the following potential conflicts of interest with respect to the research, authorship, and/or publication of this article: TS: Grants from EFIC-Grünenthal, Novartis Pharmaceuticals Switzerland, the Swiss MS Society, and the Swiss National Research Foundation. Institution received payments for steering committee/consultation and speaking activities from Actelion, ATI, Biogen Idec, Electrocore, Eli Lilly, Janssen, Merck Serono, Mitsubishi Pharma, Novartis, Roche, Sanofi, and Teva. LK: Institution (University Hospital Basel) has received the following exclusively for research support: steering committee, advisory board, and consultancy fees (Actelion, Bayer HealthCare, Biogen, BMS, Genzyme, Janssen, Merck, Novartis, Roche, Sanofi, Santhera, and TG Therapeutics); speaker fees (Bayer HealthCare, Biogen, Merck, Novartis, Roche, and Sanofi); support of educational activities (Allergan, Bayer HealthCare, Biogen, CSL Behring, Desitin, Genzyme, Merck, Novartis, Pfizer, Roche, Sanofi, Shire, and Teva); license fees for Neurostatus products; and grants (Bayer HealthCare, Biogen, European Union, InnoSwiss, Merck, Novartis, Roche, Swiss MS Society, and Swiss National Research Foundation). MPS: Consulting fees from Biogen, Genzyme, Merck Serono, Novartis, Roche, Synthon, and Teva. AEM: Consulting fees from Accordant Health Services, Adamas, Biogen, Celgene, EMD Serono, Genentech/Roche, Mapi-Pharma, and Novartis; contracted research for Biogen, Genentech, MedDay, Novartis, Mallinckrodt, and Roche. EMP: Employee of Sanofi at the time of the analysis; currently employed by Jazz Pharmaceuticals, Cambridge, MA, USA. SC: Employee of Sanofi at the time of the analysis; currently employed by Steven Cavalier Consulting, LLC. JW: Employee of MIAC AG in Basel, Switzerland; served on scientific advisory boards for Actelion, Bayer, Biogen, Idorsia, Novartis, Roche, Sanofi, and Teva.

Figures

Figure 1.
Figure 1.
TEMSO core and extension study design (a), and flow diagram based on treatment group and the number of patients from each treatment group in each BVL subgroup after stratification (b). BVL: brain volume loss; ITT: intent to treat; PASAT-3: Paced Auditory Serial Addition Test 3; R: randomization; SIENA: structural image evaluation using normalization of atrophy. †ITT population with valid scans at baseline and year 2. ‡Least BVL from core study baseline to week 108: ⩽0.52%. §Intermediate BVL from core study baseline to week 108: >0.52%–2.18%. ¶Most BVL from core study baseline to week 108: >2.18%.
Figure 2.
Figure 2.
Change in PASAT-3 Z-score through the TEMSO core study in placebo and teriflunomide 14 mg patients (a), and through the TEMSO core study and its extension period in pooled teriflunomide 14 mg patients (b). CEL: contrast-enhancing lesion; EDSS: Expanded Disability Status Scale; LS: least squares; PASAT-3: Paced Auditory Serial Addition Test 3; SE: standard error. †Last visit of core study, up to week 96.p-Values were generated using analysis of covariance adjusted for age, baseline Z-score, baseline EDSS strata, baseline CEL status, treatment, and quartile category. Panel A analytic sample is all participants in the randomized population (randomized to placebo or teriflunomide 14 mg) with PASAT scores available at baseline and week 96. Panel B analytic sample is all participants after teriflunomide 14 mg initiation (either at baseline or in the extension) with PASAT scores available at the specific week reported.
Figure 3.
Figure 3.
Change in PASAT-3 Z-score by BVL group through the TEMSO extension period. BVL: brain volume loss; CEL: contrast-enhancing lesion; EDSS: Expanded Disability Status Scale; LS: least squares; PASAT-3: Paced Auditory Serial Addition Test 3; SE: standard error. Least BVL: ⩽0.52%; intermediate BVL: >0.52%–2.18%; most BVL: >2.18%. p-Values were generated using analysis of covariance adjusted for age, baseline Z-score, baseline EDSS strata, baseline CEL status (yes/no), treatment, and quartile category. The analytic sample is all patients (placebo and both teriflunomide groups) with valid MRIs at baseline and week 108 (for categorizing BVL) who have PASAT scores at baseline and the specific week reported.
Figure 4.
Figure 4.
Percentage of relative contributions of each surrogate individually (a) and combined (b). BVL: brain volume loss; CEL: contrast-enhancing lesion; PTE: proportion of treatment effect; T2w: T2 weighted. The analytic sample is patients with valid MRIs at baseline and week 108.

References

    1. Vollmer T, Signorovitch J, Huynh L, et al.. The natural history of brain volume loss among patients with multiple sclerosis: A systematic literature review and meta-analysis. J Neurol Sci 2015; 357: 8–18.
    1. Coles AJ, Cox A, Le Page E, et al.. The window of therapeutic opportunity in multiple sclerosis: Evidence from monoclonal antibody therapy. J Neurol 2006; 253(1): 98–108.
    1. Popescu V, Agosta F, Hulst HE, et al.. Brain atrophy and lesion load predict long term disability in multiple sclerosis. J Neurol Neurosurg Psych 2013; 84(10): 1082–1091.
    1. Sormani MP, Arnold DL, De Stefano N. Treatment effect on brain atrophy correlates with treatment effect on disability in multiple sclerosis. Ann Neurol 2014; 75: 43–49.
    1. Vollmer T, Huynh L, Kelley C, et al.. Relationship between brain volume loss and cognitive outcomes among patients with multiple sclerosis: A systematic literature review. Neurol Sci 2016; 37(2): 165–179.
    1. Calabrese M, Agosta F, Rinaldi F, et al.. Cortical lesions and atrophy associated with cognitive impairment in relapsing-remitting multiple sclerosis. Arch Neurol 2009; 66(9): 1144–1150.
    1. Furby J, Hayton T, Anderson V, et al.. Magnetic resonance imaging measures of brain and spinal cord atrophy correlate with clinical impairment in secondary progressive multiple sclerosis. Mult Scler 2008; 14(8): 1068–1075.
    1. Corallo F, Rifici C, Bonanno L, et al.. The role of teriflunamide in multiple sclerosis patient: An observational study. Psychol Health Med 2021; 1-8.
    1. Radue EW, Sprenger T, Gaetano L, et al.. Teriflunomide slows BVL in relapsing MS: A reanalysis of the TEMSO MRI data set using SIENA. Neurol Neuroimmunol Neuroinflamm 2017; 4(5): e390.
    1. Sprenger T, Kappos L, Radue EW, et al.. Association of brain volume loss and long-term disability outcomes in patients with multiple sclerosis treated with teriflunomide. Mult Scler 2020; 26(10): 1207–1216.
    1. O’Connor P, Wolinsky JS, Confavreux C, et al.. Randomized trial of oral teriflunomide for relapsing multiple sclerosis. N Engl J Med 2011; 365: 1293–1303.
    1. O’Connor P, Comi G, Freedman MS, et al.. Long-term safety and efficacy of teriflunomide: Nine-year follow-up of the randomized TEMSO study. Neurology 2016; 86: 920–930.
    1. Lazeron RH, Boringa JB, Schouten M, et al.. Brain atrophy and lesion load as explaining parameters for cognitive impairment in multiple sclerosis. Mult Scler 2005; 11(5): 524–531.
    1. Cohen JA, Cutter GR, Fischer JS, et al.. Use of the multiple sclerosis functional composite as an outcome measure in a phase 3 clinical trial. Arch Neurol 2001; 58(6): 961–967.
    1. Forn C, Belenguer A, Parcet-Ibars MA, et al.. Information-processing speed is the primary deficit underlying the poor performance of multiple sclerosis patients in the Paced Auditory Serial Addition Test (PASAT). J Clin Exp Neuropsychol 2008; 30(7): 789–796.
    1. Kappos L, Radue EW, O’Connor P, et al.. A placebo-controlled trial of oral fingolimod in relapsing multiple sclerosis. N Engl J Med 2010; 362(5): 387–401.
    1. 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(6): 545–556.
    1. Arnold DL, Gold R, Kappos L, et al.. Effects of delayed-release dimethyl fumarate on MRI measures in the Phase 3 DEFINE study. J Neurol 2014; 261(9): 1794–1802.
    1. 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: 1098–1107.
    1. Fischer J, Jak A, Kniker J, et al.. Multiple sclerosis functional composite (MSFC) administration and scoring manual. New York: National Multiple Sclerosis Society.
    1. Healy BC, Engler D, Glanz B, et al.. Assessment of definitions of sustained disease progression in relapsing-remitting multiple sclerosis. Mult Scler Int 2013; 2013: 189624.
    1. Prentice RL. Surrogate endpoints in clinical trials: Definition and operational criteria. Stat Med 1989; 8(4): 431–440.
    1. Lechner-Scott J, Sprenger T, Sormani MP, et al.. Evaluation of long-term treatment of teriflunomide on cognitive outcomes and association with brain volume change: Data from the TEMSO study and its extension. In: 11th congress of the Pan-Asian Committee for treatment and research in multiple sclerosis, 1-3 November 2018, Sydney, NSW, Australia, Multiple Sclerosis Journal 2019; 25(3): 432–492.
    1. Jasperse B, Vrenken H, Sanz-Arigita E, et al.. Regional brain atrophy development is related to specific aspects of clinical dysfunction in multiple sclerosis. Neuroimage 2007; 38(3): 529–537.
    1. Di Filippo M, Portaccio E, Mancini A, et al.. Multiple sclerosis and cognition: Synaptic failure and network dysfunction. Nat Rev Neurosci 2018; 19(10): 599–609.
    1. Rogers JM, Fox AM. Event-related potential practice effects on the Paced Auditory Serial Addition Test (PASAT). Adv Cogn Psychol 2012; 8(4): 281–291.
    1. Barker-Collo SL. Within session practice effects on the PASAT in clients with multiple sclerosis. Arch Clin Neuropsychol 2005; 20(2): 145–152.

Source: PubMed

3
Se inscrever