Page last updated: 2024-11-13

teriflunomide

Description Research Excerpts Clinical Trials Roles Classes Pathways Study Profile Bioassays Related Drugs Related Conditions Protein Interactions Research Growth Market Indicators

Cross-References

ID SourceID
PubMed CID54684141
CHEBI ID68540
SCHEMBL ID22661
MeSH IDM0519665

Synonyms (104)

Synonym
rs-61980
aubagio
hmr-1726
a-771726
su-0020
teriflunomide [inn]
(z)-2-cyano-alpha'alpha'alpha-trifluoro-3-hydroxy-p-crotonotoluidide
rs 61980
n-(4-trifluoromethylphenyl)-2-cyano-2-hydroxycrotonamide
2-hydroxyethylidene-cyanoacetic acid-4-trifluoromethyl anilide
hmr 1726
flucyamide
a 771726
2-cyano-3-hydroxy-n-(4-(trifluoromethyl)phenyl)-2-butenamide
2-butenamide, 2-cyano-3-hydroxy-n-(4-(trifluoromethyl)phenyl)-
2-butenamide, 2-cyano-3-hydroxy-n-[4-(trifluoromethyl)phenyl]-
teriflunomide ,
su 20
108605-62-5
a77 1726
a771726
a77-1726
(z)-2-cyano-3-hydroxy-n-[4-(trifluoromethyl)phenyl]but-2-enamide
(2z)-2-cyano-3-hydroxy-n-[4-(trifluoromethyl)phenyl]but-2-enamide
A26 ,
chebi:68540 ,
hmr1726
(2z)-2-[hydroxy-[4-(trifluoromethyl)anilino]methylidene]-3-oxobutanenitrile
bdbm50018011
A801897
a 1726
(z)-2-cyano-3-hydroxy-n-(4-(trifluoromethyl)phenyl)-2-butenamide
(2z)-2-cyano-3-hydroxy-n-(4-(trifluoromethyl)phenyl)-2-butenamide
D10172
163451-81-8
aubagio (tn)
teriflunomide (usan)
(z)-2-cyano-alpha,alpha,alpha-trifluoro-3-hydroxy-p-crotonotoluidide
teriflunomide [usan:inn]
1c058ikg3b ,
a 77-1726
unii-1c058ikg3b
2-butenamide, 2-cyano-3-hydroxy-n-(4-(trifluoromethyl)phenyl)-, (z)-
S4169
AKOS015994773
teriflunomide(random configuration)
teriflunomide [who-dd]
teriflunomide [vandf]
leflunomide impurity b [ep impurity]
teriflunomide [ep monograph]
2-butenamide, 2-cyano-3-hydroxy-n-(4-(trifluoromethyl)phenyl)-, (2z)-
teriflunomide [orange book]
teriflunomide [mi]
teriflunomide [usan]
teriflunomida
teriflunomidum
gtpl6844
n-(4-trifluoromethylphenyl)-2-cyano-3-hydroxycrotonamide
SCHEMBL22661
teriflunomide, a77 1726
LE-0275
2-cyano-3-hydroxy-n-(4-(trifluoromethyl)phenyl)but-2-enamide
T3287
(z)-2-cyano-3-hydroxy-n-(4-(trifluoromethyl)phenyl)but-2-enamide
2-cyano-3-oh-n-(4-trifluoromethylphenyl)croton amide
2-cyano-3-hydroxy-n-[4-(trifluoromethyl)phenyl]-2-butenamide
2-butenamide, 2-cyano-3-hydroxy-n-[4-(trifluoromethyl)phenyl]-, (2z)-
AC-26446
n-[4-(trifluoromethyl)phenyl]-2-cyano-3-hydroxycrotonamide
(z)-2-cyano-3-hydroxy-n-[4-(trifluoromethyl)phenyl]-2-butenamide
AB01565775_02
mfcd00910058
(2z)-2-cyano-3-hydroxy-n-[4-(trifluoromethyl)phenyl]but-2-enamide (teriflunomide)
J-010046
SW219377-1
teriflunomide(a-771726)
DB08880
a 77-1726;a771726;hmr1726;cas# 108605-62-5
DTXSID80893457 ,
Q3077133
(z)-2-cyano-3-hydroxy-n-(4-(trifluoromethyl)phenyl)but-2-enamide.
leflunomide ep impurity b
teriflunomide; leflunomide usp rc b; cyano keto leflunomide impurity; n-(4-trifluoromethylphenyl)-2-cyano-3-hydroxycrotonamide; 2-cyano-3-hydroxy-n-[4-(trifluoromethyl)phenyl]-2-beuteamide
dimethyl aprobarbital
NCGC00263218-13
CCG-267145
NCGC00263218-07
a 1726, a77-1726, a771726, flucyamide, hmr 1726, n-(4-trifluoromethylphenyl)-2-cyano-3-hydroxycrotoamide, su 20
A882574
2-cyano-3-hydroxy-n-[4-(trifluoromethyl)phenyl]-2z-butenamide
nsc-766118
nsc766118
a77 1726 (e/z) mixture
CS-0033021
(e/z)-teriflunomide
EN300-189832
2-cyano-3-hydroxy-n-[4-(trifluoromethyl)phenyl]but-2-enamide
HY-110159
l04aa31
leflunomide impurity b (ep impurity)
teriflunomide (ep monograph)
dtxcid401323471
(2z)-2-cyano-3-hydroxy-n-(4-(trifluoromethyl)phenyl)but-2-enamide
Z1878131003

Research Excerpts

Overview

Teriflunomide is a disease modifying treatment (DMT) approved for relapsing-remitting multiple sclerosis (RRMS) in adults and children. It is an oral medication that has shown promise in improving outcomes for pediatric MS patients.

ExcerptReferenceRelevance
"Teriflunomide is an inhibitor of pyrimidine synthesis available as a first-line treatment for relapsing-remitting multiple sclerosis. "( Hepatotoxicity associated with the use of teriflunomide in a patient with multiple sclerosis: A case report.
Domingos, JA; Ferreira, CM; Gubert, VT; Guerra-Shinohara, EM; Monreal, MTFD; Oliveira, VM; Salgado, PR; Vasconcelos-Pereira, EF, 2021
)
2.33
"Teriflunomide is a once-daily oral immunomodulator that has demonstrated efficacy and acceptable safety in clinical trials of adults with relapsing forms of MS (RMS)."( Effectiveness and safety of switching to teriflunomide in older patients with relapsing multiple sclerosis: A real-world retrospective multicenter analysis.
Becker, D; Berkovich, R; Bora, A; Bzdek, KG; Chavin, J; Dumlao, M; Hua, LH; Lublin, AL; Luo, KL; Negroski, D; Quach, C; Ranno, AE; Rawlings, AM; Sellers, D; Wells, DP; Wynn, D, 2023
)
1.9
"Teriflunomide is a disease modifying treatment (DMT) approved for relapsing-remitting multiple sclerosis (RRMS) in adults and children. "( Teriflunomide Concentrations in Cerebrospinal Fluid and Plasma in Patients with Multiple Sclerosis: A Pharmacokinetic Study.
Farman, H; Lycke, J; Nordin, A, 2023
)
3.8
"Teriflunomide is an oral medication that has shown promise in improving outcomes for pediatric MS patients, including reduced relapse rates and improved quality of life. "( Teriflunomide: an oral therapy for first-line treatment of children and adolescents living with relapsing-remitting multiple sclerosis.
Comi, G; Costa, GD,
)
3.02
"Teriflunomide is an oral medication approved for the treatment of patients with multiple sclerosis. "( Neuropathy in multiple sclerosis patients treated with teriflunomide.
Bulut, A; Kilic, AK; Sahbaz, G; Suzan, AA, 2023
)
2.6
"Teriflunomide is an oral disease-modifying therapy approved for treatment of relapsing forms of MS."( Teriflunomide and Epstein-Barr virus in a Spanish multiple sclerosis cohort:
Aladro, Y; Álvarez-Lafuente, R; Arroyo, R; Casanova-Peño, I; Comabella, M; Costa-Frossard, L; Domínguez-Mozo, MI; García-Domínguez, JM; García-Martínez, MÁ; González-Suárez, I; Martínez-Ginés, ML; Montalbán, X; Pilo, B; Villar, LM; Villarrubia, N, 2023
)
3.07
"Teriflunomide is an oral monotherapy used to treat relapsing multiple sclerosis. "( Lymphocytic colitis in the setting of teriflunomide use for relapsing multiple sclerosis.
Bovell, K; McEwan, L; Morrow, SA; Son, M; Ubaidat, M, 2020
)
2.27
"Teriflunomide is an oral first-line disease modifying treatment (DMT) for patients with relapsing-remitting multiple sclerosis (RRMS). "( Rebound after discontinuation of teriflunomide in patients with multiple sclerosis: 2 case reports.
Dotor García-Soto, J; Eichau, S; Fuerte-Hortigón, A; Hiraldo, J; López Ruiz, R; Navarro-Mascarell, G; Páramo, MD; Ruiz-Peña, JL; Sánchez Fernández, F, 2020
)
2.28
"Teriflunomide (TER) is an immunomodulatory agent. "( Teriflunomide inhibits activation-induced CD25 expression on T cells and may affect Foxp3-expressing regulatory T cells.
Jasiecka-Mikołajczyk, A; Socha, P, 2020
)
3.44
"Teriflunomide is an immunomodulatory agent approved for treatment of MS through inhibition of lymphocyte proliferation."( Effects of Teriflunomide on B Cell Subsets in MuSK-Induced Experimental Autoimmune Myasthenia Gravis and Multiple Sclerosis.
Hajtovic, S; Kurtuncu, M; Lazaridis, K; Turkoglu, R; Tuzun, E; Tzartos, J; Ulusoy, C; Yilmaz, V, 2021
)
1.73
"Teriflunomide is a disease-modifying drug that has been approved for treatment of relapsing-remitting multiple sclerosis. "( Twin pregnancy outcome following teriflunomide treatment in a relapsing-remitting multiple sclerosis patient: A case report.
Bramanti, P; Corallo, F; D'Aleo, G; Di Cara, M; Donato, A; Rifici, C; Sessa, E, 2020
)
2.28
"Teriflunomide is an oral disease modifying therapy for relapsing-remitting multiple sclerosis (RRMS). "( Inflammatory colitis associated with Teriflunomide.
Esfahani, NZ; Parikh, DA; Romba, MC; von Geldern, G; Wundes, A, 2020
)
2.27
"Teriflunomide is a once-daily oral immunomodulator used for the treatment of relapsing remitting forms of MS."( Fatigue in teriflunomide-treated patients with relapsing remitting multiple sclerosis in the real-world Teri-FAST study.
de Sèze, J; Delabrousse-Mayoux, JP; Devy, R; Gherib, A; Kabir, M; Planque, E; Vandhuick, O, 2021
)
1.73
"Teriflunomide is an immunomodulatory drug approved for Multiple Sclerosis (MS) treatment that inhibits dihydroorotate dehydrogenase, a mitochondrial enzyme involved in the de novo pyrimidine synthesis pathway."( COVID-19 in teriflunomide-treated patients with multiple sclerosis: A case report and literature review.
Capone, F; Di Lazzaro, V; Luce, T; Magliozzi, A; Motolese, F; Rossi, M, 2021
)
1.72
"Teriflunomide is a once-daily, oral disease-modifying therapy (DMT) for relapsing forms of multiple sclerosis (MS). "( Real-world outcomes for a complete nationwide cohort of more than 3200 teriflunomide-treated multiple sclerosis patients in The Danish Multiple Sclerosis Registry.
Bramow, S; Buron, MD; Geertsen, SS; Hilt, C; Illes, Z; Kant, M; Magyari, M; Mezei, Z; Papp, V; Rasmussen, PV; Roshanisefat, H; Sejbæk, T; Sellebjerg, F; Siersma, V; van Wingerden, J; Weglewski, A, 2021
)
2.3
"Teriflunomide is an oral disease modifying therapy for patients with relapsing remitting multiple sclerosis. "( Pulmonary embolism in patients with multiple sclerosis treated with teriflunomide: A series of three cases.
Brecl Jakob, G; Gomezelj, S; Krajnc, N; Šega Jazbec, S, 2021
)
2.3
"Teriflunomide is a drug with immunosuppressive and selective immunomodulatory action, characterized by anti-inflammatory and antiproliferative properties. "( The role of teriflunomide in Multiple Sclerosis patient: an observational study.
Bonanno, L; Bramanti, P; Ciurleo, R; Corallo, F; D'Aleo, G; Di Cara, M; Lo Buono, V; Marino, S; Rifici, C; Sessa, E; Torre, V; Venuti, G, 2022
)
2.54
"Teriflunomide 14 mg is a once-daily oral disease-modifying treatment for relapsing-remitting multiple sclerosis. "( Teriflunomide for multiple sclerosis in real-world setting.
Elkjaer, ML; Illes, Z; Molnar, T, 2017
)
3.34
"Teriflunomide is a more focused drug, acting as an inhibitor of pyrimidines synthesis, important for rapidly dividing cells such as activated lymphocytes."( Mechanism of action of three newly registered drugs for multiple sclerosis treatment.
Cudnoch-Jędrzejewska, A; Członkowski, A; Kasarełło, K; Mirowska-Guzel, D, 2017
)
1.18
"Teriflunomide is an oral therapy approved for the treatment of relapsing remitting multiple sclerosis (MS), showing both anti-inflammatory and antiviral properties. "( Treatment of Theiler's virus-induced demyelinating disease with teriflunomide.
DiSano, KD; Gilli, F; Li, L; Pachner, AR; Royce, DB, 2017
)
2.14
"Teriflunomide is an oral therapy approved for relapsing forms of multiple sclerosis which has been shown to reduce relapse rate and disability progression. "( A case of lymphoma in a patient on teriflunomide treatment for relapsing multiple sclerosis.
Alhendi, R; Gouverneur, A; Landais, A; Teron-Aboud, B, 2017
)
2.17
"Teriflunomide is an oral disease modifying therapy approved for the treatment of relapsing forms of multiple sclerosis. "( Tolerability and efficacy of colestipol hydrochloride for accelerated elimination of teriflunomide.
Aungst, A; Casady, L; Dixon, C; Maldonado, J; McCoy, B; Moreo, N; Robertson, D, 2017
)
2.12
"Teriflunomide is a once-daily oral immunomodulator approved for the treatment of relapsing-remitting multiple sclerosis (RRMS) with a consistent safety profile in clinical trials. "( Teeth loss after teriflunomide treatment: Casual or causal? A short case series.
Brescia Morra, V; Carotenuto, A; Foschi, M; Frattaruolo, N; Lugaresi, A; Mancinelli, L; Piattelli, M; Scandellari, C; Vacchiano, V, 2018
)
2.26
"Teriflunomide is a once-daily oral immunomodulatory agent approved in 80 countries for the treatment of patients with relapsing multiple sclerosis (RMS). "( Cost-effectiveness of Teriflunomide Compared to Interferon Beta-1b for Relapsing Multiple Sclerosis Patients in China.
Chirikov, V; Du, F; Gao, X; Liu, L; Liu, R; Mao, N; Xu, Y; Yeh, YC, 2019
)
2.27
"Teriflunomide is a cost-effective therapy over a lifetime time horizon compared to interferon beta-1b in the treatment of RMS patients in China. "( Cost-effectiveness of Teriflunomide Compared to Interferon Beta-1b for Relapsing Multiple Sclerosis Patients in China.
Chirikov, V; Du, F; Gao, X; Liu, L; Liu, R; Mao, N; Xu, Y; Yeh, YC, 2019
)
2.27
"Teriflunomide is an immunomodulatory drug that received FDA approval for the treatment of relapsing forms of multiple sclerosis (MS) in September 2012. "( Pharmacokinetic evaluation of teriflunomide for the treatment of multiple sclerosis.
O'Doherty, C; Polasek, TM; Rowland, A; Sorich, MJ; Wiese, MD, 2013
)
2.12
"Teriflunomide is a new compound which has recently been approved as a first-line treatment of relapsing forms of MS in the USA and Australia."( [Teriflunomide for treatment of multiple sclerosis].
Hartung, HP; Kieseier, BC; Menge, T; Meyer Zu Hörste, G; Stüve, O; Warnke, C; Wiendl, H, 2013
)
2.02
"Teriflunomide is a new oral disease-modifying therapy approved for the treatment of relapsing MS."( Teriflunomide reduces relapse-related neurological sequelae, hospitalizations and steroid use.
Bégo-Le-Bagousse, G; Comi, G; Confavreux, C; Dive-Pouletty, C; Freedman, MS; Kappos, L; Lublin, FD; Miller, AE; O'Connor, PW; Olsson, TP; Wolinsky, JS, 2013
)
2.55
"Teriflunomide is a once-daily oral immunomodulatory agent recently approved in the United States for the treatment of relapsing multiple sclerosis (RMS). "( Effects of prophylactic and therapeutic teriflunomide in transcranial magnetic stimulation-induced motor-evoked potentials in the dark agouti rat model of experimental autoimmune encephalomyelitis.
Hanak, S; Iglesias-Bregna, D; Ji, Z; Liu, L; McMonagle-Strucko, K; Petty, M; Zhang, D, 2013
)
2.1
"Teriflunomide is an inhibitor of dihydro-orotate dehydrogenase (DHODH), and is hypothesized to ameliorate multiple sclerosis by reducing proliferation of stimulated lymphocytes. "( The effects of teriflunomide on lymphocyte subpopulations in human peripheral blood mononuclear cells in vitro.
Arendt, C; Delohery, T; Jones, C; Li, L; Liu, J; Zhang, D, 2013
)
2.19
"Teriflunomide is an inhibitor of de novo pyrimidine synthesis, reducing lymphocyte proliferation, amongst other immunomodulatory effects; autoimmunity is believed to be one of the potential mechanisms of disease for multiple sclerosis."( Teriflunomide: a review of its use in relapsing multiple sclerosis.
Garnock-Jones, KP, 2013
)
2.55
"Teriflunomide is an effective and safe oral treatment option for relapsing MS. "( Teriflunomide for the treatment of relapsing multiple sclerosis: a review of clinical data.
Brunetti, L; Maroney, M; Ryan, M; Wagner, ML, 2013
)
3.28
"Teriflunomide is a new active drug which has recently been approved as a first-line treatment of relapsing forms of MS in the US, Australia, Argentina, and the European Union. "( Teriflunomide for the treatment of multiple sclerosis.
Kieseier, BC; Stüve, O; Warnke, C, 2013
)
3.28
"Teriflunomide is a once-daily, orally administered, disease-modifying drug. "( [Teriflunomide is a new peroral disease-modifying drug for multiple sclerosis (a review)].
Boĭko, AN; Boĭko, OV; Petrov, AM; Stoliarov, ID, 2013
)
2.74
"Teriflunomide is an oral disease-modifying therapy approved for treatment of relapsing or relapsing-remitting multiple sclerosis. "( Oral teriflunomide for patients with relapsing multiple sclerosis (TOWER): a randomised, double-blind, placebo-controlled, phase 3 trial.
Bagulho, T; Comi, G; Confavreux, C; Delhay, JL; Dukovic, D; Freedman, MS; Kappos, L; Miller, AE; O'Connor, P; Olsson, TP; Truffinet, P; Wolinsky, JS, 2014
)
2.36
"Teriflunomide is a safe new oral medication for treating RMS. "( Teriflunomide: a novel oral treatment for relapsing multiple sclerosis.
Carle, D; Freedman, MS; Sartori, A, 2014
)
3.29
"Teriflunomide acts as a specific inhibitor of the de novo pyrimidine biosynthesis."( Molecular pharmacodynamics of new oral drugs used in the treatment of multiple sclerosis.
di Nuzzo, L; Nasca, C; Nicoletti, F; Orlando, R, 2014
)
1.12
"Teriflunomide (Aubagio®) is a once-daily oral immunomodulatory disease modifying therapy (DMT) presently approved in several regions, including Europe, North America, Latin America and Australia, for the treatment of relapsing forms of multiple sclerosis (RMS; RRMS). "( Teriflunomide (Aubagio®) for the treatment of multiple sclerosis.
Bar-Or, A, 2014
)
3.29
"Teriflunomide is a once-daily oral immunomodulator approved for the treatment of relapsing-remitting multiple sclerosis. "( Teriflunomide reduces relapses with sequelae and relapses leading to hospitalizations: results from the TOWER study.
Bozzi, S; Comi, G; Dive-Pouletty, C; Freedman, MS; Kappos, L; Macdonell, R; Mäurer, M; Miller, AE; O'Connor, PW; Olsson, TP; Wolinsky, JS, 2014
)
3.29
"Teriflunomide is a once-daily oral immunomodulator approved for the treatment of relapsing-remitting multiple sclerosis. "( Oral teriflunomide for patients with a first clinical episode suggestive of multiple sclerosis (TOPIC): a randomised, double-blind, placebo-controlled, phase 3 trial.
Bauer, D; Benamor, M; Comi, G; Freedman, MS; Kappos, L; Miller, AE; O'Connor, PW; Olsson, TP; Truffinet, P; Wolinsky, JS, 2014
)
2.36
"Teriflunomide is a new oral disease-modifying drug (DMD), recently approved for the first-line treatment of relapsing multiple sclerosis (MS). "( Safety of teriflunomide for the management of relapsing-remitting multiple sclerosis.
Kappos, L; Papadopoulou, A; Sprenger, T, 2015
)
2.26
"Teriflunomide is a once-daily oral agent that has been licensed in the EU since August 2013 for the treatment of adult patients with relapsing-remitting multiple sclerosis (RRMS). "( Teriflunomide in Patients with Relapsing-Remitting Forms of Multiple Sclerosis.
Chan, A; Comabella, M; de Seze, J, 2016
)
3.32
"Teriflunomide is a pyrimidine synthesis inhibitor approved by both the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA) as a DMT for adults with relapsing-remitting MS (RRMS)."( Teriflunomide for multiple sclerosis.
Chu, L; Dai, Q; He, D; Li, Y; Zhang, C; Zhang, Y; Zhao, X, 2016
)
2.6
"Teriflunomide is an immunomodulatory drug that exerts an inhibitory effect on T cell activation in central nervous system of the patients with multiple sclerosis."( Effect of teriflunomide on QuantiFERON-TB Gold results.
Bua, A; Molicotti, P; Ruggeri, M; Zanetti, S, 2017
)
1.58
"Teriflunomide is a novel inhibitor of suicidal erythrocyte death."( Inhibition by Teriflunomide of Erythrocyte Cell Membrane Scrambling Following Energy Depletion, Oxidative Stress and Ionomycin.
Bissinger, R; Lang, F; Zierle, J, 2016
)
2.24
"Teriflunomide is a once-daily oral immunomodulator approved for relapsing-remitting multiple sclerosis (MS). "( The efficacy of teriflunomide in patients who received prior disease-modifying treatments: Subgroup analyses of the teriflunomide phase 3 TEMSO and TOWER studies.
Benamor, M; Comi, G; Freedman, MS; Kappos, L; Miller, AE; O'Connor, PW; Olsson, TP; Thangavelu, K; Truffinet, P; Wolinsky, JS, 2018
)
2.27
"Teriflunomide is an orally available anti-inflammatory drug that prevents T and B cell proliferation and function by inhibition of dihydroorotate dehydrogenase. "( Teriflunomide reduces behavioral, electrophysiological, and histopathological deficits in the Dark Agouti rat model of experimental autoimmune encephalomyelitis.
Dang, C; Hanak, S; Harvey, B; Iglesias-Bregna, D; Liang, J; McMonagle-Strucko, K; Merrill, JE; Pu, SF; Zhu, B, 2009
)
3.24
"Teriflunomide is a new oral disease-modifying therapy for relapsing forms of multiple sclerosis."( Randomized trial of oral teriflunomide for relapsing multiple sclerosis.
Benzerdjeb, H; Comi, G; Confavreux, C; Freedman, MS; Kappos, L; Miller, A; O'Connor, P; Olsson, TP; Truffinet, P; Wang, L; Wolinsky, JS, 2011
)
2.12

Effects

Teriflunomide 7mg has a higher incidence of diarrhea (RR=1.73, 95% CI: 1.32-2.26) and hair thinning. While terifl unomide 14mg has an higher probability of diarrhea and nausea.

Teriflunomide has been shown to slow cortical gray matter (GM) atrophy in patients with multiple sclerosis (MS) It has been reported to inhibit microglial activation in experimental models of traumatic brain injury.

ExcerptReferenceRelevance
"Teriflunomide 7mg has a higher incidence of diarrhea (RR=1.73, 95% CI: 1.32-2.26) and hair thinning (RR=1.99, 95% CI: 1.4-2.81), while teriflunomide 14mg has a higher incidence of diarrhea (RR=1.71, 95% CI: 1.34-2.18), hair thinning (RR=2.81, 95% CI: 2.02-3.91) and nausea (RR=1.65, 95% CI: 1.03-2.31) compared with placebo."( The efficacy and safety of teriflunomide based therapy in patients with relapsing multiple sclerosis: A meta-analysis of randomized controlled trials.
Fang, J; Lu, X; Wang, J; Xu, M; Zhu, X, 2016
)
1.45
"Teriflunomide has an acceptable and manageable safety and tolerability profile."( Pathophysiology of multiple sclerosis and the place of teriflunomide.
Gold, R; Wolinsky, JS, 2011
)
1.34
"Teriflunomide has been approved as a first-line treatment for relapsing remitting MS."( Teriflunomide as a therapeutic means for myelin repair.
Akkermann, R; Göttle, P; Groh, J; Gruchot, J; Hartung, HP; Küry, P; Martini, R; Prigione, A; Reiche, L; Rychlik, N; Samper Agrelo, I; Werner, L; Zink, A, 2023
)
3.07
"Teriflunomide has been shown to slow cortical gray matter (GM) atrophy in patients with multiple sclerosis (MS). "( Teriflunomide's effect on humoral response to Epstein-Barr virus and development of cortical gray matter pathology in multiple sclerosis.
Bergsland, N; Durfee, J; Hagemeier, J; Kolb, C; Ramanathan, M; Ramasamy, DP; Weinstock-Guttman, B; Zivadinov, R, 2019
)
3.4
"Teriflunomide has been approved for multiple sclerosis (MS) in 2012 for its immunoregulatory function."( Teriflunomide suppresses T helper cells and dendritic cells to alleviate experimental autoimmune uveitis.
Chen, X; Chen, Y; Li, H; Li, Y; Li, Z; Liang, D; Su, W; Tian, Y; Xie, Y; Yu, J; Zhu, W, 2019
)
2.68
"Teriflunomide has been reported to inhibit microglial activation in experimental models of traumatic brain injury. "( Neuroprotective Action of Teriflunomide in a Mouse Model of Transient Middle Cerebral Artery Occlusion.
Cui, K; Fu, X; Gao, Y; Li, X; Liao, L; Liu, C; Liu, X; Lu, H; Lu, Z; Man, J; Wang, J; Yu, L; Zhang, D; Zhang, Y; Zhang, Z, 2020
)
2.3
"Teriflunomide has been approved as a first-line treatment for RRMS in the USA and the European Union."( Teriflunomide promotes oligodendroglial differentiation and myelination.
Göttle, P; Hartung, HP; Kremer, D; Küry, P; Manousi, A; Reiche, L, 2018
)
2.64
"Teriflunomide has shown a favorable safety and efficacy profile in RRMS and is a therapeutic option for a distinct group of adult patients with RRMS."( From Leflunomide to Teriflunomide: Drug Development and Immunosuppressive Oral Drugs in the Treatment of Multiple Sclerosis.
Aly, L; Hemmer, B; Korn, T, 2017
)
2.22

Treatment

Treatment with teriflunomide resulted in reduced clinical and pathological scores of retinal inflammations, accompanied by decreased intraocular infiltration of Th17 and Th1 cells. Both doses significantly reduced total lesion volume and number of gadolinium-enhancing T1 lesions.

ExcerptReferenceRelevance
"Teriflunomide treatment (10 mg/kg/day, intraperitoneal) was initiated to one group of mice (n = 21) following the third immunization and continued for 5 weeks."( The Beneficial Clinical Effects of Teriflunomide in Experimental Autoimmune Myasthenia Gravis and the Investigation of the Possible Immunological Mechanisms.
Eken, A; Koseoglu, E; Muhtaroglu, S; Sungur, N; Zararsiz, G, 2023
)
1.91
"Teriflunomide treatment has effectively ameliorated MuSK-autoimmunity and thus may putatively be used in long-term management of MuSK-MG as an auxiliary treatment method."( Effects of Teriflunomide on B Cell Subsets in MuSK-Induced Experimental Autoimmune Myasthenia Gravis and Multiple Sclerosis.
Hajtovic, S; Kurtuncu, M; Lazaridis, K; Turkoglu, R; Tuzun, E; Tzartos, J; Ulusoy, C; Yilmaz, V, 2021
)
1.73
"Teriflunomide-treated patients generally mounted effective immune responses to seasonal influenza vaccination, consistent with preservation of protective immune responses."( Teriflunomide effect on immune response to influenza vaccine in patients with multiple sclerosis.
Bar-Or, A; Bauer, D; Benamor, M; Chambers, S; Freedman, MS; Jodl, S; Kremenchutzky, M; Menguy-Vacheron, F; O'Connor, PW; Truffinet, P, 2013
)
3.28
"Teriflunomide-treated patients spent fewer nights in hospital for relapse (p < 0.01)."( Teriflunomide reduces relapse-related neurological sequelae, hospitalizations and steroid use.
Bégo-Le-Bagousse, G; Comi, G; Confavreux, C; Dive-Pouletty, C; Freedman, MS; Kappos, L; Lublin, FD; Miller, AE; O'Connor, PW; Olsson, TP; Wolinsky, JS, 2013
)
2.55
"Teriflunomide treatment was also associated with significant efficacy on MRI measures of disease activity in TEMSO; both doses significantly reduced total lesion volume and number of gadolinium-enhancing T1 lesions."( Teriflunomide: a once-daily oral medication for the treatment of relapsing forms of multiple sclerosis.
Miller, AE, 2015
)
2.58
"Teriflunomide-treated patients continued the original dose; those previously receiving placebo were randomized 1:1 to teriflunomide 14 mg or 7 mg."( Long-term safety and efficacy of teriflunomide: Nine-year follow-up of the randomized TEMSO study.
Benamor, M; Bouchard, JP; Comi, G; Freedman, MS; Kappos, L; Lawson, VJ; Lebrun-Frenay, C; Liang, J; Mares, J; Miller, AE; O'Connor, P; Thangavelu, K; Truffinet, P; Wolinsky, JS, 2016
)
1.44
"Treatment with teriflunomide resulted in an unadjusted risk reduction of 63% and an adjusted risk reduction of 72%, relative to placebo, in preventing a first clinical demyelinating event. "( Teriflunomide and Time to Clinical Multiple Sclerosis in Patients With Radiologically Isolated Syndrome: The TERIS Randomized Clinical Trial.
Azevedo, C; Bourre, B; Bovis, F; Ciron, J; Clavelou, P; Cohen, M; De Seze, J; Derache, N; Durand-Dubief, F; Efendi, H; Hoepner, R; Kantarci, OH; Karabudak, R; Labauge, P; Landes-Chateau, C; Le Page, E; Lebrun-Frénay, C; Mondot, L; Okuda, DT; Papeix, C; Pelletier, D; Siva, A; Sormani, MP; Terzi, M; Thouvenot, E; Turan, OF; Vermersch, P; Wiertlewski, S; Yucear, N, 2023
)
2.71
"Treatment with teriflunomide was associated with a reduction of the levels of IgG antibody titers against EBV and HHV-6. "( Teriflunomide and Epstein-Barr virus in a Spanish multiple sclerosis cohort:
Aladro, Y; Álvarez-Lafuente, R; Arroyo, R; Casanova-Peño, I; Comabella, M; Costa-Frossard, L; Domínguez-Mozo, MI; García-Domínguez, JM; García-Martínez, MÁ; González-Suárez, I; Martínez-Ginés, ML; Montalbán, X; Pilo, B; Villar, LM; Villarrubia, N, 2023
)
2.71
"Treatment with teriflunomide resulted in reduced clinical and pathological scores of retinal inflammations, accompanied by decreased intraocular infiltration of Th17 and Th1 cells in EAU mice."( Teriflunomide suppresses T helper cells and dendritic cells to alleviate experimental autoimmune uveitis.
Chen, X; Chen, Y; Li, H; Li, Y; Li, Z; Liang, D; Su, W; Tian, Y; Xie, Y; Yu, J; Zhu, W, 2019
)
2.3
"Upon treatment with teriflunomide, cytokine secretion was decreased (CXCL10, 3-fold; CCL2, 2.5-fold; IL-6, 2.2-fold; p < 0.001) and monomethylfumarate treatment led to 2.9-fold lower CXCL10 secretion (p < 0.001)."( Teriflunomide and monomethylfumarate target HIV-induced neuroinflammation and neurotoxicity.
Ambrosius, B; Chan, A; Faissner, S; Gold, R; Grewe, B; Grunwald, T; Guse, K; von Lehe, M, 2017
)
2.21

Toxicity

Teriflunomide was as effective and safe in the Chinese subpopulation as it was in the overall population of patients in the TOWER trial. Nail loss might represent a new, reversible, adverse event associated with terifl unomide treatment.

ExcerptReferenceRelevance
" These data demonstrate that the parent forms of leflunomide and A77 1726 are more toxic to hepatocytes than their poorly characterized metabolites, indicating that the metabolic process of leflunomide is a detoxification step rather than an initiating event leading to toxicity."( Hepatic cytochrome P450s attenuate the cytotoxicity induced by leflunomide and its active metabolite A77 1726 in primary cultured rat hepatocytes.
Greenhaw, J; Salminen, WF; Shi, Q; Yang, X, 2011
)
0.37
"2%) discontinued (19% due to treatment-emergent adverse events (TEAEs))."( 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.
Bar-Or, A; Benzerdjeb, H; Confavreux, C; Freedman, MS; Li, DK; O'Connor, PW; Reiman, LE; Traboulsee, AL; Truffinet, P; Wang, D, 2012
)
0.64
" We summarize adverse events observed in the Phase II and III clinical trials and the safety data from the long-term extension phases as well as the > 15-year post-marketing experience with the parent drug, leflunomide."( Safety of teriflunomide for the management of relapsing-remitting multiple sclerosis.
Kappos, L; Papadopoulou, A; Sprenger, T, 2015
)
0.82
" Safety assessments included adverse events, laboratory parameters, and physical examinations."( Pooled safety and tolerability data from four placebo-controlled teriflunomide studies and extensions.
Benamor, M; Comi, G; Dukovic, D; Freedman, MS; Kappos, L; Leist, TP; Miller, AE; O'Connor, PW; Olsson, TP; Truffinet, P; Wolinsky, JS, 2016
)
0.67
"In Pool 1, the number of patients experiencing adverse events and serious adverse events was similar in the three treatment groups."( Pooled safety and tolerability data from four placebo-controlled teriflunomide studies and extensions.
Benamor, M; Comi, G; Dukovic, D; Freedman, MS; Kappos, L; Leist, TP; Miller, AE; O'Connor, PW; Olsson, TP; Truffinet, P; Wolinsky, JS, 2016
)
0.67
" The most common adverse events (AEs) matched those in the core study."( Long-term safety and efficacy of teriflunomide: Nine-year follow-up of the randomized TEMSO study.
Benamor, M; Bouchard, JP; Comi, G; Freedman, MS; Kappos, L; Lawson, VJ; Lebrun-Frenay, C; Liang, J; Mares, J; Miller, AE; O'Connor, P; Thangavelu, K; Truffinet, P; Wolinsky, JS, 2016
)
0.72
" However, the incidence of serious adverse events was similar across groups."( The efficacy and safety of teriflunomide based therapy in patients with relapsing multiple sclerosis: A meta-analysis of randomized controlled trials.
Fang, J; Lu, X; Wang, J; Xu, M; Zhu, X, 2016
)
0.73
" Adverse effects of teriflunomide are well characterized and can be considered manageable."( Comparison of efficacy and safety of oral agents for the treatment of relapsing-remitting multiple sclerosis.
Bramanti, P; Guarnera, C; Mazzon, E, 2017
)
0.78
"Nail loss might represent a new, reversible, adverse event associated with teriflunomide treatment."( "Nail loss after teriflunomide treatment: A new potential adverse event".
Amerio, P; De Luca, G; di Ioia, M; Di Tommaso, V; Lugaresi, A; Mancinelli, L; Onofrj, M, 2017
)
1.02
" Mode of action, adverse effects, reported risks for infections and malignancies, and pregnancy related issues are discussed in the review."( Managing the side effects of multiple sclerosis therapy: pharmacotherapy options for patients.
Rommer, PS; Zettl, UK, 2018
)
0.48
"To describe DILI events with MS drugs by analyzing the FDA Adverse Event Reporting System."( Liver injury with drugs used for multiple sclerosis: A contemporary analysis of the FDA Adverse Event Reporting System.
Antonazzo, IC; Baldin, E; Bjornevik, K; De Ponti, F; Forcesi, E; Muratori, L; Poluzzi, E; Raschi, E; Riise, T, 2019
)
0.51
" Reductions in ARR and 12-w CDW associated with teriflunomide 14 mg were comparable between the Asian and overall populations, as were the rates for adverse events and serious adverse events, with no new or unexpected safety findings."( Efficacy and safety of teriflunomide in Asian patients with relapsing forms of multiple sclerosis: A subgroup analysis of the phase 3 TOWER study.
Benamor, M; Freedman, MS; Macdonell, R; Miller, AE; Thangavelu, K; Truffinet, P; Vucic, S; Xu, X, 2019
)
1.08
" There were no differences across all treatment groups in the proportion of patients with treatment-emergent adverse events (TEAEs; 72."( Efficacy and Safety of Teriflunomide in Chinese Patients with Relapsing Forms of Multiple Sclerosis: A Subgroup Analysis of the Phase 3 TOWER Study.
Bu, BT; Cheng, Y; Dong, HQ; Dong, WL; Fan, DS; Hou, SF; Hu, XQ; Huang, DH; Huang, YN; Jin, T; Li, HF; Li, JM; Li, L; Lu, CZ; Peng, H; Qiu, W; Wang, BJ; Wu, WP; Xu, XH; Zhang, CD; Zhang, MN; Zhang, X; Zhang, XH; Zhao, G; Zhou, D, 2018
)
0.79
"Teriflunomide was as effective and safe in the Chinese subpopulation as it was in the overall population of patients in the TOWER trial."( Efficacy and Safety of Teriflunomide in Chinese Patients with Relapsing Forms of Multiple Sclerosis: A Subgroup Analysis of the Phase 3 TOWER Study.
Bu, BT; Cheng, Y; Dong, HQ; Dong, WL; Fan, DS; Hou, SF; Hu, XQ; Huang, DH; Huang, YN; Jin, T; Li, HF; Li, JM; Li, L; Lu, CZ; Peng, H; Qiu, W; Wang, BJ; Wu, WP; Xu, XH; Zhang, CD; Zhang, MN; Zhang, X; Zhang, XH; Zhao, G; Zhou, D, 2018
)
2.23
" The overall frequency of adverse events (AEs) was comparable across treatment groups, but a higher proportion of patients in the teriflunomide 7 mg/14 mg (12."( Long-term safety and efficacy of teriflunomide in patients with relapsing multiple sclerosis: Results from the TOWER extension study.
Benamor, M; Chavin, J; Comi, G; Delhay, JL; Freedman, MS; Hu, X; Kappos, L; Lublin, AL; Miller, AE; Olsson, TP; Purvis, A; Truffinet, P; Wolinsky, JS; Xu, X, 2020
)
1.04
" The incidence of adverse events was also assessed."( Prior treatment status: impact on the efficacy and safety of teriflunomide in multiple sclerosis.
Chavin, J; Comi, G; Coyle, PK; Edwards, KR; Freedman, MS; Gold, R; Kim, BJ; Korideck, H; Meca-Lallana, JE; Parajeles, A; Poole, EM; Vermersch, P, 2020
)
0.8
" We examined laboratory test abnormalities and adverse health conditions in new users."( Short-term laboratory and related safety outcomes for the multiple sclerosis oral disease-modifying therapies: an observational study.
Carruthers, R; Evans, C; Kingwell, E; Marrie, RA; Tremlett, H; Walld, R; Zhang, T; Zhu, F, 2021
)
0.62
"The short-term, real-world incidences of abnormal laboratory results or adverse events were consistent with the pivotal clinical trial findings."( Short-term laboratory and related safety outcomes for the multiple sclerosis oral disease-modifying therapies: an observational study.
Carruthers, R; Evans, C; Kingwell, E; Marrie, RA; Tremlett, H; Walld, R; Zhang, T; Zhu, F, 2021
)
0.62
" Common adverse effects of leflunomide were hyperlipidemia, leucopenia, neutropenia and liver-function alteration."( Efficacy and safety of dihydroorotate dehydrogenase (DHODH) inhibitors "leflunomide" and "teriflunomide" in Covid-19: A narrative review.
Avti, P; Bhattacharyya, A; Chhimpa, N; Kaur, H; Kumar, S; Medhi, B; Prajapat, M; Prakash, A; Sarma, P; Sharma, S; Singh, A; Singh, R; Thota, P, 2021
)
0.84
" Adverse events occurred in 96 (88%) patients in the teriflunomide group and 47 (82%) patients in the placebo group; serious adverse events occurred in 12 (11%) patients in the teriflunomide group and 6 (11%) patients in the placebo group."( Safety and efficacy of teriflunomide in paediatric multiple sclerosis (TERIKIDS): a multicentre, double-blind, phase 3, randomised, placebo-controlled trial.
Arnold, DL; Banwell, B; Benamor, M; Chitnis, T; Cui, LY; Deiva, K; Gücüyener, K; Hu, W; Kappos, L; Le-Halpere, A; Saubadu, S; Skripchenko, N; Tardieu, M; Truffinet, P, 2021
)
1.18
" Moreover, the safety was assessed by blood routine, liver and kidney function, and a questionnaire to report adverse reactions."( Serological markers exploration and real-world effectiveness and safety of teriflunomide in south Chinese patients with multiple sclerosis.
Cai, H; Chen, S; Fang, L; Jiang, F; Li, H; Li, J; Yang, H; Yin, J; Zeng, Q; Zhou, R, 2022
)
0.95
" Teriflunomide was associated with mild or moderate discomfort, and discontinuation rates due to adverse events were low."( Serological markers exploration and real-world effectiveness and safety of teriflunomide in south Chinese patients with multiple sclerosis.
Cai, H; Chen, S; Fang, L; Jiang, F; Li, H; Li, J; Yang, H; Yin, J; Zeng, Q; Zhou, R, 2022
)
1.86
"Teriflunomide-induced liver injury in patients with multiple sclerosis is a serious adverse reaction."( Hepatotoxicity associated with the use of teriflunomide in a patient with multiple sclerosis: A case report.
Domingos, JA; Ferreira, CM; Gubert, VT; Guerra-Shinohara, EM; Monreal, MTFD; Oliveira, VM; Salgado, PR; Vasconcelos-Pereira, EF, 2021
)
2.33
" However, patients taking certain oral DMTs may experience gastrointestinal (GI)-related adverse events (AEs), particularly at dose titration."( The Patient and Clinician Assessment of Gastrointestinal (GI) Related Adverse Events Associated with Oral Disease-Modifying Therapies in Multiple Sclerosis: A Qualitative Study.
Cano, S; Elliott, E; Jivraj, F; Kang, S; Kapadia, S; Reedie, S; Rock, M; Strzok, S, 2022
)
0.72
"Thus, the assessment of 24 weeks treatment demonstrated that DIV is a highly effective, safe and convenient option for the treatment of RRMS patients, both naive and previously treated with disease modifying therapy."( [Efficacy and safety of divozilimab during 24-week treatment of multiple sclerosis patients in randomized double-blind placebo-controlled clinical trial BCD-132-2].
Alifirova, VM; Artemyeva, AV; Bolsun, DD; Boyko, AN; Goncharova, ZA; Greshnova, IV; Khabirov, FA; Kotov, SV; Linkova, YN; Lukashevich, IG; Malkova, NA; Mishin, GN; Parshina, EV; Poverennova, IY; Prakhova, LN; Shchur, SG; Sivertseva, SA; Smagina, IV; Totolyan, NA; Trinitatsky, YV; Trushnikova, TN; Zaslavsky, LG; Zinkina-Orikhan, AV, 2023
)
0.91
" This study aims to investigate and compare adherence, adverse event (AE) profiles, and frequencies, main reasons for treatment discontinuation under Teriflunomide (TERI), Dimethyl Fumarate (DMF), and Fingolimod (FNG) for relapsing-remitting MS (RRMS) patients."( Real-life outcomes for oral disease-modifying treatments of relapsing-remitting multiple sclerosis patients: Adherence and adverse event profiles from Marmara University.
Engin, E; Günal, D; Sünter, G; Vural, E; Yıldırım, KA, 2023
)
1.11
" The most common reasons for treatment discontinuation in order of frequency were adverse events, the progression of the disease, and the persistence of relapses."( Real-life outcomes for oral disease-modifying treatments of relapsing-remitting multiple sclerosis patients: Adherence and adverse event profiles from Marmara University.
Engin, E; Günal, D; Sünter, G; Vural, E; Yıldırım, KA, 2023
)
0.91
"To compare adverse effects of immunotherapies for people with MS or clinically isolated syndrome (CIS), and to rank these treatments according to their relative risks of adverse effects through network meta-analyses (NMAs)."( Adverse effects of immunotherapies for multiple sclerosis: a network meta-analysis.
Benedetti, MD; Capobussi, M; Castellini, G; Featherstone, R; Filippini, G; Frau, S; Gonzalez-Lorenzo, M; Lucenteforte, E; Perduca, V; Tramacere, I; Virgili, G, 2023
)
0.91
" Our review, along with other work in the literature, confirms poor-quality reporting of adverse events from RCTs of interventions."( Adverse effects of immunotherapies for multiple sclerosis: a network meta-analysis.
Benedetti, MD; Capobussi, M; Castellini, G; Featherstone, R; Filippini, G; Frau, S; Gonzalez-Lorenzo, M; Lucenteforte, E; Perduca, V; Tramacere, I; Virgili, G, 2023
)
0.91

Pharmacokinetics

Plasma teriflunomide (active metabolite of leflunomid) concentrations were determined, and pharmacokinetic parameters were calculated. Given the lack of pharmacokinetics evaluation of terifnomide in the Iranian context, the present 2-way crossover study aimed to assess the pharmacokeretic properties and bioequivalence of 2 terif lunomides.

ExcerptReferenceRelevance
" The pharmacokinetic results showed that it exhibited first order kinetic characteristics."( Pharmacokinetics of leflunomide in Chinese healthy volunteers.
Jin, Y; Li, CY; Li, J; Li, YH; Xu, SY; Yao, HW; Zhang, YF, 2002
)
0.31
" A steady-state infusion model best described the pharmacokinetic data."( Population pharmacokinetics and association between A77 1726 plasma concentrations and disease activity measures following administration of leflunomide to people with rheumatoid arthritis.
Chan, V; Charles, BG; Tett, SE, 2005
)
0.33
" A population pharmacokinetic model was developed to estimate the oral clearance (CL/F) and volume of distribution (V/F)."( Investigation of the influence of CYP1A2 and CYP2C19 genetic polymorphism on 2-Cyano-3-hydroxy-N-[4-(trifluoromethyl)phenyl]-2-butenamide (A77 1726) pharmacokinetics in leflunomide-treated patients with rheumatoid arthritis.
Bohanec Grabar, P; Dolzan, V; Grabnar, I; Logar, D; Mrhar, A; Peterlin Masic, L; Rozman, B; Suput, D; Tomsic, M; Trdan, T, 2009
)
0.35
" This study aimed to determine if a pharmacodynamic relationship exists between BK viral load reduction and leflunomide metabolite, A77 1726, serum concentrations."( Leflunomide efficacy and pharmacodynamics for the treatment of BK viral infection.
Baliga, P; Bratton, C; Chavin, K; Krisl, JC; McGillicuddy, J; Pilch, N; Taber, DJ; Thomas, B, 2012
)
0.38
" Plasma teriflunomide (active metabolite of leflunomide) concentrations were determined, and pharmacokinetic parameters were calculated."( Pharmacokinetics and bioequivalence evaluation of leflunomide tablets in Korean healthy volunteers.
Kim, EY; Kim, HS; Lim, YJ; Moon, BS; Oh, M; Shim, EJ; Shin, JG; Shon, JH; Song, GS, 2013
)
0.82
" They were applied to a pharmacokinetic study in rats by a single oral dose."( Comparison of LC-UV and LC-MS methods for simultaneous determination of teriflunomide, dimethyl fumarate and fampridine in human plasma: application to rat pharmacokinetic study.
Raja, RK; Suneetha, A, 2016
)
0.67
"Based on a pharmacokinetic (PK) study assessing the relative bioavailability of teriflunomide sodium compared to leflunomide, a population pharmacokinetic (Pop PK) analysis was firstly conducted using non-linear mixed effect model."( A population pharmacokinetic study to accelerate early phase clinical development for a novel drug, teriflunomide sodium, to treat systemic lupus erythematosus.
Chen, X; Hu, P; Jiang, J; Liu, D; Wu, Y; Yao, X, 2019
)
0.96
" Given the lack of pharmacokinetic evaluation of teriflunomide in the Iranian context, the present 2-way crossover study aimed to assess the pharmacokinetic properties and bioequivalence of 2 teriflunomide formulations."( Pharmacokinetics and Bioequivalence Studies of Teriflunomide in Healthy Iranian Volunteers.
Dibaei, M; Ghasemian, E; Rouini, MR, 2020
)
1.07

Bioavailability

ExcerptReferenceRelevance
" Identical areas under the curve suggest bioavailability was 100% (AUCp."( Blood distribution and single-dose pharmacokinetics of leflunomide.
Dias, VC; LeGatt, DF; Lucien, J; Yatscoff, RW, 1995
)
0.29
" The bioavailability after a single oral dose of leflunomide was 100%."( Pharmacokinetics and pharmacodynamics of A77 1726 and leflunomide in domestic cats.
Chen, YJ; Craigmill, A; Gregory, CR; Kyles, AE; Mehl, ML; Tell, L, 2012
)
0.38
" However, no published data is available regarding the bioavailability of this in the Indian population."( Comparative bioequivalence study of leflunomide tablets in Indian healthy volunteers.
Agarwal, S; Chattaraj, TK; Das, A; Ghosh, D; Pal, TK; Sarkar, AK, 2012
)
0.38
" Moreover, an oral bioavailability of 40% for compound 50 was determined from in vivo pharmacokinetic studies."( Novel selective and potent inhibitors of malaria parasite dihydroorotate dehydrogenase: discovery and optimization of dihydrothiophenone derivatives.
Diao, Y; Han, D; Huang, J; Li, H; Ren, X; Sun, D; Xu, M; Xu, Y; Zhao, Z; Zhou, H; Zhu, J; Zhu, L, 2013
)
0.39
"Based on a pharmacokinetic (PK) study assessing the relative bioavailability of teriflunomide sodium compared to leflunomide, a population pharmacokinetic (Pop PK) analysis was firstly conducted using non-linear mixed effect model."( A population pharmacokinetic study to accelerate early phase clinical development for a novel drug, teriflunomide sodium, to treat systemic lupus erythematosus.
Chen, X; Hu, P; Jiang, J; Liu, D; Wu, Y; Yao, X, 2019
)
0.96

Dosage Studied

Colestipol HCl may offer an alternative method for accelerated elimination of teriflunomide with potentially improved tolerability and more favorable dosing and administration options. The developed EHC Pop PK model exhibited the ability to predict PK profiles of terflaminomide in patients after long-term dosing. It could be utilized to support phase II trial design.

ExcerptRelevanceReference
" Such information would prove invaluable in determining the appropriate medium for analysis and optimal immunosuppressive dosing regimes."( Blood distribution and single-dose pharmacokinetics of leflunomide.
Dias, VC; LeGatt, DF; Lucien, J; Yatscoff, RW, 1995
)
0.29
"01), but this would not be clinically important in terms of dosage changes."( Population pharmacokinetics and association between A77 1726 plasma concentrations and disease activity measures following administration of leflunomide to people with rheumatoid arthritis.
Chan, V; Charles, BG; Tett, SE, 2005
)
0.33
" The marked variability in pharmacokinetics suggests a place for individualized dosing of leflunomide in RA therapy."( Population pharmacokinetics and association between A77 1726 plasma concentrations and disease activity measures following administration of leflunomide to people with rheumatoid arthritis.
Chan, V; Charles, BG; Tett, SE, 2005
)
0.33
" Leflunomide was dosed to a targeted blood level of active metabolite, A77 1726, of 50 microg/ml to 100 microg/ml (150 microM to 300 microM)."( Treatment of renal allograft polyoma BK virus infection with leflunomide.
Atwood, W; Foster, P; Garfinkel, M; Gillen, D; Harland, R; Javaid, B; Jordan, J; Josephson, MA; Kadambi, P; Meehan, S; Millis, MJ; Sadhu, M; Thistlethwaite, RJ; Williams, J, 2006
)
0.33
" Neurological evaluation demonstrated that prophylactic dosing of teriflunomide at 3 and 10 mg/kg delayed disease onset and reduced maximal and cumulative scores."( Teriflunomide reduces behavioral, electrophysiological, and histopathological deficits in the Dark Agouti rat model of experimental autoimmune encephalomyelitis.
Dang, C; Hanak, S; Harvey, B; Iglesias-Bregna, D; Liang, J; McMonagle-Strucko, K; Merrill, JE; Pu, SF; Zhu, B, 2009
)
2.03
" To examine details of the relationships between leflunomide dosing and patient response, it is necessary to have an assay that is sufficiently sensitive to measure the minor fraction of free teriflunomide in patient samples."( Quantitation of total and free teriflunomide (A77 1726) in human plasma by LC-MS/MS.
Cleland, L; Hopkins, A; James, M; Proudman, S; Rakhila, H; Rozek, T; Wiese, M, 2011
)
0.85
"Teriflunomide - a dihydroorotate dehydrogenase (DHODH) inhibitor; AL8697 - a selective p38 MAPK inhibitor; and tofacitinib - a Janus kinase (JAK) inhibitor; were selected as representatives of their class and dose-response studies carried out using a therapeutic 10-day administration scheme in arthritic rats."( Profiling of dihydroorotate dehydrogenase, p38 and JAK inhibitors in the rat adjuvant-induced arthritis model: a translational study.
Balagué, C; Godessart, N; Pont, M; Prats, N, 2012
)
1.82
" Leflunomide can be used for RA patients on chronic dialysis without any dosage modification."( Leflunomide in dialysis patients with rheumatoid arthritis--a pharmacokinetic study.
Bergner, R; Löffler, C; Peters, L; Schmitt, V, 2013
)
0.39
" While both the 7 and the 14 mg/day dosages are discussed, the 7 mg/day dosage is not approved in the EU."( Teriflunomide: a review of its use in relapsing multiple sclerosis.
Garnock-Jones, KP, 2013
)
1.83
" The intravenous DMT natalizumab (Tysabri; dosed monthly) provides high therapeutic efficacy and good compliance but is considered a second-line intervention because of the associated increased risk for progressive multifocal leukoencephalopathy."( An update on new and emerging therapies for relapsing-remitting multiple sclerosis.
Weinstock-Guttman, B, 2013
)
0.39
"A total of 5 new agents and 1 new dosage formulation were approved by the FDA for the treatment of RRMS since 2010."( New FDA-Approved Disease-Modifying Therapies for Multiple Sclerosis.
Aloi, JJ; English, C, 2015
)
0.42
" After oral dosing (15 mg/kg), the plasma exposure (AUC0-t) of leflunomide increased to 3-fold in HRN mice compared with wild type mice."( Inhibition of hepatic cytochrome P450 enzymes and sodium/bile acid cotransporter exacerbates leflunomide-induced hepatotoxicity.
Cao, YY; Chen, C; Lin, YF; Luan, Y; Ma, LL; Ni, X; Pan, GY; Wang, J; Wang, L; Wu, ZT; Zhang, XF, 2016
)
0.43
" Currently, AEPs with oral cholestyramine or activated charcoal are available but are restricted by adverse effects, limited administration routes, and dosing frequencies."( Tolerability and efficacy of colestipol hydrochloride for accelerated elimination of teriflunomide.
Aungst, A; Casady, L; Dixon, C; Maldonado, J; McCoy, B; Moreo, N; Robertson, D, 2017
)
0.68
" Although colestipol HCl did not completely eliminate teriflunomide with the same effectiveness as cholestyramine, it may offer an alternative method for accelerated elimination of teriflunomide with potentially improved tolerability and more favorable dosing and administration options."( Tolerability and efficacy of colestipol hydrochloride for accelerated elimination of teriflunomide.
Aungst, A; Casady, L; Dixon, C; Maldonado, J; McCoy, B; Moreo, N; Robertson, D, 2017
)
0.93
"We report that the subcutaneous delivery of TEF-MTX-HAp-NP was most effective as it successfully reduced the dosage by half for maximizing therapeutic efficacy and minimizing side effects."( Co-Delivery of Teriflunomide and Methotrexate from Hydroxyapatite Nanoparticles for the Treatment of Rheumatoid Arthritis: In Vitro Characterization, Pharmacodynamic and Biochemical Investigations.
Ahmad, FJ; Kumar, V; Leekha, A; Pandey, S; Rai, N; Talegaonkar, S; Verma, AK, 2018
)
0.83
" In order to predict teriflunomide PK profiles for multiple dosing of teriflunomide sodium in SLE patients, a model integrating enterohepatic circulation (EHC) mechanism was utilized to simulate the teriflunomide PK profile after multiple dosing of 20 mg/day leflunomide, and compare it to the teriflunomide PK profile in a 20 mg/day leflunomide multiple dose study in rheumatoid arthritis patients."( A population pharmacokinetic study to accelerate early phase clinical development for a novel drug, teriflunomide sodium, to treat systemic lupus erythematosus.
Chen, X; Hu, P; Jiang, J; Liu, D; Wu, Y; Yao, X, 2019
)
1.05
" After this model was confirmed to capture EHC characteristics of teriflunomide in both healthy subjects and patients with rheumatoid arthritis after single and multiple dosing leflunomide, it was applied to suggest dose regimen of teriflunomide sodium in phase II study."( A population pharmacokinetic study to accelerate early phase clinical development for a novel drug, teriflunomide sodium, to treat systemic lupus erythematosus.
Chen, X; Hu, P; Jiang, J; Liu, D; Wu, Y; Yao, X, 2019
)
0.97
" The developed EHC Pop PK model exhibited the ability to predict PK profiles of teriflunomide in patients after long-term dosing and could be utilized to support phase II trial design."( A population pharmacokinetic study to accelerate early phase clinical development for a novel drug, teriflunomide sodium, to treat systemic lupus erythematosus.
Chen, X; Hu, P; Jiang, J; Liu, D; Wu, Y; Yao, X, 2019
)
0.96
" In vivo, at a dosage not efficacious in wt animals, teri treatment ameliorated clinical EAE in abcg2-KO mice which was accompanied by higher spinal cord tissue concentrations of teri."( Functional relevance of the multi-drug transporter abcg2 on teriflunomide therapy in an animal model of multiple sclerosis.
Chan, A; Demir, S; Engelhardt, B; Guse, K; Hermann, DM; Hoepner, R; Lühder, F; Pedreiturria, X; Pistor, M; Remlinger, J; Salmen, A; Thiele Née Schrewe, L; Tietz, S; Turner, T; Wiese, S, 2020
)
0.8
" However, adjustments to dosing schedules may help de-risk the chance of infection further and reduce the concerns of people with MS being treated during the COVID-19 pandemic."( The underpinning biology relating to multiple sclerosis disease modifying treatments during the COVID-19 pandemic.
Amor, S; Baker, D; Giovannoni, G; Kang, AS; Schmierer, K, 2020
)
0.56
"Findings will provide timely information on the safety, efficacy, and optimal dosing of t-PA to treat moderate/severe COVID-19-induced ARDS, which can be rapidly adapted to a phase III trial (NCT04357730; FDA IND 149634)."(
Abbasi, S; Abd El-Wahab, A; Abdallah, M; Abebe, G; Aca-Aca, G; Adama, S; Adefegha, SA; Adidigue-Ndiome, R; Adiseshaiah, P; Adrario, E; Aghajanian, C; Agnese, W; Ahmad, A; Ahmad, I; Ahmed, MFE; Akcay, OF; Akinmoladun, AC; Akutagawa, T; Alakavuklar, MA; Álava-Rabasa, S; Albaladejo-Florín, MJ; Alexandra, AJE; Alfawares, R; Alferiev, IS; Alghamdi, HS; Ali, I; Allard, B; Allen, JD; Almada, E; Alobaid, A; Alonso, GL; Alqahtani, YS; Alqarawi, W; Alsaleh, H; Alyami, BA; Amaral, BPD; Amaro, JT; Amin, SAW; Amodio, E; Amoo, ZA; Andia Biraro, I; Angiolella, L; Anheyer, D; Anlay, DZ; Annex, BH; Antonio-Aguirre, B; Apple, S; Arbuznikov, AV; Arinsoy, T; Armstrong, DK; Ash, S; Aslam, M; Asrie, F; Astur, DC; Atzrodt, J; Au, DW; Aucoin, M; Auerbach, EJ; Azarian, S; Ba, D; Bai, Z; Baisch, PRM; Balkissou, AD; Baltzopoulos, V; Banaszewski, M; Banerjee, S; Bao, Y; Baradwan, A; Barandika, JF; Barger, PM; Barion, MRL; Barrett, CD; Basudan, AM; Baur, LE; Baz-Rodríguez, SA; Beamer, P; Beaulant, A; Becker, DF; Beckers, C; Bedel, J; Bedlack, R; Bermúdez de Castro, JM; Berry, JD; Berthier, C; Bhattacharya, D; Biadgo, B; Bianco, G; Bianco, M; Bibi, S; Bigliardi, AP; Billheimer, D; Birnie, DH; Biswas, K; Blair, HC; Bognetti, P; Bolan, PJ; Bolla, JR; Bolze, A; Bonnaillie, P; Borlimi, R; Bórquez, J; Bottari, NB; Boulleys-Nana, JR; Brighetti, G; Brodeur, GM; Budnyak, T; Budnyk, S; Bukirwa, VD; Bulman, DM; Burm, R; Busman-Sahay, K; Butcher, TW; Cai, C; Cai, H; Cai, L; Cairati, M; Calvano, CD; Camacho-Ordóñez, A; Camela, E; Cameron, T; Campbell, BS; Cansian, RL; Cao, Y; Caporale, AS; Carciofi, AC; Cardozo, V; Carè, J; Carlos, AF; Carozza, R; Carroll, CJW; Carsetti, A; Carubelli, V; Casarotta, E; Casas, M; Caselli, G; Castillo-Lora, J; Cataldi, TRI; Cavalcante, ELB; Cavaleiro, A; Cayci, Z; Cebrián-Tarancón, C; Cedrone, E; Cella, D; Cereda, C; Ceretti, A; Ceroni, M; Cha, YH; Chai, X; Chang, EF; Chang, TS; Chanteux, H; Chao, M; Chaplin, BP; Chaturvedi, S; Chaturvedi, V; Chaudhary, DK; Chen, A; Chen, C; Chen, HY; Chen, J; Chen, JJ; Chen, K; Chen, L; Chen, Q; Chen, R; Chen, SY; Chen, TY; Chen, WM; Chen, X; Chen, Y; Cheng, G; Cheng, GJ; Cheng, J; Cheng, YH; Cheon, HG; Chew, KW; Chhoker, S; Chiu, WN; Choi, ES; Choi, MJ; Choi, SD; Chokshi, S; Chorny, M; Chu, KI; Chu, WJ; Church, AL; Cirrincione, A; Clamp, AR; Cleff, MB; Cohen, M; Coleman, RL; Collins, SL; Colombo, N; Conduit, N; Cong, WL; Connelly, MA; Connor, J; Cooley, K; Correa Ramos Leal, I; Cose, S; Costantino, C; Cottrell, M; Cui, L; Cundall, J; Cutaia, C; Cutler, CW; Cuypers, ML; da Silva Júnior, FMR; Dahal, RH; Damiani, E; Damtie, D; Dan-Li, W; Dang, Z; Dasa, SSK; Davin, A; Davis, DR; de Andrade, CM; de Jong, PL; de Oliveira, D; de Paula Dorigam, JC; Dean, A; Deepa, M; Delatour, C; Dell'Aiera, S; Delley, MF; den Boer, RB; Deng, L; Deng, Q; Depner, RM; Derdau, V; Derici, U; DeSantis, AJ; Desmarini, D; Diffo-Sonkoue, L; Divizia, M; Djenabou, A; Djordjevic, JT; Dobrovolskaia, MA; Domizi, R; Donati, A; Dong, Y; Dos Santos, M; Dos Santos, MP; Douglas, RG; Duarte, PF; Dullaart, RPF; Duscha, BD; Edwards, LA; Edwards, TE; Eichenwald, EC; El-Baba, TJ; Elashiry, M; Elashiry, MM; Elashry, SH; Elliott, A; Elsayed, R; Emerson, MS; Emmanuel, YO; Emory, TH; Endale-Mangamba, LM; Enten, GA; Estefanía-Fernández, K; Estes, JD; Estrada-Mena, FJ; Evans, S; Ezra, L; Faria de, RO; Farraj, AK; Favre, C; Feng, B; Feng, J; Feng, L; Feng, W; Feng, X; Feng, Z; Fernandes, CLF; Fernández-Cuadros, ME; Fernie, AR; Ferrari, D; Florindo, PR; Fong, PC; Fontes, EPB; Fontinha, D; Fornari, VJ; Fox, NP; Fu, Q; Fujitaka, Y; Fukuhara, K; Fumeaux, T; Fuqua, C; Fustinoni, S; Gabbanelli, V; Gaikwad, S; Gall, ET; Galli, A; Gancedo, MA; Gandhi, MM; Gao, D; Gao, K; Gao, M; Gao, Q; Gao, X; Gao, Y; Gaponenko, V; Garber, A; Garcia, EM; García-Campos, C; García-Donas, J; García-Pérez, AL; Gasparri, F; Ge, C; Ge, D; Ge, JB; Ge, X; George, I; George, LA; Germani, G; Ghassemi Tabrizi, S; Gibon, Y; Gillent, E; Gillies, RS; Gilmour, MI; Goble, S; Goh, JC; Goiri, F; Goldfinger, LE; Golian, M; Gómez, MA; Gonçalves, J; Góngora-García, OR; Gonul, I; González, MA; Govers, TM; Grant, PC; Gray, EH; Gray, JE; Green, MS; Greenwald, I; Gregory, MJ; Gretzke, D; Griffin-Nolan, RJ; Griffith, DC; Gruppen, EG; Guaita, A; Guan, P; Guan, X; Guerci, P; Guerrero, DT; Guo, M; Guo, P; Guo, R; Guo, X; Gupta, J; Guz, G; Hajizadeh, N; Hamada, H; Haman-Wabi, AB; Han, TT; Hannan, N; Hao, S; Harjola, VP; Harmon, M; Hartmann, MSM; Hartwig, JF; Hasani, M; Hawthorne, WJ; Haykal-Coates, N; Hazari, MS; He, DL; He, P; He, SG; Héau, C; Hebbar Kannur, K; Helvaci, O; Heuberger, DM; Hidalgo, F; Hilty, MP; Hirata, K; Hirsch, A; Hoffman, AM; Hoffmann, JF; Holloway, RW; Holmes, RK; Hong, S; Hongisto, M; Hopf, NB; Hörlein, R; Hoshino, N; Hou, Y; Hoven, NF; Hsieh, YY; Hsu, CT; Hu, CW; Hu, JH; Hu, MY; Hu, Y; Hu, Z; Huang, C; Huang, D; Huang, DQ; Huang, L; Huang, Q; Huang, R; Huang, S; Huang, SC; Huang, W; Huang, Y; Huffman, KM; Hung, CH; Hung, CT; Huurman, R; Hwang, SM; Hyun, S; Ibrahim, AM; Iddi-Faical, A; Immordino, P; Isla, MI; Jacquemond, V; Jacques, T; Jankowska, E; Jansen, JA; Jäntti, T; Jaque-Fernandez, F; Jarvis, GA; Jatt, LP; Jeon, JW; Jeong, SH; Jhunjhunwala, R; Ji, F; Jia, X; Jia, Y; Jian-Bo, Z; Jiang, GD; Jiang, L; Jiang, W; Jiang, WD; Jiang, Z; Jiménez-Hoyos, CA; Jin, S; Jobling, MG; John, CM; John, T; Johnson, CB; Jones, KI; Jones, WS; Joseph, OO; Ju, C; Judeinstein, P; Junges, A; Junnarkar, M; Jurkko, R; Kaleka, CC; Kamath, AV; Kang, X; Kantsadi, AL; Kapoor, M; Karim, Z; Kashuba, ADM; Kassa, E; Kasztura, M; Kataja, A; Katoh, T; Kaufman, JS; Kaupp, M; Kehinde, O; Kehrenberg, C; Kemper, N; Kerr, CW; Khan, AU; Khan, MF; Khan, ZUH; Khojasteh, SC; Kilburn, S; Kim, CG; Kim, DU; Kim, DY; Kim, HJ; Kim, J; Kim, OH; Kim, YH; King, C; Klein, A; Klingler, L; Knapp, AK; Ko, TK; Kodavanti, UP; Kolla, V; Kong, L; Kong, RY; Kong, X; Kore, S; Kortz, U; Korucu, B; Kovacs, A; Krahnert, I; Kraus, WE; Kuang, SY; Kuehn-Hajder, JE; Kurz, M; Kuśtrowski, P; Kwak, YD; Kyttaris, VC; Laga, SM; Laguerre, A; Laloo, A; Langaro, MC; Langham, MC; Lao, X; Larocca, MC; Lassus, J; Lattimer, TA; Lazar, S; Le, MH; Leal, DB; Leal, M; Leary, A; Ledermann, JA; Lee, JF; Lee, MV; Lee, NH; Leeds, CM; Leeds, JS; Lefrandt, JD; Leicht, AS; Leonard, M; Lev, S; Levy, K; Li, B; Li, C; Li, CM; Li, DH; Li, H; Li, J; Li, L; Li, LJ; Li, N; Li, P; Li, T; Li, X; Li, XH; Li, XQ; Li, XX; Li, Y; Li, Z; Li, ZY; Liao, YF; Lin, CC; Lin, MH; Lin, Y; Ling, Y; Links, TP; Lira-Romero, E; Liu, C; Liu, D; Liu, H; Liu, J; Liu, L; Liu, LP; Liu, M; Liu, T; Liu, W; Liu, X; Liu, XH; Liu, Y; Liuwantara, D; Ljumanovic, N; Lobo, L; Lokhande, K; Lopes, A; Lopes, RMRM; López-Gutiérrez, JC; López-Muñoz, MJ; López-Santamaría, M; Lorenzo, C; Lorusso, D; Losito, I; Lu, C; Lu, H; Lu, HZ; Lu, SH; Lu, SN; Lu, Y; Lu, ZY; Luboga, F; Luo, JJ; Luo, KL; Luo, Y; Lutomski, CA; Lv, W; M Piedade, MF; Ma, J; Ma, JQ; Ma, JX; Ma, N; Ma, P; Ma, S; Maciel, M; Madureira, M; Maganaris, C; Maginn, EJ; Mahnashi, MH; Maierhofer, M; Majetschak, M; Malla, TR; Maloney, L; Mann, DL; Mansuri, A; Marelli, E; Margulis, CJ; Marrella, A; Martin, BL; Martín-Francés, L; Martínez de Pinillos, M; Martínez-Navarro, EM; Martinez-Quintanilla Jimenez, D; Martínez-Velasco, A; Martínez-Villaseñor, L; Martinón-Torres, M; Martins, BA; Massongo, M; Mathew, AP; Mathews, D; Matsui, J; Matsumoto, KI; Mau, T; Maves, RC; Mayclin, SJ; Mayer, JM; Maynard, ND; Mayr, T; Mboowa, MG; McEvoy, MP; McIntyre, RC; McKay, JA; McPhail, MJW; McVeigh, AL; Mebazaa, A; Medici, V; Medina, DN; Mehmood, T; Mei-Li, C; Melku, M; Meloncelli, S; Mendes, GC; Mendoza-Velásquez, C; Mercadante, R; Mercado, MI; Merenda, MEZ; Meunier, J; Mi, SL; Michels, M; Mijatovic, V; Mikhailov, V; Milheiro, SA; Miller, DC; Ming, F; Mitsuishi, M; Miyashita, T; Mo, J; Mo, S; Modesto-Mata, M; Moeller, S; Monte, A; Monteiro, L; Montomoli, J; Moore, EE; Moore, HB; Moore, PK; Mor, MK; Moratalla-López, N; Moratilla Lapeña, L; Moreira, R; Moreno, MA; Mörk, AC; Morton, M; Mosier, JM; Mou, LH; Mougharbel, AS; Muccillo-Baisch, AL; Muñoz-Serrano, AJ; Mustafa, B; Nair, GM; Nakanishi, I; Nakanjako, D; Naraparaju, K; Nawani, N; Neffati, R; Neil, EC; Neilipovitz, D; Neira-Borrajo, I; Nelson, MT; Nery, PB; Nese, M; Nguyen, F; Nguyen, MH; Niazy, AA; Nicolaï, J; Nogueira, F; Norbäck, D; Novaretti, JV; O'Donnell, T; O'Dowd, A; O'Malley, DM; Oaknin, A; Ogata, K; Ohkubo, K; Ojha, M; Olaleye, MT; Olawande, B; Olomo, EJ; Ong, EWY; Ono, A; Onwumere, J; Ortiz Bibriesca, DM; Ou, X; Oza, AM; Ozturk, K; Özütemiz, C; Palacio-Pastrana, C; Palaparthi, A; Palevsky, PM; Pan, K; Pantanetti, S; Papachristou, DJ; Pariani, A; Parikh, CR; Parissis, J; Paroul, N; Parry, S; Patel, N; Patel, SM; Patel, VC; Pawar, S; Pefura-Yone, EW; Peixoto Andrade, BCO; Pelepenko, LE; Peña-Lora, D; Peng, S; Pérez-Moro, OS; Perez-Ortiz, AC; Perry, LM; Peter, CM; Phillips, NJ; Phillips, P; Pia Tek, J; Piner, LW; Pinto, EA; Pinto, SN; Piyachaturawat, P; Poka-Mayap, V; Polledri, E; Poloni, TE; Ponessa, G; Poole, ST; Post, AK; Potter, TM; Pressly, BB; Prouty, MG; Prudêncio, M; Pulkki, K; Pupier, C; Qian, H; Qian, ZP; Qiu, Y; Qu, G; Rahimi, S; Rahman, AU; Ramadan, H; Ramanna, S; Ramirez, I; Randolph, GJ; Rasheed, A; Rault, J; Raviprakash, V; Reale, E; Redpath, C; Rema, V; Remucal, CK; Remy, D; Ren, T; Ribeiro, LB; Riboli, G; Richards, J; Rieger, V; Rieusset, J; Riva, A; Rivabella Maknis, T; Robbins, JL; Robinson, CV; Roche-Campo, F; Rodriguez, R; Rodríguez-de-Cía, J; Rollenhagen, JE; Rosen, EP; Rub, D; Rubin, N; Rubin, NT; Ruurda, JP; Saad, O; Sabell, T; Saber, SE; Sabet, M; Sadek, MM; Saejio, A; Salinas, RM; Saliu, IO; Sande, D; Sang, D; Sangenito, LS; Santos, ALSD; Sarmiento Caldas, MC; Sassaroli, S; Sassi, V; Sato, J; Sauaia, A; Saunders, K; Saunders, PR; Savarino, SJ; Scambia, G; Scanlon, N; Schetinger, MR; Schinkel, AFL; Schladweiler, MC; Schofield, CJ; Schuepbach, RA; Schulz, J; Schwartz, N; Scorcella, C; Seeley, J; Seemann, F; Seinige, D; Sengoku, T; Seravalli, J; Sgromo, B; Shaheen, MY; Shan, L; Shanmugam, S; Shao, H; Sharma, S; Shaw, KJ; Shen, BQ; Shen, CH; Shen, P; Shen, S; Shen, Y; Shen, Z; Shi, J; Shi-Li, L; Shimoda, K; Shoji, Y; Shun, C; Silva, MA; Silva-Cardoso, J; Simas, NK; Simirgiotis, MJ; Sincock, SA; Singh, MP; Sionis, A; Siu, J; Sivieri, EM; Sjerps, MJ; Skoczen, SL; Slabon, A; Slette, IJ; Smith, MD; Smith, S; Smith, TG; Snapp, KS; Snow, SJ; Soares, MCF; Soberman, D; Solares, MD; Soliman, I; Song, J; Sorooshian, A; Sorrell, TC; Spinar, J; Staudt, A; Steinhart, C; Stern, ST; Stevens, DM; Stiers, KM; Stimming, U; Su, YG; Subbian, V; Suga, H; Sukhija-Cohen, A; Suksamrarn, A; Suksen, K; Sun, J; Sun, M; Sun, P; Sun, W; Sun, XF; Sun, Y; Sundell, J; Susan, LF; Sutjarit, N; Swamy, KV; Swisher, EM; Sykes, C; Takahashi, JA; Talmor, DS; Tan, B; Tan, ZK; Tang, L; Tang, S; Tanner, JJ; Tanwar, M; Tarazi, Z; Tarvasmäki, T; Tay, FR; Teketel, A; Temitayo, GI; Thersleff, T; Thiessen Philbrook, H; Thompson, LC; Thongon, N; Tian, B; Tian, F; Tian, Q; Timothy, AT; Tingle, MD; Titze, IR; Tolppanen, H; Tong, W; Toyoda, H; Tronconi, L; Tseng, CH; Tu, H; Tu, YJ; Tung, SY; Turpault, S; Tuynman, JB; Uemoto, AT; Ugurlu, M; Ullah, S; Underwood, RS; Ungell, AL; Usandizaga-Elio, I; Vakonakis, I; van Boxel, GI; van den Beucken, JJJP; van der Boom, T; van Slegtenhorst, MA; Vanni, JR; Vaquera, A; Vasconcellos, RS; Velayos, M; Vena, R; Ventura, G; Verso, MG; Vincent, RP; Vitale, F; Vitali, S; Vlek, SL; Vleugels, MPH; Volkmann, N; Vukelic, M; Wagner Mackenzie, B; Wairagala, P; Waller, SB; Wan, J; Wan, MT; Wan, Y; Wang, CC; Wang, H; Wang, J; Wang, JF; Wang, K; Wang, L; Wang, M; Wang, S; Wang, WM; Wang, X; Wang, Y; Wang, YD; Wang, YF; Wang, Z; Wang, ZG; Warriner, K; Weberpals, JI; Weerachayaphorn, J; Wehrli, FW; Wei, J; Wei, KL; Weinheimer, CJ; Weisbord, SD; Wen, S; Wendel Garcia, PD; Williams, JW; Williams, R; Winkler, C; Wirman, AP; Wong, S; Woods, CM; Wu, B; Wu, C; Wu, F; Wu, P; Wu, S; Wu, Y; Wu, YN; Wu, ZH; Wurtzel, JGT; Xia, L; Xia, Z; Xia, ZZ; Xiao, H; Xie, C; Xin, ZM; Xing, Y; Xing, Z; Xu, S; Xu, SB; Xu, T; Xu, X; Xu, Y; Xue, L; Xun, J; Yaffe, MB; Yalew, A; Yamamoto, S; Yan, D; Yan, H; Yan, S; Yan, X; Yang, AD; Yang, E; Yang, H; Yang, J; Yang, JL; Yang, K; Yang, M; Yang, P; Yang, Q; Yang, S; Yang, W; Yang, X; Yang, Y; Yao, JC; Yao, WL; Yao, Y; Yaqub, TB; Ye, J; Ye, W; Yen, CW; Yeter, HH; Yin, C; Yip, V; Yong-Yi, J; Yu, HJ; Yu, MF; Yu, S; Yu, W; Yu, WW; Yu, X; Yuan, P; Yuan, Q; Yue, XY; Zaia, AA; Zakhary, SY; Zalwango, F; Zamalloa, A; Zamparo, P; Zampini, IC; Zani, JL; Zeitoun, R; Zeng, N; Zenteno, JC; Zepeda-Palacio, C; Zhai, C; Zhang, B; Zhang, G; Zhang, J; Zhang, K; Zhang, Q; Zhang, R; Zhang, T; Zhang, X; Zhang, Y; Zhang, YY; Zhao, B; Zhao, D; Zhao, G; Zhao, H; Zhao, Q; Zhao, R; Zhao, S; Zhao, T; Zhao, X; Zhao, XA; Zhao, Y; Zhao, Z; Zheng, Z; Zhi-Min, G; Zhou, CL; Zhou, HD; Zhou, J; Zhou, W; Zhou, XQ; Zhou, Z; Zhu, C; Zhu, H; Zhu, L; Zhu, Y; Zitzmann, N; Zou, L; Zou, Y, 2022
)
0.72
" The present strategy for electrochemical sensor have been also showed excellent recovery in synthetic serum samples and tablet dosage form with the recoveries 97."( A sensitive and selective electrochemical sensor based on molecularly imprinted polymer for the assay of teriflunomide.
Armutcu, C; Bellur Atici, E; Cetinkaya, A; Çorman, ME; Ozkan, SA; Uzun, L, 2022
)
0.94
" No significant baseline covariates affected the ponesimod effects and, consequently, dosage adjustments are not warranted by these analyses."( An exposure-response analysis of ponesimod clinical efficacy in a randomized phase III study in patients with relapsing multiple sclerosis.
Burcklen, M; Kracker, H; Olsson Gisleskog, P; Pérez-Ruixo, JJ; Poggesi, I; Sidorenko, T; Valenzuela, B, 2022
)
0.72
"Therapeutic drug monitoring (TDM) is extremely helpful in individualizing dosage regimen of drugs with narrow therapeutic ranges."( Saliva as Blood Alternative in Therapeutic Monitoring of Teriflunomide-Development and Validation of the Novel Analytical Method.
Giebułtowicz, J; Michorowska, S; Raćkowska, E; Sankowski, B; Sikora, M, 2022
)
0.97
" The goal of this work was to develop a population PK model for A771726 and propose an optimal individualized dosing strategy."( Development of a population pharmacokinetic model and optimal dosing regimen of leflunomide in Korean population.
Chae, D; Park, K; Shin, Y, 2023
)
0.91
" For dose optimization, simulating a set of 1000 virtual subjects from the developed model and dividing the subjects into 5 groups with WT of 50, 60, 70, 80, 90 kg, respectively, the optimal dose was explored that achieves the drug concentration most similar to the target, which was defined as the concentration for the 70 kg subject treated with the current standard dosage regimen (the loading dose of 100 mg QD for 3 days, followed by the maintenance dose of 20 mg QD)."( Development of a population pharmacokinetic model and optimal dosing regimen of leflunomide in Korean population.
Chae, D; Park, K; Shin, Y, 2023
)
0.91
"0 years), or under treatment with alemtuzumab (N = 12, 9 females, median time from last dosing 15."( In-depth characterization of long-term humoral and cellular immune responses to COVID-19m-RNA vaccination in multiple sclerosis patients treated with teriflunomide or alemtuzumab.
Achiron, A; Didikin, M; Dolev, M; Dreyer-Alster, S; Falb, R; Gurevich, M; Harari, G; Magalashvili, D; Mandel, M; Menascu, S; Sonis, P; Warszawer, Y, 2023
)
1.11
"The article provides an introduction to the mechanism of action of teriflunomide, reviews the clinical trials conducted on the safety and efficacy of the drug, and the optimal dosing and monitoring strategies."( Teriflunomide: an oral therapy for first-line treatment of children and adolescents living with relapsing-remitting multiple sclerosis.
Comi, G; Costa, GD,
)
1.81
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Roles (5)

RoleDescription
EC 1.3.98.1 [dihydroorotate oxidase (fumarate)] inhibitorAn EC 1.3.98.* (oxidoreductase acting on CH-CH group of donors, with other, known, acceptors) inhibitor that interferes with the action of dihydroorotate oxidase (fumarate), EC 1.3.98.1 (formerly EC 1.3.3.1).
tyrosine kinase inhibitorAny protein kinase inhibitor that interferes with the action of tyrosine kinase.
hepatotoxic agentA role played by a chemical compound exihibiting itself through the ability to induce damage to the liver in animals.
drug metabolitenull
non-steroidal anti-inflammatory drugAn anti-inflammatory drug that is not a steroid. In addition to anti-inflammatory actions, non-steroidal anti-inflammatory drugs have analgesic, antipyretic, and platelet-inhibitory actions. They act by blocking the synthesis of prostaglandins by inhibiting cyclooxygenase, which converts arachidonic acid to cyclic endoperoxides, precursors of prostaglandins.
[role information is derived from Chemical Entities of Biological Interest (ChEBI), Hastings J, Owen G, Dekker A, Ennis M, Kale N, Muthukrishnan V, Turner S, Swainston N, Mendes P, Steinbeck C. (2016). ChEBI in 2016: Improved services and an expanding collection of metabolites. Nucleic Acids Res]

Drug Classes (6)

ClassDescription
nitrileA compound having the structure RC#N; thus a C-substituted derivative of hydrocyanic acid, HC#N. In systematic nomenclature, the suffix nitrile denotes the triply bound #N atom, not the carbon atom attached to it.
enolAlkenols; the term refers specifically to vinylic alcohols, which have the structure HOCR'=CR2. Enols are tautomeric with aldehydes (R' = H) or ketones (R' =/= H).
aromatic amideAn amide in which the amide linkage is bonded directly to an aromatic system.
enamideAn alpha,beta-unsaturated carboxylic acid amide of general formula R(1)R(2)C=CR(3)-C(=O)NR(4)R(5) in which the amide C=O function is conjugated to a C=C double bond at the alpha,beta position.
(trifluoromethyl)benzenesAn organofluorine compound that is (trifluoromethyl)benzene and derivatives arising from substitution of one or more of the phenyl hydrogens.
secondary carboxamideA carboxamide resulting from the formal condensation of a carboxylic acid with a primary amine; formula RC(=O)NHR(1).
[compound class information is derived from Chemical Entities of Biological Interest (ChEBI), Hastings J, Owen G, Dekker A, Ennis M, Kale N, Muthukrishnan V, Turner S, Swainston N, Mendes P, Steinbeck C. (2016). ChEBI in 2016: Improved services and an expanding collection of metabolites. Nucleic Acids Res]

Pathways (1)

PathwayProteinsCompounds
Pyrimidine synthesis and deprivation pathway (COVID-19 Disease Maps)1329

Protein Targets (12)

Potency Measurements

ProteinTaxonomyMeasurementAverage (µ)Min (ref.)Avg (ref.)Max (ref.)Bioassay(s)
PPM1D proteinHomo sapiens (human)Potency18.55690.00529.466132.9993AID1347411
Interferon betaHomo sapiens (human)Potency18.55690.00339.158239.8107AID1347411
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Inhibition Measurements

ProteinTaxonomyMeasurementAverageMin (ref.)Avg (ref.)Max (ref.)Bioassay(s)
Dihydroorotate dehydrogenase Schistosoma mansoniIC50 (µMol)50.00000.01901.94088.8000AID1592257; AID1604306
Dihydroorotate dehydrogenase (quinone), mitochondrialMus musculus (house mouse)IC50 (µMol)0.12130.03000.10300.1560AID1152593; AID1648546; AID254751; AID55726
Dihydrofolate reductaseHomo sapiens (human)IC50 (µMol)0.38800.00060.87267.3000AID1439522
Cytochrome P450 3A4Homo sapiens (human)IC50 (µMol)0.30000.00011.753610.0000AID1648541
Cytochrome P450 3A5Homo sapiens (human)IC50 (µMol)0.30000.00330.70736.2000AID1648541
Dihydroorotate dehydrogenase (quinone), mitochondrialHomo sapiens (human)IC50 (µMol)0.62950.00050.742710.0000AID1152596; AID1152602; AID1196386; AID1196770; AID1233660; AID1304357; AID1505686; AID1592258; AID1604292; AID1648541; AID1773961; AID1801348; AID1821365; AID1884238; AID254980; AID275414; AID354303; AID634769; AID706970; AID774130
Dihydroorotate dehydrogenase (quinone), mitochondrialHomo sapiens (human)Ki0.92070.01200.50372.7000AID275414; AID354303; AID55745
Stromal interaction molecule 1Homo sapiens (human)IC50 (µMol)4.30000.50002.78334.3000AID1683722
Dihydroorotate dehydrogenase Plasmodium falciparum (malaria parasite P. falciparum)IC50 (µMol)98.87250.23002.64439.4800AID1152595; AID275413; AID354302; AID706969
Dihydroorotate dehydrogenase Plasmodium falciparum (malaria parasite P. falciparum)Ki22.67500.03000.03000.0300AID275413; AID354302
Dihydroorotate dehydrogenase (quinone), mitochondrialRattus norvegicus (Norway rat)IC50 (µMol)0.01720.00900.14100.7000AID1152597; AID1648547; AID254736; AID55727; AID634771; AID644163
Calcium release-activated calcium channel protein 1Homo sapiens (human)IC50 (µMol)4.30000.50004.25009.8000AID1683722
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Activation Measurements

ProteinTaxonomyMeasurementAverageMin (ref.)Avg (ref.)Max (ref.)Bioassay(s)
Dihydroorotate dehydrogenase (quinone), mitochondrialHomo sapiens (human)EC50 (µMol)8.90630.05494.68438.9125AID1687332
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Biological Processes (81)

Processvia Protein(s)Taxonomy
tetrahydrobiopterin biosynthetic processDihydrofolate reductaseHomo sapiens (human)
one-carbon metabolic processDihydrofolate reductaseHomo sapiens (human)
negative regulation of translationDihydrofolate reductaseHomo sapiens (human)
axon regenerationDihydrofolate reductaseHomo sapiens (human)
response to methotrexateDihydrofolate reductaseHomo sapiens (human)
dihydrofolate metabolic processDihydrofolate reductaseHomo sapiens (human)
tetrahydrofolate metabolic processDihydrofolate reductaseHomo sapiens (human)
tetrahydrofolate biosynthetic processDihydrofolate reductaseHomo sapiens (human)
folic acid metabolic processDihydrofolate reductaseHomo sapiens (human)
positive regulation of nitric-oxide synthase activityDihydrofolate reductaseHomo sapiens (human)
regulation of removal of superoxide radicalsDihydrofolate reductaseHomo sapiens (human)
cell surface receptor signaling pathway via JAK-STATInterferon betaHomo sapiens (human)
response to exogenous dsRNAInterferon betaHomo sapiens (human)
B cell activation involved in immune responseInterferon betaHomo sapiens (human)
cell surface receptor signaling pathwayInterferon betaHomo sapiens (human)
cell surface receptor signaling pathway via JAK-STATInterferon betaHomo sapiens (human)
response to virusInterferon betaHomo sapiens (human)
positive regulation of autophagyInterferon betaHomo sapiens (human)
cytokine-mediated signaling pathwayInterferon betaHomo sapiens (human)
natural killer cell activationInterferon betaHomo sapiens (human)
positive regulation of peptidyl-serine phosphorylation of STAT proteinInterferon betaHomo sapiens (human)
cellular response to interferon-betaInterferon betaHomo sapiens (human)
B cell proliferationInterferon betaHomo sapiens (human)
negative regulation of viral genome replicationInterferon betaHomo sapiens (human)
innate immune responseInterferon betaHomo sapiens (human)
positive regulation of innate immune responseInterferon betaHomo sapiens (human)
regulation of MHC class I biosynthetic processInterferon betaHomo sapiens (human)
negative regulation of T cell differentiationInterferon betaHomo sapiens (human)
positive regulation of transcription by RNA polymerase IIInterferon betaHomo sapiens (human)
defense response to virusInterferon betaHomo sapiens (human)
type I interferon-mediated signaling pathwayInterferon betaHomo sapiens (human)
neuron cellular homeostasisInterferon betaHomo sapiens (human)
cellular response to exogenous dsRNAInterferon betaHomo sapiens (human)
cellular response to virusInterferon betaHomo sapiens (human)
negative regulation of Lewy body formationInterferon betaHomo sapiens (human)
negative regulation of T-helper 2 cell cytokine productionInterferon betaHomo sapiens (human)
positive regulation of apoptotic signaling pathwayInterferon betaHomo sapiens (human)
response to exogenous dsRNAInterferon betaHomo sapiens (human)
B cell differentiationInterferon betaHomo sapiens (human)
natural killer cell activation involved in immune responseInterferon betaHomo sapiens (human)
adaptive immune responseInterferon betaHomo sapiens (human)
T cell activation involved in immune responseInterferon betaHomo sapiens (human)
humoral immune responseInterferon betaHomo sapiens (human)
lipid hydroxylationCytochrome P450 3A4Homo sapiens (human)
lipid metabolic processCytochrome P450 3A4Homo sapiens (human)
steroid catabolic processCytochrome P450 3A4Homo sapiens (human)
xenobiotic metabolic processCytochrome P450 3A4Homo sapiens (human)
steroid metabolic processCytochrome P450 3A4Homo sapiens (human)
cholesterol metabolic processCytochrome P450 3A4Homo sapiens (human)
androgen metabolic processCytochrome P450 3A4Homo sapiens (human)
estrogen metabolic processCytochrome P450 3A4Homo sapiens (human)
alkaloid catabolic processCytochrome P450 3A4Homo sapiens (human)
monoterpenoid metabolic processCytochrome P450 3A4Homo sapiens (human)
calcitriol biosynthetic process from calciolCytochrome P450 3A4Homo sapiens (human)
xenobiotic catabolic processCytochrome P450 3A4Homo sapiens (human)
vitamin D metabolic processCytochrome P450 3A4Homo sapiens (human)
vitamin D catabolic processCytochrome P450 3A4Homo sapiens (human)
retinol metabolic processCytochrome P450 3A4Homo sapiens (human)
retinoic acid metabolic processCytochrome P450 3A4Homo sapiens (human)
long-chain fatty acid biosynthetic processCytochrome P450 3A4Homo sapiens (human)
aflatoxin metabolic processCytochrome P450 3A4Homo sapiens (human)
oxidative demethylationCytochrome P450 3A4Homo sapiens (human)
lipid hydroxylationCytochrome P450 3A5Homo sapiens (human)
xenobiotic metabolic processCytochrome P450 3A5Homo sapiens (human)
steroid metabolic processCytochrome P450 3A5Homo sapiens (human)
estrogen metabolic processCytochrome P450 3A5Homo sapiens (human)
alkaloid catabolic processCytochrome P450 3A5Homo sapiens (human)
xenobiotic catabolic processCytochrome P450 3A5Homo sapiens (human)
retinol metabolic processCytochrome P450 3A5Homo sapiens (human)
retinoic acid metabolic processCytochrome P450 3A5Homo sapiens (human)
aflatoxin metabolic processCytochrome P450 3A5Homo sapiens (human)
oxidative demethylationCytochrome P450 3A5Homo sapiens (human)
UDP biosynthetic processDihydroorotate dehydrogenase (quinone), mitochondrialHomo sapiens (human)
'de novo' UMP biosynthetic processDihydroorotate dehydrogenase (quinone), mitochondrialHomo sapiens (human)
pyrimidine ribonucleotide biosynthetic processDihydroorotate dehydrogenase (quinone), mitochondrialHomo sapiens (human)
'de novo' pyrimidine nucleobase biosynthetic processDihydroorotate dehydrogenase (quinone), mitochondrialHomo sapiens (human)
store-operated calcium entryStromal interaction molecule 1Homo sapiens (human)
detection of calcium ionStromal interaction molecule 1Homo sapiens (human)
activation of store-operated calcium channel activityStromal interaction molecule 1Homo sapiens (human)
positive regulation of adenylate cyclase activityStromal interaction molecule 1Homo sapiens (human)
positive regulation of angiogenesisStromal interaction molecule 1Homo sapiens (human)
regulation of calcium ion transportStromal interaction molecule 1Homo sapiens (human)
enamel mineralizationStromal interaction molecule 1Homo sapiens (human)
regulation of store-operated calcium entryStromal interaction molecule 1Homo sapiens (human)
intracellular calcium ion homeostasisStromal interaction molecule 1Homo sapiens (human)
store-operated calcium entryCalcium release-activated calcium channel protein 1Homo sapiens (human)
adaptive immune responseCalcium release-activated calcium channel protein 1Homo sapiens (human)
phospholipase C-activating G protein-coupled receptor signaling pathwayCalcium release-activated calcium channel protein 1Homo sapiens (human)
positive regulation of insulin secretionCalcium release-activated calcium channel protein 1Homo sapiens (human)
positive regulation of adenylate cyclase activityCalcium release-activated calcium channel protein 1Homo sapiens (human)
regulation of calcium ion transportCalcium release-activated calcium channel protein 1Homo sapiens (human)
positive regulation of calcium ion transportCalcium release-activated calcium channel protein 1Homo sapiens (human)
mammary gland epithelium developmentCalcium release-activated calcium channel protein 1Homo sapiens (human)
calcium ion importCalcium release-activated calcium channel protein 1Homo sapiens (human)
calcium ion transmembrane transportCalcium release-activated calcium channel protein 1Homo sapiens (human)
ion channel modulating, G protein-coupled receptor signaling pathwayCalcium release-activated calcium channel protein 1Homo sapiens (human)
ligand-gated ion channel signaling pathwayCalcium release-activated calcium channel protein 1Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Molecular Functions (45)

Processvia Protein(s)Taxonomy
mRNA regulatory element binding translation repressor activityDihydrofolate reductaseHomo sapiens (human)
mRNA bindingDihydrofolate reductaseHomo sapiens (human)
dihydrofolate reductase activityDihydrofolate reductaseHomo sapiens (human)
folic acid bindingDihydrofolate reductaseHomo sapiens (human)
NADPH bindingDihydrofolate reductaseHomo sapiens (human)
sequence-specific mRNA bindingDihydrofolate reductaseHomo sapiens (human)
NADP bindingDihydrofolate reductaseHomo sapiens (human)
cytokine activityInterferon betaHomo sapiens (human)
cytokine receptor bindingInterferon betaHomo sapiens (human)
type I interferon receptor bindingInterferon betaHomo sapiens (human)
protein bindingInterferon betaHomo sapiens (human)
chloramphenicol O-acetyltransferase activityInterferon betaHomo sapiens (human)
monooxygenase activityCytochrome P450 3A4Homo sapiens (human)
steroid bindingCytochrome P450 3A4Homo sapiens (human)
iron ion bindingCytochrome P450 3A4Homo sapiens (human)
protein bindingCytochrome P450 3A4Homo sapiens (human)
steroid hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
retinoic acid 4-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
oxidoreductase activityCytochrome P450 3A4Homo sapiens (human)
oxygen bindingCytochrome P450 3A4Homo sapiens (human)
enzyme bindingCytochrome P450 3A4Homo sapiens (human)
heme bindingCytochrome P450 3A4Homo sapiens (human)
vitamin D3 25-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
caffeine oxidase activityCytochrome P450 3A4Homo sapiens (human)
quinine 3-monooxygenase activityCytochrome P450 3A4Homo sapiens (human)
testosterone 6-beta-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
1-alpha,25-dihydroxyvitamin D3 23-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
anandamide 8,9 epoxidase activityCytochrome P450 3A4Homo sapiens (human)
anandamide 11,12 epoxidase activityCytochrome P450 3A4Homo sapiens (human)
anandamide 14,15 epoxidase activityCytochrome P450 3A4Homo sapiens (human)
aromatase activityCytochrome P450 3A4Homo sapiens (human)
vitamin D 24-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
estrogen 16-alpha-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
estrogen 2-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
1,8-cineole 2-exo-monooxygenase activityCytochrome P450 3A4Homo sapiens (human)
monooxygenase activityCytochrome P450 3A5Homo sapiens (human)
iron ion bindingCytochrome P450 3A5Homo sapiens (human)
protein bindingCytochrome P450 3A5Homo sapiens (human)
retinoic acid 4-hydroxylase activityCytochrome P450 3A5Homo sapiens (human)
oxidoreductase activityCytochrome P450 3A5Homo sapiens (human)
oxygen bindingCytochrome P450 3A5Homo sapiens (human)
heme bindingCytochrome P450 3A5Homo sapiens (human)
aromatase activityCytochrome P450 3A5Homo sapiens (human)
estrogen 16-alpha-hydroxylase activityCytochrome P450 3A5Homo sapiens (human)
testosterone 6-beta-hydroxylase activityCytochrome P450 3A5Homo sapiens (human)
dihydroorotase activityDihydroorotate dehydrogenase (quinone), mitochondrialHomo sapiens (human)
protein bindingDihydroorotate dehydrogenase (quinone), mitochondrialHomo sapiens (human)
dihydroorotate dehydrogenase (quinone) activityDihydroorotate dehydrogenase (quinone), mitochondrialHomo sapiens (human)
dihydroorotate dehydrogenase activityDihydroorotate dehydrogenase (quinone), mitochondrialHomo sapiens (human)
protease bindingStromal interaction molecule 1Homo sapiens (human)
calcium channel regulator activityStromal interaction molecule 1Homo sapiens (human)
calcium ion bindingStromal interaction molecule 1Homo sapiens (human)
protein bindingStromal interaction molecule 1Homo sapiens (human)
identical protein bindingStromal interaction molecule 1Homo sapiens (human)
microtubule plus-end bindingStromal interaction molecule 1Homo sapiens (human)
calcium channel activityCalcium release-activated calcium channel protein 1Homo sapiens (human)
protein bindingCalcium release-activated calcium channel protein 1Homo sapiens (human)
calmodulin bindingCalcium release-activated calcium channel protein 1Homo sapiens (human)
store-operated calcium channel activityCalcium release-activated calcium channel protein 1Homo sapiens (human)
identical protein bindingCalcium release-activated calcium channel protein 1Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Ceullar Components (19)

Processvia Protein(s)Taxonomy
mitochondrionDihydrofolate reductaseHomo sapiens (human)
cytosolDihydrofolate reductaseHomo sapiens (human)
mitochondrionDihydrofolate reductaseHomo sapiens (human)
extracellular spaceInterferon betaHomo sapiens (human)
extracellular regionInterferon betaHomo sapiens (human)
cytoplasmCytochrome P450 3A4Homo sapiens (human)
endoplasmic reticulum membraneCytochrome P450 3A4Homo sapiens (human)
intracellular membrane-bounded organelleCytochrome P450 3A4Homo sapiens (human)
endoplasmic reticulum membraneCytochrome P450 3A5Homo sapiens (human)
intracellular membrane-bounded organelleCytochrome P450 3A5Homo sapiens (human)
nucleoplasmDihydroorotate dehydrogenase (quinone), mitochondrialHomo sapiens (human)
mitochondrionDihydroorotate dehydrogenase (quinone), mitochondrialHomo sapiens (human)
mitochondrial inner membraneDihydroorotate dehydrogenase (quinone), mitochondrialHomo sapiens (human)
cytosolDihydroorotate dehydrogenase (quinone), mitochondrialHomo sapiens (human)
mitochondrial inner membraneDihydroorotate dehydrogenase (quinone), mitochondrialHomo sapiens (human)
endoplasmic reticulumStromal interaction molecule 1Homo sapiens (human)
endoplasmic reticulum membraneStromal interaction molecule 1Homo sapiens (human)
microtubuleStromal interaction molecule 1Homo sapiens (human)
plasma membraneStromal interaction molecule 1Homo sapiens (human)
cortical endoplasmic reticulumStromal interaction molecule 1Homo sapiens (human)
sarcoplasmic reticulum membraneStromal interaction molecule 1Homo sapiens (human)
plasma membrane raftStromal interaction molecule 1Homo sapiens (human)
endoplasmic reticulumStromal interaction molecule 1Homo sapiens (human)
plasma membraneStromal interaction molecule 1Homo sapiens (human)
plasma membraneCalcium release-activated calcium channel protein 1Homo sapiens (human)
membraneCalcium release-activated calcium channel protein 1Homo sapiens (human)
basolateral plasma membraneCalcium release-activated calcium channel protein 1Homo sapiens (human)
plasma membrane raftCalcium release-activated calcium channel protein 1Homo sapiens (human)
membrane raftCalcium release-activated calcium channel protein 1Homo sapiens (human)
calcium channel complexCalcium release-activated calcium channel protein 1Homo sapiens (human)
membraneCalcium release-activated calcium channel protein 1Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Bioassays (108)

Assay IDTitleYearJournalArticle
AID1347411qHTS to identify inhibitors of the type 1 interferon - major histocompatibility complex class I in skeletal muscle: primary screen against the NCATS Mechanism Interrogation Plate v5.0 (MIPE) Libary2020ACS chemical biology, 07-17, Volume: 15, Issue:7
High-Throughput Screening to Identify Inhibitors of the Type I Interferon-Major Histocompatibility Complex Class I Pathway in Skeletal Muscle.
AID1592264Selectivity Index, ratio of IC50 for Schistosoma mansoni DHODH to IC50 for human DHODH to IC502019European journal of medicinal chemistry, Apr-01, Volume: 167Ligand-based design, synthesis and biochemical evaluation of potent and selective inhibitors of Schistosoma mansoni dihydroorotate dehydrogenase.
AID1505687Antiproliferative activity against human Jurkat T cells assessed as DNA content after 72 hrs by Hoechst 33258 dye-based fluorescence assay2018Journal of medicinal chemistry, Jul-26, Volume: 61, Issue:14
Targeting Myeloid Differentiation Using Potent 2-Hydroxypyrazolo[1,5- a]pyridine Scaffold-Based Human Dihydroorotate Dehydrogenase Inhibitors.
AID1196386Inhibition of His-tagged human DHODH assessed as reduction of DCIP by spectrophotometry2015Journal of medicinal chemistry, Jan-22, Volume: 58, Issue:2
Original 2-(3-alkoxy-1H-pyrazol-1-yl)pyrimidine derivatives as inhibitors of human dihydroorotate dehydrogenase (DHODH).
AID275414Inhibition of human recombinant DHODH2007Journal of medicinal chemistry, Jan-25, Volume: 50, Issue:2
Design and synthesis of potent inhibitors of the malaria parasite dihydroorotate dehydrogenase.
AID1196381Antiviral activity against Measles virus expressing firefly luciferase rMV2/Luc infected in human HEK293 cells assessed as bioluminescence level after 6 mins by luminometry2015Journal of medicinal chemistry, Jan-22, Volume: 58, Issue:2
Original 2-(3-alkoxy-1H-pyrazol-1-yl)pyrimidine derivatives as inhibitors of human dihydroorotate dehydrogenase (DHODH).
AID1152605Inhibition of C-terminal His6-tagged human DHODH (amino acid residues 30 to 396) R136A mutant expressed in Escherichia coli BL21 cells assessed as orotic acid production using dihydroorotate as substrate by direct assay in presence of oxygen depleting sys2014Journal of medicinal chemistry, Jun-26, Volume: 57, Issue:12
Fluorine modulates species selectivity in the triazolopyrimidine class of Plasmodium falciparum dihydroorotate dehydrogenase inhibitors.
AID354302Inhibition of ubiquinone binding site of Plasmodium falciparum dihydroorotate dehydrogenase2009Journal of medicinal chemistry, May-14, Volume: 52, Issue:9
Structure-based design, synthesis, and characterization of inhibitors of human and Plasmodium falciparum dihydroorotate dehydrogenases.
AID1884236Inhibition of human N-terminal SUMO-tagged DHODH expressed in Escherichia coli BL21(DE3) at 1 uM using dihydroorotate as substrate by DCIP based microplate reader analysis relative to control2022European journal of medicinal chemistry, Aug-05, Volume: 238A novel series of teriflunomide derivatives as orally active inhibitors of human dihydroorotate dehydrogenase for the treatment of colorectal carcinoma.
AID254751Inhibitory activity against mouse dihydroorotate dehydrogenase2005Bioorganic & medicinal chemistry letters, Nov-01, Volume: 15, Issue:21
SAR, species specificity, and cellular activity of cyclopentene dicarboxylic acid amides as DHODH inhibitors.
AID695196Antiarthritic activity in rat with several spinal cord injury assessed as increase in locomotory function2012ACS medicinal chemistry letters, Oct-11, Volume: 3, Issue:10
Novel use of leflunomide and malononitrilamides: patent highlight.
AID634770Half life in rat at 1 mg/kg, iv2011Bioorganic & medicinal chemistry letters, Dec-15, Volume: 21, Issue:24
Biaryl analogues of teriflunomide as potent DHODH inhibitors.
AID254736Inhibitory activity against rat dihydroorotate dehydrogenase2005Bioorganic & medicinal chemistry letters, Nov-01, Volume: 15, Issue:21
SAR, species specificity, and cellular activity of cyclopentene dicarboxylic acid amides as DHODH inhibitors.
AID1592258Inhibition of human DHODH using DHO as substrate measured at 4 secs interval for 60 secs by DCIP reduction based indirect assay2019European journal of medicinal chemistry, Apr-01, Volume: 167Ligand-based design, synthesis and biochemical evaluation of potent and selective inhibitors of Schistosoma mansoni dihydroorotate dehydrogenase.
AID1648547Inhibition of C-terminal His6-tagged rat DHODH expressed in Escherichia coli BL21(DE3) cells using L-DHO as substrate by DCIP dye based assay2020Journal of medicinal chemistry, 05-14, Volume: 63, Issue:9
Lead Optimization of a Pyrrole-Based Dihydroorotate Dehydrogenase Inhibitor Series for the Treatment of Malaria.
AID567061Chemical stability of the compound in phosphate buffer at pH 7.4 assessed as 3-monosubstituted 1,2,5-oxadiazole level at 100 uM after 24 hrs by RP-HPLC analysis2011European journal of medicinal chemistry, Jan, Volume: 46, Issue:1
1,2,5-Oxadiazole analogues of leflunomide and related compounds.
AID1439522Inhibition of recombinant human N-terminal truncated GST-tagged DHFR (31 to 395 residues) expressed in Escherichia coli BL21(DE3) pyrD using DHO as substrate preincubated for 5 mins followed by substrate addition measured for 5 mins by DCIP oxidation base
AID370563Inhibition of Escherichia coli D-alanine-D-alanine ligase B assessed as residual activity at 2 mM preincubated for 30 mins by malachite green assay2009Bioorganic & medicinal chemistry, Feb-01, Volume: 17, Issue:3
ATP competitive inhibitors of D-alanine-D-alanine ligase based on protein kinase inhibitor scaffolds.
AID1439528Immunosuppressive activity against human PBMC assessed as inhibition of PHA-stimulated cell proliferation preincubated for 2 hrs followed by PHA stimulation for 72 hrs by BrdU incorporation assay
AID644163Inhibition of dihydroorotate dehydrogenase in Wistar rat liver mitochondrial/microsomal membranes measured for 5 mins by 2,6-dichlorophenolindophenol reduction-based spectrophotometry2012European journal of medicinal chemistry, Mar, Volume: 49New inhibitors of dihydroorotate dehydrogenase (DHODH) based on the 4-hydroxy-1,2,5-oxadiazol-3-yl (hydroxyfurazanyl) scaffold.
AID774130Inhibition of N-terminal His10-tagged human DHODH (Met30 to Arg396) expressed in Escherichia coli BL21 (DE3) using dihydroorotate as substrate measured every 30 secs for 6 mins by DCIP dye-based assay2013Journal of medicinal chemistry, Oct-24, Volume: 56, Issue:20
Novel selective and potent inhibitors of malaria parasite dihydroorotate dehydrogenase: discovery and optimization of dihydrothiophenone derivatives.
AID275415Selectivity for Plasmodium falciparum recombinant DHODH over human recombinant DHODH2007Journal of medicinal chemistry, Jan-25, Volume: 50, Issue:2
Design and synthesis of potent inhibitors of the malaria parasite dihydroorotate dehydrogenase.
AID275413Inhibition of recombinant Plasmodium falciparum DHODH2007Journal of medicinal chemistry, Jan-25, Volume: 50, Issue:2
Design and synthesis of potent inhibitors of the malaria parasite dihydroorotate dehydrogenase.
AID1152604Inhibition of C-terminal His6-tagged human DHODH (amino acid residues 30 to 396) H56A mutant expressed in Escherichia coli BL21 cells assessed as orotic acid production using dihydroorotate as substrate by direct assay in presence of oxygen depleting syst2014Journal of medicinal chemistry, Jun-26, Volume: 57, Issue:12
Fluorine modulates species selectivity in the triazolopyrimidine class of Plasmodium falciparum dihydroorotate dehydrogenase inhibitors.
AID155445In vitro inhibition of lymphocyte activated CD25 antigens induced by monoclonal antibody1997Journal of medicinal chemistry, Jun-20, Volume: 40, Issue:13
A new rational hypothesis for the pharmacophore of the active metabolite of leflunomide, a potent immunosuppressive drug.
AID634774Metabolic stability in human microsomes assessed as compound degradation at 5 uM after 30 mins2011Bioorganic & medicinal chemistry letters, Dec-15, Volume: 21, Issue:24
Biaryl analogues of teriflunomide as potent DHODH inhibitors.
AID1233659Antiviral activity against measles virus rMV2/Luc infected in HEK-293T cells assessed as inhibition of virus growth incubated for 24 hrs by firefly luciferase reporter gene assay2015Journal of medicinal chemistry, Jul-23, Volume: 58, Issue:14
Original 2-(3-Alkoxy-1H-pyrazol-1-yl)azines Inhibitors of Human Dihydroorotate Dehydrogenase (DHODH).
AID255000Inhibitory activity (proliferation) against human peripheral blood mononuclear cells stimulated with Phytohemagglutininin-L2005Bioorganic & medicinal chemistry letters, Nov-01, Volume: 15, Issue:21
SAR, species specificity, and cellular activity of cyclopentene dicarboxylic acid amides as DHODH inhibitors.
AID1505691Immunosuppressive activity against human PBMC assessed as inhibition of PHA-stimulated cell proliferation preincubated for 2 hrs followed by PHA stimulation for 72 hrs in presence of exogenous uridine by Hoechst 33258 dye-based fluorescence assay2018Journal of medicinal chemistry, Jul-26, Volume: 61, Issue:14
Targeting Myeloid Differentiation Using Potent 2-Hydroxypyrazolo[1,5- a]pyridine Scaffold-Based Human Dihydroorotate Dehydrogenase Inhibitors.
AID55727Inhibitory concentration tested against enzyme dihydroorotate dehydrogenase in rat1996Journal of medicinal chemistry, Nov-08, Volume: 39, Issue:23
Synthesis, structure-activity relationships, and pharmacokinetic properties of dihydroorotate dehydrogenase inhibitors: 2-cyano-3-cyclopropyl-3-hydroxy-N-[3'-methyl-4'-(trifluoromethyl)phenyl ] propenamide and related compounds.
AID706969Inhibition of Plasmodium falciparum DHODH expressed in Escherichia coli BL21 (DE3) using L-dihydroorotate as substrate after 10 mins by DCIP dye reduction assay2012Journal of medicinal chemistry, Oct-11, Volume: 55, Issue:19
Discovery of diverse human dihydroorotate dehydrogenase inhibitors as immunosuppressive agents by structure-based virtual screening.
AID1152595Inhibition of N-terminal His6-tagged recombinant Plasmodium falciparum DHODH (amino acid residues 158 to 569) expressed in Escherichia coli BL21 cells assessed as orotic acid production using dihydroorotate as substrate by DCIP dye-based assay2014Journal of medicinal chemistry, Jun-26, Volume: 57, Issue:12
Fluorine modulates species selectivity in the triazolopyrimidine class of Plasmodium falciparum dihydroorotate dehydrogenase inhibitors.
AID1152598Inhibition of dog DHODH (amino acid residues 48 to 414) expressed in Escherichia coli BL21 cells assessed as orotic acid production using dihydroorotate as substrate by DCIP dye-based assay2014Journal of medicinal chemistry, Jun-26, Volume: 57, Issue:12
Fluorine modulates species selectivity in the triazolopyrimidine class of Plasmodium falciparum dihydroorotate dehydrogenase inhibitors.
AID1884237Inhibition of human N-terminal SUMO-tagged DHODH expressed in Escherichia coli BL21(DE3) at 100 nM using dihydroorotate as substrate by DCIP based microplate reader analysis relative to control2022European journal of medicinal chemistry, Aug-05, Volume: 238A novel series of teriflunomide derivatives as orally active inhibitors of human dihydroorotate dehydrogenase for the treatment of colorectal carcinoma.
AID1538415Inhibition of C-terminal 6-His tagged recombinant human DHODH N-terminal domain delta29 mutant expressed in Escherichia coli BL21(DE3) using dihydroorotate as substrate measured every 30 secs for 6 mins by DCIP dye based assay2019Journal of medicinal chemistry, 04-25, Volume: 62, Issue:8
Discovery, Optimization, and Target Identification of Novel Potent Broad-Spectrum Antiviral Inhibitors.
AID210279Inhibition of growth of T-cell lymphoblastoma; range 1-10 uM2001Journal of medicinal chemistry, Feb-01, Volume: 44, Issue:3
New dermatological agents for the treatment of psoriasis.
AID1773983Antitumor activity against patient-derived breast cancer cells xenografted in NOD/SCID mouse assessed as tumor growth suppression at 20 mg/kg, ip administered every 2 days and measured after 21 days
AID567060Inhibition of DHODH in Wistar rat liver homogenates at 100 uM by DCIP reduction assay2011European journal of medicinal chemistry, Jan, Volume: 46, Issue:1
1,2,5-Oxadiazole analogues of leflunomide and related compounds.
AID1505695n-Octanol/PBS distribution coefficient, log D of the compound at pH 7.4 after 20 mins by UV-spectrometric based shake-flask method2018Journal of medicinal chemistry, Jul-26, Volume: 61, Issue:14
Targeting Myeloid Differentiation Using Potent 2-Hydroxypyrazolo[1,5- a]pyridine Scaffold-Based Human Dihydroorotate Dehydrogenase Inhibitors.
AID729491Half life in human2013Journal of medicinal chemistry, Apr-25, Volume: 56, Issue:8
On dihydroorotate dehydrogenases and their inhibitors and uses.
AID1152594Antimalarial activity against Plasmodium falciparum 3D7 infected in RBC after 72 hrs by SYBR green dye based flow cytometry2014Journal of medicinal chemistry, Jun-26, Volume: 57, Issue:12
Fluorine modulates species selectivity in the triazolopyrimidine class of Plasmodium falciparum dihydroorotate dehydrogenase inhibitors.
AID275418Inhibition of Plasmodium falciparum DHODH F188A mutant2007Journal of medicinal chemistry, Jan-25, Volume: 50, Issue:2
Design and synthesis of potent inhibitors of the malaria parasite dihydroorotate dehydrogenase.
AID1505693Solubility of the compound in PBS at pH 7.4 at 1 mg after 24 hrs by HPLC-UV analysis2018Journal of medicinal chemistry, Jul-26, Volume: 61, Issue:14
Targeting Myeloid Differentiation Using Potent 2-Hydroxypyrazolo[1,5- a]pyridine Scaffold-Based Human Dihydroorotate Dehydrogenase Inhibitors.
AID1505686Inhibition of recombinant N-terminal GST-tagged human DHODH (31 to 395 residues) expressed in Escherichia coli BL21(DE3) assessed as inhibition of DCIP reduction using dihydroorotate as substrate preincubated for 5 mins followed by substrate addition meas2018Journal of medicinal chemistry, Jul-26, Volume: 61, Issue:14
Targeting Myeloid Differentiation Using Potent 2-Hydroxypyrazolo[1,5- a]pyridine Scaffold-Based Human Dihydroorotate Dehydrogenase Inhibitors.
AID1683722Inhibition of STIM1/Orai1 (unknown origin) expressed in HEK293 cells assessed as reduction in tBHQ-induced calcium response2020Journal of medicinal chemistry, 12-10, Volume: 63, Issue:23
Store-Operated Calcium Entry as a Therapeutic Target in Acute Pancreatitis: Discovery and Development of Drug-Like SOCE Inhibitors.
AID354303Inhibition of ubiquinone binding site of human dihydroorotate dehydrogenase2009Journal of medicinal chemistry, May-14, Volume: 52, Issue:9
Structure-based design, synthesis, and characterization of inhibitors of human and Plasmodium falciparum dihydroorotate dehydrogenases.
AID1233688Inhibition of cell proliferation of human Jurkat cells incubated for 72 hrs by Celltiter-Glo assay2015Journal of medicinal chemistry, Jul-23, Volume: 58, Issue:14
Original 2-(3-Alkoxy-1H-pyrazol-1-yl)azines Inhibitors of Human Dihydroorotate Dehydrogenase (DHODH).
AID449195Antimalarial activity against Plasmodium falciparum W2mef at 10 uM after 48 hrs by hoechst 33342-thiazole orange stain based flow cytometry assay2009Bioorganic & medicinal chemistry letters, Sep-15, Volume: 19, Issue:18
Addressing the malaria drug resistance challenge using flow cytometry to discover new antimalarials.
AID1604292Inhibition of human DHODH2019European journal of medicinal chemistry, Dec-01, Volume: 183Dihydroorotate dehydrogenase inhibitors in anti-infective drug research.
AID1439523Antiproliferative activity against human Jurkat T cells assessed as DNA content after 72 hrs by Hoechst 33258 dye based fluorometric method
AID55745Inhibition of dihydroorotate dehydrogenase2001Journal of medicinal chemistry, Feb-01, Volume: 44, Issue:3
New dermatological agents for the treatment of psoriasis.
AID1439527Antiproliferative activity against human Jurkat T cells assessed as DNA content up to 200 uM after 72 hrs in presence of exogenous uridine by Hoechst 33258 dye based fluorometric method
AID1439529Solubility in pH 7.4 PBS buffer at 1 mg after 24 hrs by HPLC-UV method
AID1773961Inhibition of recombinant N-terminal His-tagged human DHODH (31 to 395 residues) expressed in Escherichia coli using DL-dihydroorotic acid as substrate and Q0 as coenzyme by DCIP based assay
AID1687332Inhibition of DHODH in human HEK-293 cells transfected with ISRE-luciferase reporter gene assessed as increase in IFN-alpha mediated cellular response upto 72 hrs by luciferase reporter gene assay2020European journal of medicinal chemistry, Jan-15, Volume: 186Cerpegin-derived furo[3,4-c]pyridine-3,4(1H,5H)-diones enhance cellular response to interferons by de novo pyrimidine biosynthesis inhibition.
AID1773960Cytotoxicity against human MDA-MB-468 cells assessed as reduction in cell viability incubated for 24 hrs by CellTiter-Glo luminescent assay
AID1884238Inhibition of human N-terminal SUMO-tagged DHODH expressed in Escherichia coli BL21(DE3) using dihydroorotate substrate by DCIP based microplate reader analysis2022European journal of medicinal chemistry, Aug-05, Volume: 238A novel series of teriflunomide derivatives as orally active inhibitors of human dihydroorotate dehydrogenase for the treatment of colorectal carcinoma.
AID1152606Inhibition of C-terminal His6-tagged human DHODH (amino acid residues 30 to 396) L359A mutant expressed in Escherichia coli BL21 cells assessed as orotic acid production using dihydroorotate as substrate by direct assay in presence of oxygen depleting sys2014Journal of medicinal chemistry, Jun-26, Volume: 57, Issue:12
Fluorine modulates species selectivity in the triazolopyrimidine class of Plasmodium falciparum dihydroorotate dehydrogenase inhibitors.
AID706966Immunosuppressive activity in ICR mouse B cells assessed as inhibition of LPS-stimulated cell proliferation after 72 hrs by MTT assay2012Journal of medicinal chemistry, Oct-11, Volume: 55, Issue:19
Discovery of diverse human dihydroorotate dehydrogenase inhibitors as immunosuppressive agents by structure-based virtual screening.
AID706970Inhibition of human DHODH expressed in Escherichia coli BL21(DE3) using L-dihydroorotate as substrate after 10 mins by DCIP dye reduction assay2012Journal of medicinal chemistry, Oct-11, Volume: 55, Issue:19
Discovery of diverse human dihydroorotate dehydrogenase inhibitors as immunosuppressive agents by structure-based virtual screening.
AID634771Inhibition of rat dihydroorotate dehydrogenase2011Bioorganic & medicinal chemistry letters, Dec-15, Volume: 21, Issue:24
Biaryl analogues of teriflunomide as potent DHODH inhibitors.
AID706779Cytotoxicity against human Jurkat cells after 4 days by MTT assay2012Journal of medicinal chemistry, Oct-11, Volume: 55, Issue:19
Discovery of diverse human dihydroorotate dehydrogenase inhibitors as immunosuppressive agents by structure-based virtual screening.
AID377224Inhibition of IL2-dependent mouse CTLL2 cell proliferation after 48 hrs by MTT assay2000Journal of natural products, Sep, Volume: 63, Issue:9
In vitro antiinflammatory effects of neolignan woorenosides from the rhizomes of Coptis japonica.
AID1648541Inhibition of C-terminal His6-tagged human DHODH expressed in Escherichia coli BL21(DE3) cells using L-DHO as substrate by DCIP dye based assay2020Journal of medicinal chemistry, 05-14, Volume: 63, Issue:9
Lead Optimization of a Pyrrole-Based Dihydroorotate Dehydrogenase Inhibitor Series for the Treatment of Malaria.
AID634776Antiinflammatory activity in rat Freund's adjuvant induced-arthritis model assessed as inhibition of paw edema administered 10 days post challenge once daily for 10 days2011Bioorganic & medicinal chemistry letters, Dec-15, Volume: 21, Issue:24
Biaryl analogues of teriflunomide as potent DHODH inhibitors.
AID1439524Cytotoxicity against human Jurkat T cells assessed as compound concentration required to induce up to 30% cell death after 72 hrs by Hoechst 33258 dye based fluorometric method
AID1196770Inhibition of human DHODH using dihydroorotate substrate by DCIP assay2015Journal of medicinal chemistry, Feb-12, Volume: 58, Issue:3
Design, synthesis, X-ray crystallographic analysis, and biological evaluation of thiazole derivatives as potent and selective inhibitors of human dihydroorotate dehydrogenase.
AID1592252Inhibition of Schistosoma mansoni DHODH assessed as remaining enzyme activity at 500 uM using DHO as substrate measured at 4 secs interval for 60 secs by DCIP reduction based indirect assay relative to control2019European journal of medicinal chemistry, Apr-01, Volume: 167Ligand-based design, synthesis and biochemical evaluation of potent and selective inhibitors of Schistosoma mansoni dihydroorotate dehydrogenase.
AID1683728Cytotoxicity in HEK293 cells assessed as cell viability at 50 uM after 72 hrs by MTT assay2020Journal of medicinal chemistry, 12-10, Volume: 63, Issue:23
Store-Operated Calcium Entry as a Therapeutic Target in Acute Pancreatitis: Discovery and Development of Drug-Like SOCE Inhibitors.
AID1233660Inhibition of human recombinant DHODH2015Journal of medicinal chemistry, Jul-23, Volume: 58, Issue:14
Original 2-(3-Alkoxy-1H-pyrazol-1-yl)azines Inhibitors of Human Dihydroorotate Dehydrogenase (DHODH).
AID155444In vitro inhibition of lymphocyte activated CD2 antigens induced by monoclonal antibody1997Journal of medicinal chemistry, Jun-20, Volume: 40, Issue:13
A new rational hypothesis for the pharmacophore of the active metabolite of leflunomide, a potent immunosuppressive drug.
AID155446In vitro inhibition of lymphocyte activated CD71 antigens induced by monoclonal antibody1997Journal of medicinal chemistry, Jun-20, Volume: 40, Issue:13
A new rational hypothesis for the pharmacophore of the active metabolite of leflunomide, a potent immunosuppressive drug.
AID1821365Inhibition of human recombinant DHODH expressed in Escherichia coli BL21 (DE3) using dihydroorotate as substrate and CoQ6 as co-substrate incubated for 10 mins by DCIP based multimode microplate reader analysis
AID634772Antiproliferative activity against PHA-stimulated human PBMC assessed as inhibition of [3H]-thymidine incorporation after 72 hrs by BrdU-ELISA assay2011Bioorganic & medicinal chemistry letters, Dec-15, Volume: 21, Issue:24
Biaryl analogues of teriflunomide as potent DHODH inhibitors.
AID1304356Inhibition of human DHODH assessed as decrease in DCIP at 10 uM using dihydroorotate as substrate measured every 30 seconds for 6 mins2016Bioorganic & medicinal chemistry letters, 07-01, Volume: 26, Issue:13
Design, synthesis and inhibitory activity against human dihydroorotate dehydrogenase (hDHODH) of 1,3-benzoazole derivatives bearing amide units.
AID1152593Inhibition of mouse DHODH (amino acid residues 30 to 396) expressed in Escherichia coli BL21 cells assessed as orotic acid production using dihydroorotate as substrate by DCIP dye-based assay2014Journal of medicinal chemistry, Jun-26, Volume: 57, Issue:12
Fluorine modulates species selectivity in the triazolopyrimidine class of Plasmodium falciparum dihydroorotate dehydrogenase inhibitors.
AID140308The compound was tested for in vitro mixed lymphocyte reaction.1997Journal of medicinal chemistry, Jun-20, Volume: 40, Issue:13
A new rational hypothesis for the pharmacophore of the active metabolite of leflunomide, a potent immunosuppressive drug.
AID1152597Inhibition of rat DHODH (amino acid residues 30 to 396) expressed in Escherichia coli BL21 cells assessed as orotic acid production using dihydroorotate as substrate by DCIP dye-based assay2014Journal of medicinal chemistry, Jun-26, Volume: 57, Issue:12
Fluorine modulates species selectivity in the triazolopyrimidine class of Plasmodium falciparum dihydroorotate dehydrogenase inhibitors.
AID1505694Solubility of the compound in PBS at pH 7.4 at 1 mg after 24 hrs in presence of 2% DMSO by HPLC-UV analysis2018Journal of medicinal chemistry, Jul-26, Volume: 61, Issue:14
Targeting Myeloid Differentiation Using Potent 2-Hydroxypyrazolo[1,5- a]pyridine Scaffold-Based Human Dihydroorotate Dehydrogenase Inhibitors.
AID1152607Ratio of IC50 for C-terminal His6-tagged human DHODH (amino acid residues 30 to 396) R136A mutant expressed in Escherichia coli BL21 cells to IC50 for C-terminal His6-tagged wild type human DHODH (amino acid residues 30 to 396) expressed in Escherichia co2014Journal of medicinal chemistry, Jun-26, Volume: 57, Issue:12
Fluorine modulates species selectivity in the triazolopyrimidine class of Plasmodium falciparum dihydroorotate dehydrogenase inhibitors.
AID634773Metabolic stability in rat microsomes assessed as compound degradation at 5 uM after 30 mins2011Bioorganic & medicinal chemistry letters, Dec-15, Volume: 21, Issue:24
Biaryl analogues of teriflunomide as potent DHODH inhibitors.
AID1592257Inhibition of Schistosoma mansoni DHODH using DHO as substrate measured at 4 secs interval for 60 secs by DCIP reduction based indirect assay2019European journal of medicinal chemistry, Apr-01, Volume: 167Ligand-based design, synthesis and biochemical evaluation of potent and selective inhibitors of Schistosoma mansoni dihydroorotate dehydrogenase.
AID1304357Inhibition of human DHODH assessed as decrease in DCIP using dihydroorotate as substrate measured every 30 seconds for 6 mins2016Bioorganic & medicinal chemistry letters, 07-01, Volume: 26, Issue:13
Design, synthesis and inhibitory activity against human dihydroorotate dehydrogenase (hDHODH) of 1,3-benzoazole derivatives bearing amide units.
AID377223Inhibition of Concanavalin A-stimulated cell proliferation in mouse splenocytes after 48 hrs by MTT assay2000Journal of natural products, Sep, Volume: 63, Issue:9
In vitro antiinflammatory effects of neolignan woorenosides from the rhizomes of Coptis japonica.
AID1505689Cytotoxicity against human Jurkat T cells assessed as compound concentration required to induce =>30% cell death after 72 hrs by CellTox green assay2018Journal of medicinal chemistry, Jul-26, Volume: 61, Issue:14
Targeting Myeloid Differentiation Using Potent 2-Hydroxypyrazolo[1,5- a]pyridine Scaffold-Based Human Dihydroorotate Dehydrogenase Inhibitors.
AID1821363Inhibition of human GAL4-fused RORgammat LBD transfected in HEK293T cell measured after 24 hrs by by dual-glo luciferase assay
AID1505688Antiproliferative activity against human Jurkat T cells assessed as DNA content after 72 hrs in presence of exogenous uridine by Hoechst 33258 dye-based fluorescence assay2018Journal of medicinal chemistry, Jul-26, Volume: 61, Issue:14
Targeting Myeloid Differentiation Using Potent 2-Hydroxypyrazolo[1,5- a]pyridine Scaffold-Based Human Dihydroorotate Dehydrogenase Inhibitors.
AID254980Inhibition of N-terminally truncated recombinant human dihydroorotate dehydrogenase using in vitro enzyme assay2005Bioorganic & medicinal chemistry letters, Nov-01, Volume: 15, Issue:21
SAR, species specificity, and cellular activity of cyclopentene dicarboxylic acid amides as DHODH inhibitors.
AID55726Inhibitory concentration tested on enzyme dihydroorotate dehydrogenase in mouse1996Journal of medicinal chemistry, Nov-08, Volume: 39, Issue:23
Synthesis, structure-activity relationships, and pharmacokinetic properties of dihydroorotate dehydrogenase inhibitors: 2-cyano-3-cyclopropyl-3-hydroxy-N-[3'-methyl-4'-(trifluoromethyl)phenyl ] propenamide and related compounds.
AID1439530Stability in human Jurkat cells assessed as parent compound remaining at 100 uM after 72 hrs by HPLC-UV method
AID695197Antiarthritic activity in rat with several spinal cord injury assessed as increase in voiding function2012ACS medicinal chemistry letters, Oct-11, Volume: 3, Issue:10
Novel use of leflunomide and malononitrilamides: patent highlight.
AID567059Inhibition of DHODH in Wistar rat liver homogenates by DCIP reduction assay2011European journal of medicinal chemistry, Jan, Volume: 46, Issue:1
1,2,5-Oxadiazole analogues of leflunomide and related compounds.
AID1604308Selectivity ratio of IC50 for Schistosoma mansoni DHODH to IC50 for human DHODH2019European journal of medicinal chemistry, Dec-01, Volume: 183Dihydroorotate dehydrogenase inhibitors in anti-infective drug research.
AID1648546Inhibition of C-terminal His6-tagged mouse DHODH expressed in Escherichia coli BL21(DE3) cells using L-DHO as substrate by DCIP dye based assay2020Journal of medicinal chemistry, 05-14, Volume: 63, Issue:9
Lead Optimization of a Pyrrole-Based Dihydroorotate Dehydrogenase Inhibitor Series for the Treatment of Malaria.
AID706965Immunosuppressive activity in mouse T cells assessed as inhibition of two way mixed lymphocyte reaction after 4 days by MTT assay2012Journal of medicinal chemistry, Oct-11, Volume: 55, Issue:19
Discovery of diverse human dihydroorotate dehydrogenase inhibitors as immunosuppressive agents by structure-based virtual screening.
AID1604306Inhibition of Schistosoma mansoni DHODH2019European journal of medicinal chemistry, Dec-01, Volume: 183Dihydroorotate dehydrogenase inhibitors in anti-infective drug research.
AID1648548Inhibition of C-terminal His6-tagged dog DHODH expressed in Escherichia coli BL21(DE3) cells using L-DHO as substrate by DCIP dye based assay2020Journal of medicinal chemistry, 05-14, Volume: 63, Issue:9
Lead Optimization of a Pyrrole-Based Dihydroorotate Dehydrogenase Inhibitor Series for the Treatment of Malaria.
AID1773959Cytotoxicity against human MDA-MB-231 cells assessed as reduction in cell viability incubated for 24 hrs by CellTiter-Glo luminescent assay
AID1152596Inhibition of C-terminal His6-tagged human DHODH (amino acid residues 30 to 396) expressed in Escherichia coli BL21 cells assessed as orotic acid production using dihydroorotate as substrate by DCIP dye-based assay2014Journal of medicinal chemistry, Jun-26, Volume: 57, Issue:12
Fluorine modulates species selectivity in the triazolopyrimidine class of Plasmodium falciparum dihydroorotate dehydrogenase inhibitors.
AID634769Inhibition of human dihydroorotate dehydrogenase2011Bioorganic & medicinal chemistry letters, Dec-15, Volume: 21, Issue:24
Biaryl analogues of teriflunomide as potent DHODH inhibitors.
AID1152610Ratio of IC50 for C-terminal His6-tagged human DHODH (amino acid residues 30 to 396) H56A mutant expressed in Escherichia coli BL21 cells to IC50 for C-terminal His6-tagged wild type human DHODH (amino acid residues 30 to 396) expressed in Escherichia col2014Journal of medicinal chemistry, Jun-26, Volume: 57, Issue:12
Fluorine modulates species selectivity in the triazolopyrimidine class of Plasmodium falciparum dihydroorotate dehydrogenase inhibitors.
AID1233689Antiplasmodial activity against Plasmodium falciparum 3D7 assessed as inhibition of parasite growth at 8.66 ug/ml by SYBR Green I dye based assay2015Journal of medicinal chemistry, Jul-23, Volume: 58, Issue:14
Original 2-(3-Alkoxy-1H-pyrazol-1-yl)azines Inhibitors of Human Dihydroorotate Dehydrogenase (DHODH).
AID1152603Inhibition of C-terminal His6-tagged human DHODH (amino acid residues 30 to 396) L46A mutant expressed in Escherichia coli BL21 cells assessed as orotic acid production using dihydroorotate as substrate by direct assay in presence of oxygen depleting syst2014Journal of medicinal chemistry, Jun-26, Volume: 57, Issue:12
Fluorine modulates species selectivity in the triazolopyrimidine class of Plasmodium falciparum dihydroorotate dehydrogenase inhibitors.
AID1152602Inhibition of C-terminal His6-tagged human DHODH (amino acid residues 30 to 396) expressed in Escherichia coli BL21 cells assessed as orotic acid production using dihydroorotate as substrate by direct assay in presence of oxygen depleting system2014Journal of medicinal chemistry, Jun-26, Volume: 57, Issue:12
Fluorine modulates species selectivity in the triazolopyrimidine class of Plasmodium falciparum dihydroorotate dehydrogenase inhibitors.
AID1801348DCIP-linked Assay from Article 10.1111/cbdd.12530: \\Novel Diketopiperazine Dihydroorotate Dehydrogenase Inhibitors Purified from Traditional Tibetan Animal Medicine Osteon Myospalacem Baileyi.\\2015Chemical biology & drug design, Oct, Volume: 86, Issue:4
Novel Diketopiperazine Dihydroorotate Dehydrogenase Inhibitors Purified from Traditional Tibetan Animal Medicine Osteon Myospalacem Baileyi.
AID1794808Fluorescence-based screening to identify small molecule inhibitors of Plasmodium falciparum apicoplast DNA polymerase (Pf-apPOL).2014Journal of biomolecular screening, Jul, Volume: 19, Issue:6
A High-Throughput Assay to Identify Inhibitors of the Apicoplast DNA Polymerase from Plasmodium falciparum.
AID1794808Fluorescence-based screening to identify small molecule inhibitors of Plasmodium falciparum apicoplast DNA polymerase (Pf-apPOL).
AID1346140Human dihydroorotate dehydrogenase (quinone) (Nucleoside synthesis and metabolism)2007Journal of medicinal chemistry, Jan-25, Volume: 50, Issue:2
Design and synthesis of potent inhibitors of the malaria parasite dihydroorotate dehydrogenase.
[information is prepared from bioassay data collected from National Library of Medicine (NLM), extracted Dec-2023]

Research

Studies (549)

TimeframeStudies, This Drug (%)All Drugs %
pre-19900 (0.00)18.7374
1990's42 (7.65)18.2507
2000's76 (13.84)29.6817
2010's269 (49.00)24.3611
2020's162 (29.51)2.80
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Market Indicators

Research Demand Index: 66.94

According to the monthly volume, diversity, and competition of internet searches for this compound, as well the volume and growth of publications, there is estimated to be very strong demand-to-supply ratio for research on this compound.

MetricThis Compound (vs All)
Research Demand Index66.94 (24.57)
Research Supply Index6.42 (2.92)
Research Growth Index5.18 (4.65)
Search Engine Demand Index115.06 (26.88)
Search Engine Supply Index2.00 (0.95)

This Compound (66.94)

All Compounds (24.57)

Study Types

Publication TypeThis drug (%)All Drugs (%)
Trials50 (8.91%)5.53%
Reviews101 (18.00%)6.00%
Case Studies37 (6.60%)4.05%
Observational17 (3.03%)0.25%
Other356 (63.46%)84.16%
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Clinical Trials (56)

Trial Overview

TrialPhaseEnrollmentStudy TypeStart DateStatus
An Open-label Pharmacokinetic and Tolerability Study of Teriflunomide Given as a Single 14 mg Dose in Subjects With Severe Renal Impairment, and in Matched Subjects With Normal Renal Function [NCT01239459]Phase 116 participants (Actual)Interventional2010-11-30Completed
Open-Label Rater-Blind Randomized Multi-Center Parallel-Arm Active- Comparator Study to Assess the Efficacy and Tolerability of Ofatumumab 20mg SC Monthly vs. First Line DMT - Physician's Choice in the Treatment of Newly Diagnosed RMS [NCT04788615]Phase 3186 participants (Anticipated)Interventional2021-07-23Recruiting
A Randomized, Double-blind, Double-dummy, Parallel-group Study Comparing the Efficacy and Safety of Ofatumumab Versus Teriflunomide in Patients With Relapsing Multiple Sclerosis [NCT02792218]Phase 3930 participants (Actual)Interventional2016-09-20Completed
Multicenter, Randomized, Double-blind, Parallel-group, Active-controlled, Superiority Study to Compare the Efficacy and Safety of Ponesimod to Teriflunomide in Subjects With Relapsing Multiple Sclerosis [NCT02425644]Phase 31,133 participants (Actual)Interventional2015-06-04Completed
Association of Possible Biomarkers With Disease Activity in Patients Treated With Teriflunomide (Aubagio) [NCT03561402]24 participants (Actual)Observational2016-12-01Completed
A Phase III, Multicenter, Randomized, Parallel Group, Double Blind, Double Dummy, Active Controlled Study of Evobrutinib Compared With Teriflunomide, in Participants With Relapsing Multiple Sclerosis to Evaluate Efficacy and Safety (evolutionRMS 2) [NCT04338061]Phase 31,124 participants (Actual)Interventional2020-07-02Active, not recruiting
A Randomized, Double-blind, Double-dummy, Parallel-group Study, Comparing the Efficacy and Safety of Remibrutinib Versus Teriflunomide in Participants With Relapsing Multiple Sclerosis, Followed by Extended Treatment With Open-label Remibrutinib [NCT05156281]Phase 3800 participants (Anticipated)Interventional2021-12-13Recruiting
Czech Pharmaco-epidemiological Real World Data Study Focused on Effectiveness of Different Disease Modifying Drugs [NCT05762003]17,478 participants (Actual)Observational2019-01-01Completed
International Multicenter, Randomized, Double-blind, Double-masked, Placebo-controlled Study of the Efficacy and Safety of BCD-132 Using an Active Reference Drug (Teriflunomide) for the Treatment of Patients With Multiple Sclerosis [NCT04056897]Phase 2270 participants (Anticipated)Interventional2019-06-07Active, not recruiting
Effect of Teriflunomide on Cortical Atrophy and Leptomeningeal Inflammation in Multiple Sclerosis: A Retrospective Observational Case-control Pilot Study [NCT03526224]120 participants (Actual)Observational2018-06-14Completed
Mechanistic Studies of Teriflunomide in Relapsing Remitting Multiple Sclerosis: Regulatory B Lymphocytes as Central Mediators of the Therapeutic Effects of Teriflunomide in MS [NCT03464448]30 participants (Actual)Observational2018-04-17Completed
Teriflunomide Observational Effectiveness Study [NCT02490982]106 participants (Actual)Observational2015-11-30Completed
A Randomized, Double-blind, Double-dummy, Parallel-group Study, Comparing the Efficacy and Safety of Remibrutinib Versus Teriflunomide in Participants With Relapsing Multiple Sclerosis, Followed by Extended Treatment With Open-label Remibrutinib [NCT05147220]Phase 3800 participants (Anticipated)Interventional2021-12-16Recruiting
Chi3L1: A Marker of Efficacy of Platform Treatments in Relapsing-onset Multiple Sclerosis: A Prognostic Study on Existing Clinical Data and Biological Samples [NCT04289675]63 participants (Actual)Observational [Patient Registry]2012-01-01Completed
A Phase 3, Randomized, Double-blind Efficacy and Safety Study Comparing SAR442168 to Teriflunomide (Aubagio®) in Participants With Relapsing Forms of Multiple Sclerosis [NCT04410991]Phase 3900 participants (Anticipated)Interventional2020-06-11Active, not recruiting
A Phase 3, Randomized, Double-blind Efficacy and Safety Study Comparing SAR442168 to Teriflunomide (Aubagio®) in Participants With Relapsing Forms of Multiple Sclerosis [NCT04410978]Phase 3900 participants (Anticipated)Interventional2020-06-30Active, not recruiting
Long-term Extension of the Multinational, Double-blind, Placebo Controlled Studies PDY6045 and PDY6046 to Document the Safety of Teriflunomide When Added to Treatment With Interferon-Beta or Glatiramer Acetate in Patients With Multiple Sclerosis With Rela [NCT00811395]Phase 2182 participants (Actual)Interventional2007-10-31Completed
Phase I/II Study of Teriflunomide in HTLV-1 Associated Myelopathy/Tropical Spastic Paraparesis [NCT04799288]Phase 1/Phase 224 participants (Anticipated)Interventional2021-09-24Recruiting
A Randomized, Double-blind, Double-dummy, Parallel-group Study Comparing the Efficacy and Safety of Ofatumumab Versus Teriflunomide in Patients With Relapsing Multiple Sclerosis. [NCT02792231]Phase 3955 participants (Actual)Interventional2016-08-26Completed
A Randomized, Double-Blind, Placebo-Controlled, Parallel Group Design Study to Evaluate the Efficacy and Safety of Teriflunomide in Reducing the Frequency of Relapses and Delaying the Accumulation of Physical Disability in Subjects With Multiple Sclerosis [NCT00134563]Phase 31,088 participants (Actual)Interventional2004-09-30Completed
A Randomized, Multinational, Double-Blind, Placebo-Controlled, Parallel-Group Design Pilot Study to Estimate the Tolerability, Safety, Pharmacokinetics, and Pharmacodynamic Effects of Teriflunomide for 24 Weeks When Added to Treatment With Interferon-beta [NCT00489489]Phase 2118 participants (Actual)Interventional2007-05-31Completed
Long-term Extension of the Multinational, Double-blind, Placebo Controlled Study EFC6049 (HMR1726D/3001) to Document the Safety of Two Doses of Teriflunomide (7 and 14 mg) in Patients With Multiple Sclerosis With Relapses [NCT00803049]Phase 3742 participants (Actual)Interventional2006-10-31Completed
Teriflunomide Pregnancy Outcome Exposure Registry: An OTIS Autoimmune Diseases in Pregnancy Project [NCT03198351]220 participants (Actual)Observational [Patient Registry]2013-04-25Completed
A Phase III Multicenter Randomized, Double-Blind, Double-Dummy, Parallel-Group Study to Evaluate the Efficacy and Safety of Fenebrutinib Compared With Teriflunomide in Adult Patients With Relapsing Multiple Sclerosis [NCT04586010]Phase 3736 participants (Anticipated)Interventional2021-03-17Recruiting
Evaluation of the Relationship Between ABCG2 Mutation and Teriflunomide Exposure and Safety in Chinese RMS Patients Treated With Teriflunomide 14 mg Once Daily for 24 Weeks [NCT04410965]Phase 482 participants (Actual)Interventional2020-05-20Completed
A Multi-center, Randomized, Parallel-group, Rater-blinded Study Comparing the Effectiveness and Safety of Teriflunomide and Interferon Beta-1a in Patients With Relapsing Multiple Sclerosis Plus a Long Term Extension Period [NCT00883337]Phase 3324 participants (Actual)Interventional2009-04-30Completed
A Multi-center Double-blind Parallel-group Placebo-controlled Study of the Efficacy and Safety of Teriflunomide in Patients With Relapsing Multiple Sclerosis [NCT00751881]Phase 31,169 participants (Actual)Interventional2008-08-31Completed
Phase III: UbLiTuximab in Multiple Sclerosis Treatment Effects (ULTIMATE II STUDY) [NCT03277248]Phase 3545 participants (Actual)Interventional2017-08-25Completed
An International, Multicenter, Randomized, Double-Blind, Double-Masked Study of the Efficacy and Safety of BCD-132 (JSC BIOCAD, Russia) Using an Active Reference Drug (Teriflunomide) for the Treatment of Patients With Multiple Sclerosis [NCT05385744]Phase 3336 participants (Anticipated)Interventional2021-04-05Recruiting
Multi-center, Randomized, Double-blinded Study of Teriflunomide® in Radiologically Isolated Syndrome (RIS) The TERIS Study [NCT03122652]Phase 3125 participants (Actual)Interventional2017-09-25Completed
An Open-label, Single-dose Study to Evaluate the Pharmacokinetic Profiles of 14 mg Teriflunomide Tablet in Healthy Chinese Subjects [NCT02046629]Phase 112 participants (Actual)Interventional2014-05-31Completed
A Randomized, Multinational, Double-blind, Placebo-controlled, Parallel-group Design Pilot Study to Estimate the Tolerability, Safety, Pharmacokinetics, and Pharmacodynamic Effects of Teriflunomide for 24 Weeks When Added to Treatment With Glatiramer Acet [NCT00475865]Phase 2123 participants (Actual)Interventional2007-04-30Completed
An International, Multi-center, Randomized, Double-blind, Placebo-controlled, Parallel Group Study to Evaluate the Efficacy and Safety of Two Year Treatment With Teriflunomide 7 mg Once Daily and 14 mg Once Daily Versus Placebo in Patients With a First Cl [NCT00622700]Phase 3618 participants (Actual)Interventional2008-02-29Completed
A Multi-center Double-blind Parallel-group Placebo-controlled Study of the Efficacy and Safety of Teriflunomide in Patients With Relapsing Multiple Sclerosis Who Are Treated With Interferon-beta [NCT01252355]Phase 3534 participants (Actual)Interventional2011-01-31Terminated(stopped due to Sponsor decision to prematurely stop the study, not linked to any safety concern.)
The Safety and Cost-effectiveness of Discontinuing Disease-modifying Therapies in Stable Relapsing - Onset Multiple Sclerosis (DOT-MS): a Randomized Rater-blinded Multicenter Trial. [NCT04260711]130 participants (Anticipated)Interventional2020-07-01Recruiting
A National, Multi-center Study to Evaluate the Safety of Long Term Treatment With Teriflunomide 14 mg Once Daily in Patients With a First Clinical Episode Suggestive of Multiple Sclerosis in a Long-term Extension Period [NCT02587195]Phase 35 participants (Actual)Interventional2015-12-18Completed
Determining the Effectiveness of earLy Intensive Versus Escalation Approaches for the Treatment of Relapsing-Remitting Multiple Sclerosis [NCT03535298]Phase 4800 participants (Anticipated)Interventional2019-01-03Recruiting
Master Protocol of Two Independent, Randomized, Double-blind, Phase 3 Studies Comparing Efficacy and Safety of Frexalimab (SAR441344) to Teriflunomide in Adult Participants With Relapsing Forms of Multiple Sclerosis [NCT06141473]Phase 31,400 participants (Anticipated)Interventional2023-12-13Recruiting
Exploratory Open Label Study to Investigate the Effect of Teriflunomide on Immune Cell Subsets in the Blood of Patients With Relapsing Forms of Multiple Sclerosis [NCT01863888]Phase 370 participants (Actual)Interventional2013-10-31Completed
A Phase II Study of the Safety and Efficacy of Teriflunomide (HMR1726) in Multiple Sclerosis With Relapses [NCT01487096]Phase 2179 participants (Actual)Interventional2001-04-30Completed
A Two Year, Multicenter, Randomized, Double-Blind, Placebo-Controlled, Parallel Group Trial to Evaluate Efficacy, Safety, Tolerability, and Pharmacokinetics of Teriflunomide Administered Orally Once Daily in Pediatric Patients With Relapsing Forms of Mult [NCT02201108]Phase 3166 participants (Actual)Interventional2014-07-16Active, not recruiting
A Pragmatic Trial to Evaluate the Intermediate-term Effects of Early, Aggressive Versus Escalation Therapy in People With Multiple Sclerosis [NCT03500328]900 participants (Anticipated)Interventional2018-05-02Recruiting
Extension of Protocol HMR1726D/2001, A Phase II Study of the Safety and Efficacy of Teriflunomide (HMR1726) in Multiple Sclerosis With Relapses [NCT00228163]Phase 2147 participants (Actual)Interventional2002-01-31Completed
Phase III: UbLiTuximab In Multiple Sclerosis Treatment Effects (ULTIMATE I STUDY) [NCT03277261]Phase 3549 participants (Actual)Interventional2017-09-19Completed
The Combination of Teriflunomide and High-dose Dexamethasone vs High-dose Dexamethasone Alone as First-line Treatment for Newly Diagnosed Adult Primary Immune Thrombocytopenia (ITP): A Prospective, Multicenter, Randomized Trial [NCT06176235]Phase 2132 participants (Anticipated)Interventional2023-12-19Recruiting
Study to Investigate the Immune Response to Influenza Vaccine in Patients With Multiple Sclerosis on Teriflunomide Treatment and Using a Population of Patients With Multiple Sclerosis as a Reference [NCT01403376]Phase 2128 participants (Actual)Interventional2011-09-30Completed
A Multicenter, Open-label, Randomized, Controlled, Phase 2 Trial Comparing the Efficacy and Safety of Teriflunomide Plus Danazol in Patients With Steroid-resistant/Relapse ITP [NCT06176911]Phase 2124 participants (Anticipated)Interventional2023-12-05Recruiting
MAIN STUDY: Switching Relapsing Multiple Sclerosis Patients Treated With Natalizumab at Risk for Progressive Multifocal Leukoencephalopathy to Teriflunomide: Is This Safe and Effective? SUB-STUDY: Analysis of JCV Antibody Index in MS Patients Treated With [NCT01970410]Phase 455 participants (Actual)Interventional2013-10-31Completed
Determination of Teriflunomide Concentration in Serum and Cerebrospinal Fluid From Patients With Multiple Sclerosis Treated With Teriflunomide 14 mg Daily. [NCT04129736]Phase 412 participants (Actual)Interventional2019-10-10Completed
A Phase III Multicenter Randomized, Double-Blind, Double-Dummy, Parallel-Group Study to Evaluate the Efficacy and Safety of Fenebrutinib Compared With Teriflunomide in Adult Patients With Relapsing Multiple Sclerosis [NCT04586023]Phase 3751 participants (Actual)Interventional2021-03-24Active, not recruiting
A Single Arm Phase IV Clinical Trial to Describe the Safety and Efficacy of Teriflunomide in Patients With Relapsing Forms of Multiple Sclerosis [NCT03856619]Phase 4121 participants (Actual)Interventional2019-03-27Completed
A Prospective, Single-Arm, Clinical-Setting Study to Describe Efficacy, Tolerability and Convenience of Teriflunomide Treatment Using Patient Reported Outcomes (PROs) in Relapsing Multiple Sclerosis (RMS) Patients [NCT01895335]Phase 41,001 participants (Actual)Interventional2013-06-30Completed
STATURE: A Prospective Observational Study of the relationShip beTween Oral DMT bURden and adhErence in People With MS [NCT04676204]323 participants (Anticipated)Observational2020-09-25Enrolling by invitation
A Phase IIa, Double-blind, Randomised, Placebo-controlled Study on the Efficacy and Tolerability of a 14-day Treatment With Teriflunomide vs. Placebo in Subjects With Coeliac Disease Undergoing a 3-day Gluten Challenge [NCT04806737]Phase 1/Phase 215 participants (Anticipated)Interventional2021-04-16Recruiting
A Phase III, Multicenter, Randomized, Parallel Group, Double Blind, Double Dummy, Active Controlled Study of Evobrutinib Compared With Teriflunomide, in Participants With Relapsing Multiple Sclerosis to Evaluate Efficacy and Safety (evolutionRMS 1) [NCT04338022]Phase 31,124 participants (Actual)Interventional2020-06-12Active, not recruiting
Rapid Elimination Procedure of Teriflunomide With Colestipol Hydrochloride [NCT02263547]Phase 114 participants (Actual)Interventional2015-03-31Terminated(stopped due to PK time points were not being met as expected. Determination that risks outweighed benefit.)
[information is prepared from clinicaltrials.gov, extracted Sep-2024]

Trial Outcomes

TrialOutcome
NCT00134563 (6) [back to overview]Changes From Baseline in Fatigue Impact Scale [FIS] Total Score
NCT00134563 (6) [back to overview]Annualized Relapse Rate [ARR]: Poisson Regression Estimates
NCT00134563 (6) [back to overview]Cerebral Magnetic Resonance Imaging [MRI] Assessment: Change From Baseline in Total Lesion Volume (Burden of Disease)
NCT00134563 (6) [back to overview]Time to 12-week Sustained Disability Progression: Kaplan-Meier Estimates of the Rate of Disability Progression at Timepoints
NCT00134563 (6) [back to overview]Cerebral MRI Assessment: Volume of Gd-enhancing T1-lesions Per Scan
NCT00134563 (6) [back to overview]Cerebral MRI Assessment: Number of Gd-enhancing T1-lesions Per Scan (Poisson Regression Estimates)
NCT00475865 (8) [back to overview]Cerebral MRI Assessment: Number of Gd-enhancing T1-lesions Per Scan (Poisson Regression Estimates)
NCT00475865 (8) [back to overview]Overview of Adverse Events (AE]
NCT00475865 (8) [back to overview]Cerebral Magnetic Resonance Imaging [MRI] Assessment: Change From Baseline in Total Lesion Volume (Burden of Disease)
NCT00475865 (8) [back to overview]Annualized Relapse Rate [ARR]: Poisson Regression Estimates
NCT00475865 (8) [back to overview]Overview of AE With Potential Risk of Occurrence
NCT00475865 (8) [back to overview]Pharmacokinetic [PK]: Teriflunomide Plasma Concentration
NCT00475865 (8) [back to overview]Liver Function: Number of Participants With Potentially Clinically Significant Abnormalities (PCSA)
NCT00475865 (8) [back to overview]Cerebral MRI Assessment: Volume of Gd-enhancing T1-lesions Per Scan
NCT00489489 (8) [back to overview]Pharmacokinetic [PK]: Teriflunomide Plasma Concentration
NCT00489489 (8) [back to overview]Cerebral MRI Assessment: Volume of Gd-enhancing T1-lesions Per Scan
NCT00489489 (8) [back to overview]Cerebral MRI Assessment: Number of Gd-enhancing T1-lesions Per Scan (Poisson Regression Estimates)
NCT00489489 (8) [back to overview]Cerebral Magnetic Resonance Imaging [MRI] Assessment: Change From Baseline in Total Lesion Volume (Burden of Disease)
NCT00489489 (8) [back to overview]Annualized Relapse Rate [ARR]: Poisson Regression Estimates
NCT00489489 (8) [back to overview]Overview of AE With Potential Risk of Occurrence
NCT00489489 (8) [back to overview]Liver Function: Number of Participants With Potentially Clinically Significant Abnormalities (PCSA)
NCT00489489 (8) [back to overview]Overview of Adverse Events [AE]
NCT00622700 (16) [back to overview]Core Treatment Period: Time to 12-Week Sustained Disability Progression
NCT00622700 (16) [back to overview]Core Treatment Period: Time to Conversion to Clinically Definite Multiple Sclerosis (CDMS)
NCT00622700 (16) [back to overview]Core Treatment Period: Brain Magnetic Resonance Imaging (MRI) Assessment: Change From Baseline in Total Lesion Volume at Week 108
NCT00622700 (16) [back to overview]Core Treatment Period: Brain MRI Assessment: Change From Baseline in Volume of Hypointense Post-Gadolinium T1 Lesion Component
NCT00622700 (16) [back to overview]Core Treatment Period: Annualized Relapse Rate (ARR)
NCT00622700 (16) [back to overview]Core Treatment Period: Brain MRI Assessment: Change From Baseline in Volume of T2 Lesion Component
NCT00622700 (16) [back to overview]Core Treatment Period: Brain MRI Assessment: Number of Gadolinium Enhancing (Gd-enhancing) T1-lesions Per MRI Scan (Poisson Regression Estimates)
NCT00622700 (16) [back to overview]Core Treatment Period: Brain MRI Assessment: Percent Change From Baseline in Atrophy
NCT00622700 (16) [back to overview]Extension Treatment Period: Overview of Adverse Events (AEs)
NCT00622700 (16) [back to overview]Core Treatment Period: Change From Baseline in EDSS at Week 108
NCT00622700 (16) [back to overview]Extension Treatment Period: Time to Conversion to Clinically Definite Multiple Sclerosis (CDMS)
NCT00622700 (16) [back to overview]Core Treatment Period: Brain MRI Assessment: Volume of Gadolinium Enhancing (Gd-enhancing) T1-lesions Per MRI Scan
NCT00622700 (16) [back to overview]Core Treatment Period: Change From Baseline in Fatigue Impact Scale (FIS) Total Score at Week 108
NCT00622700 (16) [back to overview]Core Treatment Period: Time to Conversion to Definite Multiple Sclerosis (DMS)
NCT00622700 (16) [back to overview]Core Treatment Period: Overview of Adverse Events (AEs)
NCT00622700 (16) [back to overview]Core Treatment Period: Liver Function: Number of Participants With Potentially Clinically Significant Abnormalities (PCSA)
NCT00751881 (13) [back to overview]Extension Treatment Period: Overview of Treatment Emergent Adverse Events (TEAE)
NCT00751881 (13) [back to overview]Extension Treatment Period: Time to Disability Progression
NCT00751881 (13) [back to overview]Core Treatment Period: Time to Disability Progression
NCT00751881 (13) [back to overview]Core Treatment Period: Change From Baseline to Week 48 in Fatigue Impact Scale (FIS) Total Score
NCT00751881 (13) [back to overview]Extension Treatment Period: ARR: Poisson Regression Estimate
NCT00751881 (13) [back to overview]Core Treatment Period: Change From Baseline to Last Visit in Short Form Generic Health Survey - 36 Items (SF-36) Summary Scores
NCT00751881 (13) [back to overview]Core Treatment Period: Change From Baseline to Week 48 in Short Form Generic Health Survey - 36 Items (SF-36) Summary Scores
NCT00751881 (13) [back to overview]Core Treatment Period: Liver Function: Number of Participants With Potentially Clinically Significant Abnormalities (PCSA)
NCT00751881 (13) [back to overview]Core Treatment Period: Overview of Adverse Events
NCT00751881 (13) [back to overview]Core Treatment Period: Time Without Relapse
NCT00751881 (13) [back to overview]Core Treatment Period: Change From Baseline to Week 48 in EDSS Total Score
NCT00751881 (13) [back to overview]Core Treatment Period: Annualized Relapse Rate (ARR): Poisson Regression Estimate
NCT00751881 (13) [back to overview]Core Treatment Period: Change From Baseline to Last Visit in Fatigue Impact Scale (FIS) Total Score
NCT00803049 (6) [back to overview]Annualized MS Relapse Rate (ARR): Poisson Regression Estimates
NCT00803049 (6) [back to overview]Magnetic Resonance Imaging (MRI) Assessment: Change From Baseline in Total Volume of Abnormal Lesions (Burden of Disease [BOD]) at Week 192 Since LTS6050 Randomization
NCT00803049 (6) [back to overview]Time to 12 Week Sustained Disability Progression (DP): Kaplan-Meier Estimates of the Rate of DP
NCT00803049 (6) [back to overview]Time to 24 Week Sustained Disability Progression (DP): Kaplan-Meier Estimates of the Rate of DP
NCT00803049 (6) [back to overview]Percentage of Participants Free of Sustained Disability Progression (DP)
NCT00803049 (6) [back to overview]Percentage of Participants With Treatment-Emergent Adverse Events (TEAEs)
NCT00811395 (9) [back to overview]Overview of Adverse Events [AE]
NCT00811395 (9) [back to overview]Overview of AE With Potential Risk of Occurence
NCT00811395 (9) [back to overview]Time to 12-week Sustained Disability Progression: Kaplan-Meier Estimates of the Rate of Disability Progression at Timepoints
NCT00811395 (9) [back to overview]Liver Function: Number of Participants With Potentially Clinically Significant Abnormalities [PCSA]
NCT00811395 (9) [back to overview]Cerebral MRI Assessment: Total Volume of Gd-enhancing T1-lesions Per Scan
NCT00811395 (9) [back to overview]Cerebral MRI Assessment: Number of Gd-enhancing T1-lesions Per Scan (Poisson Regression Estimates)
NCT00811395 (9) [back to overview]Cerebral Magnetic Resonance Imaging [MRI] Assessment: Change From Baseline in Total Lesion Volume (Burden of Disease)
NCT00811395 (9) [back to overview]Annualized Relapse Rate [ARR]: Poisson Regression Estimates
NCT00811395 (9) [back to overview]Overview of 12-week Sustained Disability Progression
NCT00883337 (8) [back to overview]Core Treatment Period: Change From Baseline in Fatigue Impact Scale (FIS) Total Score
NCT00883337 (8) [back to overview]Extension Treatment Period: ARR Poisson Regression Estimates
NCT00883337 (8) [back to overview]Core Treatment Period: Overview of Adverse Events [AE]
NCT00883337 (8) [back to overview]Core Treatment Period: Overview of Failures
NCT00883337 (8) [back to overview]Core Treatment Period: Time to Failure: Kaplan-Meier Estimates of the Rate of Failure at Timepoints
NCT00883337 (8) [back to overview]Core Treatment Period: Treatment Satisfaction Questionnaire for Medication [TSQM] Scores
NCT00883337 (8) [back to overview]Extension Treatment Period: Overview of AEs
NCT00883337 (8) [back to overview]Core Treatment Period: Annualized Relapse Rate [ARR] - Poisson Regression Estimates
NCT01252355 (7) [back to overview]Brain MRI Assessment: Change From Baseline in Total Lesion Volume (Burden of Disease) at Week 24
NCT01252355 (7) [back to overview]Brain MRI Assessment: Volume of Gd-enhancing T1-lesions Per MRI Scan
NCT01252355 (7) [back to overview]Liver Function: Number of Participants With Potentially Clinically Significant Abnormalities (PCSA)
NCT01252355 (7) [back to overview]Overview of Adverse Events (AEs)
NCT01252355 (7) [back to overview]Time to Relapse: Kaplan-Meier Estimates of the Probability of no Relapse at Week 24, 48, and 72
NCT01252355 (7) [back to overview]Brain Magnetic Resonance Imaging (MRI) Assessment: Number of Gadolinium Enhancing (Gd-enhancing) T1-lesions Per Scan (Poisson Regression Estimates)
NCT01252355 (7) [back to overview]Annualized Relapse Rate (ARR) (Poisson Regression Estimates)
NCT01403376 (5) [back to overview]Immunoglobulin Levels
NCT01403376 (5) [back to overview]Geometric Mean of Titers (GMT) Ratio Post/Pre Vaccination
NCT01403376 (5) [back to overview]Percentage of Participants With 2 Fold or More Increase in Antibody Titer at 28 Days Post Vaccination
NCT01403376 (5) [back to overview]Percentage of Participants With 4 Fold or More Increase in Antibody Titer at 28 Days Post Vaccination
NCT01403376 (5) [back to overview]Percentage of Participants With Antibody Titer ≥40 at 28 Days Post Vaccination
NCT01895335 (14) [back to overview]Time to Relapse: Kaplan-Meier Estimates of the Probability of Treated Relapse at Week 4, Week 24 and Week 48
NCT01895335 (14) [back to overview]Annualized Treated Relapse Rate
NCT01895335 (14) [back to overview]Change From Baseline in Cognition Measured by Symbol Digit Modalities Test (SDMT) Score at Week 48
NCT01895335 (14) [back to overview]Change From Baseline in Disease Progression Using Patient Determined Disease Steps (PDDS) Score at Week 48
NCT01895335 (14) [back to overview]Change From Baseline in Multiple Sclerosis International Quality of Life (MusiQoL) Score at Week 48
NCT01895335 (14) [back to overview]Change From Baseline in Stern Leisure Activity Scale at Week 48
NCT01895335 (14) [back to overview]Duration of Teriflunomide Treatment Exposure
NCT01895335 (14) [back to overview]Percentage of Participants With Treatment Compliance of ≥80% During the Study Treatment Period
NCT01895335 (14) [back to overview]Treatment Satisfaction Questionnaire for Medication (TSQM) Version 1.4 - Assessment of Global Satisfaction Subscale Score With Teriflunomide Treatment at Week 48
NCT01895335 (14) [back to overview]Change From Baseline in Multiple Sclerosis Performance Scale (MSPS) Score at Week 24 and Week 48
NCT01895335 (14) [back to overview]Change From Baseline in TSQM Scores in Participants Switching From Another Disease Modifying Therapy (DMT) at Week 4 and Week 48
NCT01895335 (14) [back to overview]Change From Week 4 in TSQM Scores in Naïve Participants to Week 48
NCT01895335 (14) [back to overview]Expanded Disability Status Scale (EDSS) Score at Baseline and Week 48
NCT01895335 (14) [back to overview]Overview of Adverse Events (AEs)
NCT01970410 (5) [back to overview]MAIN STUDY: Number of Participants Relapse Free at 24 Months
NCT01970410 (5) [back to overview]"MAIN STUDY: Time to Return of Radiological Evidence of Multiple Sclerosis Activity With New Gadolinium Enhancing (Gd+) Lesions on Cranial MRI."
NCT01970410 (5) [back to overview]MAIN STUDY: Expanded Disability Status Scale (EDSS) Sustained Progression for 3 Months as Measured by at Least 0.5 Increase From Baseline or 1 in Any EDSS Set Score
NCT01970410 (5) [back to overview]MAIN STUDY: Mean Time to New T2 or Enlarging T2 Hyperintensities on Monthly Sentinel Brain MRIs
NCT01970410 (5) [back to overview]SUB-STUDY: Number of Patients With a Reduction of Anti-JCV Antibody Index (AJAI)
NCT02201108 (22) [back to overview]Cognitive Assessment: Change From Baseline in Trail Making Test- Part A (TMT-A) Test Scores (in Seconds) at Week 96 and 192
NCT02201108 (22) [back to overview]Brain Magnetic Resonance Imaging Assessment: Change From Baseline in Volume of T2 Lesions at Weeks 24, 36, 48, 72, 96, 144 and 192
NCT02201108 (22) [back to overview]Probability of Participants Who Were Clinical Relapse Free at Weeks 24, 48, 72, 96, 120, 144, 168 and 192
NCT02201108 (22) [back to overview]Brain Magnetic Resonance Imaging (MRI) Assessment: Number of New or Enlarged T2 Lesions Per MRI Scan
NCT02201108 (22) [back to overview]Brain Magnetic Resonance Imaging Assessment: Number of New T1 Hypointense Lesions Per MRI Scan
NCT02201108 (22) [back to overview]Brain Magnetic Resonance Imaging Assessment: Number of T1 Gadolinium (Gd)-Enhancing T1 Lesions Per MRI Scan
NCT02201108 (22) [back to overview]DB: Pharmacokinetics: Steady-state Trough Concentration (Ctrough) of Teriflunomide
NCT02201108 (22) [back to overview]OL: Pharmacokinetics: Steady-state Trough Concentration (Ctrough) of Teriflunomide
NCT02201108 (22) [back to overview]OL: Time to First Confirmed Clinical Relapse
NCT02201108 (22) [back to overview]Time to First Confirmed Clinical Relapse
NCT02201108 (22) [back to overview]Brain Magnetic Resonance Imaging Assessment: Change From Baseline in Volume of T1 Hypointense Lesions
NCT02201108 (22) [back to overview]Brain Magnetic Resonance Imaging Assessment: Percent Change From Baseline in Brain Volume at Weeks 24, 36, 48, 72, 96, 144 and 192
NCT02201108 (22) [back to overview]Brain Magnetic Resonance Imaging Assessment: Percentage of Participants Free of New or Enlarged MRI T2-Lesions
NCT02201108 (22) [back to overview]Cognitive Assessment - Selective Reminding Test (SRT): Change From Baseline in Total Number of Words on Delayed Recall at Weeks 96 and 192
NCT02201108 (22) [back to overview]Cognitive Assessment: Change From Baseline in Beery Visual-motor Integration (BVMI) Scores at Weeks 96 and 192
NCT02201108 (22) [back to overview]Cognitive Assessment: Change From Baseline in Brief Visuospatial Memory Test-Revised (BVMT-R) Scores at Weeks 96 and 192
NCT02201108 (22) [back to overview]Cognitive Assessment: Change From Baseline in Delis-Kaplan Executive Function System (D-KEFS) Letter Fluency Total Correct Raw Score at Weeks 96 and 192
NCT02201108 (22) [back to overview]Cognitive Assessment: Change From Baseline in Delis-Kaplan Executive Function System Category Fluency Total Correct Raw Score at Weeks 96 and 192
NCT02201108 (22) [back to overview]Cognitive Assessment: Change From Baseline in Number of Completed Items Measured by Symbol Digit Modalities Test at Weeks 24, 48, 72, 96, 120, 144, 168 and 192
NCT02201108 (22) [back to overview]Cognitive Assessment: Change From Baseline in Total Number of Correct Substitutions Measured by Symbol Digit Modalities Test (SDMT) at Weeks 24, 48, 72, 96, 120, 144, 168 and 192
NCT02201108 (22) [back to overview]Cognitive Assessment: Change From Baseline in Trail Making Test B (TMT-B) Test Scores (in Seconds) at Weeks 96 and 192
NCT02201108 (22) [back to overview]Cognitive Assessment: Change From Baseline in Wechsler Abbreviated Scale of Intelligence-II (WASI-II) Vocabulary Total Raw Scores at Weeks 96 and 192
NCT02263547 (2) [back to overview]Secondary Outcome Measure: Mean Percentage Change of Serum Teriflunomide Levels Percentage Change of Teriflunomide Concentrations at Day 8, Day 14, Day 26 ad Day 36 Following Administration of Colestipol Hydrochloride Tablets.
NCT02263547 (2) [back to overview]Primary Outcome Measures: Teriflunomide Concentrations at Day 28
NCT02425644 (5) [back to overview]Cumulative Number of Combined Unique Active Lesions (CUAL) Per Year From Baseline to Week 108
NCT02425644 (5) [back to overview]24-Week Confirmed Disability Accumulation (CDA) Assessed From Baseline to EOS
NCT02425644 (5) [back to overview]12-Week Confirmed Disability Accumulation (CDA) Assessed From Baseline to EOS
NCT02425644 (5) [back to overview]Change From Baseline in Fatigue-related Symptoms as Measured by the Symptoms Domain of the Fatigue Symptom and Impact Questionnaire-Relapsing Multiple Sclerosis (FSIQ-RMS) Score to Week 108
NCT02425644 (5) [back to overview]Annualized Confirmed Relapse Rate
NCT02792218 (30) [back to overview]Annualized Relapse Rates (ARR) by NfL High-low Subgroups - Pooled Data
NCT02792218 (30) [back to overview]Annual Rate of Percent Change in Brain Volume Loss by NfL High-low Subgroups - Pooled Data
NCT02792218 (30) [back to overview]6-month Confirmed Worsening of at Least 20% in the Timed 25-Foot Walk (T25FW) - Pooled Data
NCT02792218 (30) [back to overview]6-month Confirmed Worsening of at Least 20% in the 9-Hole Peg Test (9HPT) - Pooled Data
NCT02792218 (30) [back to overview]6-month Confirmed Disability Worsening (6mCDW) or 6-month Confirmed Cognitive Decline (6mCCD) - Pooled Data
NCT02792218 (30) [back to overview]6-month Confirmed Disability Worsening (6mCDW) Based on EDSS > 8 Weeks After Onset of Treatment - Pooled Data
NCT02792218 (30) [back to overview]6-month Confirmed Disability Worsening (6mCDW) Based on EDSS - Study COMB157G2301
NCT02792218 (30) [back to overview]3-month Confirmed Disability Worsening (3mCDW) Based on EDSS > 8 Weeks After Onset of Treatment - Pooled Data
NCT02792218 (30) [back to overview]6-month Confirmed Disability Worsening (6mCDW) Based on EDSS - Pooled Data
NCT02792218 (30) [back to overview]6-month Confirmed Disability Improvement (6mCDI) Sustained Until End of Study (EOS) as Measured by EDSS - Pooled Data
NCT02792218 (30) [back to overview]6-month Confirmed Disability Improvement (6mCDI ) Based on EDSS - Study COMB157G2301
NCT02792218 (30) [back to overview]6-month Confirmed Disability Improvement (6mCDI ) Based on EDSS - Pooled Data
NCT02792218 (30) [back to overview]Pharmacokinetic (PK) Concentrations of Ofatumumab
NCT02792218 (30) [back to overview]Multiple Sclerosis Impact Scale (MSIS-29) Physical Impact Score Change From Baseline
NCT02792218 (30) [back to overview]Change in Cognitive Performance Measured by the Symbol Digit Modalities Test (SDMT) - Pooled Data
NCT02792218 (30) [back to overview]6-month Confirmed Cognitive Decline on Symbol Digit Modalities Test (SDMT) - Pooled Data
NCT02792218 (30) [back to overview]Percent Change in T2 Lesion Volume Relative to Baseline
NCT02792218 (30) [back to overview]Number of New or Enlarging T2 Lesions Per Year by NfL High-low Subgroups - Pooled Data
NCT02792218 (30) [back to overview]No Evidence of Disease Activity (NEDA-4)
NCT02792218 (30) [back to overview]Neurofilament Light Chain (NfL) Concentration in Serum
NCT02792218 (30) [back to overview]Multiple Sclerosis Impact Scale (MSIS-29) Psychological Impact Score Change From Baseline
NCT02792218 (30) [back to overview]3-month Confirmed Disability Worsening (3mCDW) Based on EDSS - Study COMB157G2301
NCT02792218 (30) [back to overview]3-month Confirmed Disability Worsening (3mCDW) Based on EDSS - Pooled Data
NCT02792218 (30) [back to overview]Percentage of Participants With Confirmed Relapse
NCT02792218 (30) [back to overview]Number of New or Enlarging T2 Lesions on MRI Per Year From Month 12 Until End of Study (EOS)
NCT02792218 (30) [back to overview]Number of Gd-enhancing T1 Lesions Per MRI Scan
NCT02792218 (30) [back to overview]Annualized Relapse Rate (ARR) >8 Weeks After Onset of Treatment
NCT02792218 (30) [back to overview]Number of New or Enlarging T2 Lesions on MRI Per Year (Annualized Lesion Rate)
NCT02792218 (30) [back to overview]Annualized Relapse Rate (ARR)
NCT02792218 (30) [back to overview]Annualized Rate of Brain Volume Loss Based on Assessments of Percent Brain Volume Change From Baseline
NCT02792231 (30) [back to overview]Number of Gadolinium-enhancing T1 Lesions Per MRI Scan
NCT02792231 (30) [back to overview]Annualized Relapse Rate (ARR) >8 Weeks After Onset of Treatment
NCT02792231 (30) [back to overview]Annualized Relapse Rate (ARR)
NCT02792231 (30) [back to overview]Annualized Rate of Brain Volume Loss Based on Assessments of Percent Brain Volume Change From Baseline
NCT02792231 (30) [back to overview]Number of New or Enlarging T2 Lesions on MRI Per Year From Month 12 Until End of Study (EOS)
NCT02792231 (30) [back to overview]6-month Confirmed Disability Worsening (6mCDW) Based on EDSS - Study COMB157G2302
NCT02792231 (30) [back to overview]6-month Confirmed Disability Worsening (6mCDW) Based on EDSS - Pooled Data
NCT02792231 (30) [back to overview]6-month Confirmed Disability Improvement (6mCDI) Sustained Until End of Study (EOS) as Measured by EDSS - Pooled Data
NCT02792231 (30) [back to overview]6-month Confirmed Disability Improvement (6mCDI ) Based on EDSS - Study COMB157G2302
NCT02792231 (30) [back to overview]6-month Confirmed Disability Improvement (6mCDI ) Based on EDSS - Pooled Data
NCT02792231 (30) [back to overview]6-month Confirmed Cognitive Decline on Symbol Digit Modalities Test (SDMT) - Pooled Data
NCT02792231 (30) [back to overview]3-month Confirmed Disability Worsening (3mCDW) Based on EDSS > 8 Weeks After Onset of Treatment - Pooled Data
NCT02792231 (30) [back to overview]3-month Confirmed Disability Worsening (3mCDW) Based on EDSS - Study COMB157G2302
NCT02792231 (30) [back to overview]3-month Confirmed Disability Worsening (3mCDW) Based on EDSS - Pooled Data
NCT02792231 (30) [back to overview]Participants With Confirmed Relapse
NCT02792231 (30) [back to overview]Pharmacokinetic (PK) Concentrations of Ofatumumab
NCT02792231 (30) [back to overview]Percent Change in T2 Lesion Volume Relative to Baseline
NCT02792231 (30) [back to overview]Number of New or Enlarging T2 Lesions Per Year by NfL High-low Subgroups - Pooled Data
NCT02792231 (30) [back to overview]Number of New or Enlarging T2 Lesions on MRI Per Year (Annualized Lesion Rate)
NCT02792231 (30) [back to overview]No Evidence of Disease Activity (NEDA-4)
NCT02792231 (30) [back to overview]Neurofilament Light Chain (NfL) Concentration in Serum
NCT02792231 (30) [back to overview]Multiple Sclerosis Impact Scale (MSIS-29) Psychological Impact Score Change From Baseline
NCT02792231 (30) [back to overview]Multiple Sclerosis Impact Scale (MSIS-29) Physical Impact Score Change From Baseline
NCT02792231 (30) [back to overview]Change in Cognitive Performance Measured by the Symbol Digit Modalities Test (SDMT) - Pooled Data
NCT02792231 (30) [back to overview]Brain Volume Loss by NfL High-low Subgroups - Pooled Data
NCT02792231 (30) [back to overview]Annualized Relapse Rates (ARR) by NfL High-low Subgroups - Pooled Data
NCT02792231 (30) [back to overview]6-month Confirmed Worsening of at Least 20% in the Timed 25-Foot Walk (T25FW) - Pooled Data
NCT02792231 (30) [back to overview]6-month Confirmed Worsening of at Least 20% in the 9-Hole Peg Test (9HPT) - Pooled Data
NCT02792231 (30) [back to overview]6-month Confirmed Disability Worsening (6mCDW) or 6-month Confirmed Cognitive Decline (6mCCD) - Pooled Data
NCT02792231 (30) [back to overview]6-month Confirmed Disability Worsening (6mCDW) Based on EDSS > 8 Weeks After Onset of Treatment - Pooled Data
NCT03277248 (8) [back to overview]Annualized Relapse Rate (ARR)
NCT03277248 (8) [back to overview]Percent Change From Baseline in Brain Volume
NCT03277248 (8) [back to overview]Percentage of Participants With Impaired Symbol Digit Modalities Test (SDMT)
NCT03277248 (8) [back to overview]Percentage of Participants With No Evidence of Disease Activity (NEDA)
NCT03277248 (8) [back to overview]Time to Confirmed Disability Progression (CDP) for at Least 12 Weeks
NCT03277248 (8) [back to overview]Total Number of Gadolinium (Gd)-Enhancing T1-Lesions Per Magnetic Resonance Imaging (MRI) Scan Per Participant
NCT03277248 (8) [back to overview]Total Number of New and Enlarging T2 Hyperintense Lesions (NELs) Per MRI Scan Per Participant
NCT03277248 (8) [back to overview]Percentage of Participants With Treatment-Emergent Adverse Events (TEAEs) and Treatment-Emergent Serious Adverse Events (TESAEs)
NCT03277261 (8) [back to overview]Time to Confirmed Disability Progression (CDP) for at Least 12 Weeks
NCT03277261 (8) [back to overview]Total Number of Gadolinium (Gd)-Enhancing T1-Lesions Per Magnetic Resonance Imaging (MRI) Scan Per Participant
NCT03277261 (8) [back to overview]Total Number of New and Enlarging T2 Hyperintense Lesions (NELs) Per MRI Scan Per Participant
NCT03277261 (8) [back to overview]Annualized Relapse Rate (ARR)
NCT03277261 (8) [back to overview]Percent Change From Baseline in Brain Volume
NCT03277261 (8) [back to overview]Percentage of Participants With Impaired Symbol Digit Modalities Test (SDMT)
NCT03277261 (8) [back to overview]Percentage of Participants With No Evidence of Disease Activity (NEDA)
NCT03277261 (8) [back to overview]Percentage of Participants With Treatment Emergent Adverse Events (TEAEs) and Treatment Emergent Serious Adverse Events (TESAEs)

Changes From Baseline in Fatigue Impact Scale [FIS] Total Score

"FIS is a subject-reported scale that qualifies the impact of fatigue on daily life in patients with MS. It consists of 40 statements that measure fatigue in three areas; physical, cognitive, and social.~FIS total score ranges from 0 (no problem) to 160 (extreme problem).~Least-square means were estimated using a Mixed-effect model with repeated measures [MMRM] on FIS total score data (treatment group, region of enrollment, baseline EDSS stratum, visit, treatment-by-visit interaction, baseline value, and baseline-by-visit interaction as factors)." (NCT00134563)
Timeframe: baseline (before randomization) and 108 weeks

Interventionunits on a scale (Least Squares Mean)
Placebo4.300
Teriflunomide 7 mg2.343
Teriflunomide 14 mg3.804

[back to top]

Annualized Relapse Rate [ARR]: Poisson Regression Estimates

"ARR is obtained from the total number of confirmed relapses that occured during the treatment period divided by the sum of the treatment durations.~Each episode of relapse - appearance, or worsening of a clinical symptom that was stable for at least 30 days, that persisted for a minimum of 24 hours in the absence of fever - was to be confirmed by an increase in EDSS score or Functional System scores.~To account for the different treatment durations among participants, a Poisson regression model with robust error variance was used (total number of confirmed relapses as response variable; log-transformed treatment duration as offset variable; treatment group, region of enrollment and baseline EDSS stratum as covariates)." (NCT00134563)
Timeframe: 108 weeks

Interventionrelapses per year (Number)
Placebo0.539
Teriflunomide 7 mg0.370
Teriflunomide 14 mg0.369

[back to top]

Cerebral Magnetic Resonance Imaging [MRI] Assessment: Change From Baseline in Total Lesion Volume (Burden of Disease)

Total lesion volume is the sum of the total volume of all T2-lesions and the total volume all T1-hypointense post-gadolinium lesions measured through T2/proton density scan analysis and gadolinium-enhanced T1 scan analysis. (NCT00134563)
Timeframe: baseline (before randomization) and 108 weeks

,,
Interventionmililiters (mL) (Mean)
Change in total lesion volume- Change in T1-hypointense lesion component- Change in T2-lesion component
Placebo2.2080.5331.674
Teriflunomide 14 mg0.7230.3310.392
Teriflunomide 7 mg1.3080.4990.810

[back to top]

Time to 12-week Sustained Disability Progression: Kaplan-Meier Estimates of the Rate of Disability Progression at Timepoints

"12-week sustained disability progression was defined as an increase from baseline of at least 1-point in EDSS score (at least 0.5-point for participants with baseline EDSS score >5.5) that persisted for at least 12 weeks.~Probability of disability progression at 24, 48 and 108 weeks was estimated using Kaplan-Meier method on the time to disability progression defined as the time from randomization to first EDSS increase. Participants free of disability progression (no disability progression observed on treatment) were censored at the date of the last on-treatment EDSS evaluation.~Kaplan-Meier method consists in computing probabilities of non occurrence of event at any observed time of event and multiplying successive probabilities for time ≤t by any earlier computed probabilities to estimate the probability of being event-free for the amount of time t. Probability of event at time t is 1 minus the probability of being event-free for the amount of time t." (NCT00134563)
Timeframe: 108 weeks

,,
Interventionpercent probability (Number)
Probability of disability progression at 24 weeksProbability of disability progression at 48 weeksProbability of disability progression at 108 weeks
Placebo8.616.027.3
Teriflunomide 14 mg6.211.320.2
Teriflunomide 7 mg5.813.121.7

[back to top]

Cerebral MRI Assessment: Volume of Gd-enhancing T1-lesions Per Scan

Total volume of Gd-enhancing T1-lesions per scan is obtained from the sum of the volumes of Gd-enhancing T1-lesions observed during the study divided by the total number of scans performed during the study. (NCT00134563)
Timeframe: 108 weeks

Interventionmililiters (mL) (Mean)
Placebo0.102
Teriflunomide 7 mg0.059
Teriflunomide 14 mg0.025

[back to top]

Cerebral MRI Assessment: Number of Gd-enhancing T1-lesions Per Scan (Poisson Regression Estimates)

"Number of Gd-enhancing T1-lesions per scan is obtained from the total number of Gd-enhancing T1-lesions observed during the study divided by the total number of scans performed during the study.~To account for the different number of scans among participants, a Poisson regression model with robust error variance was used (total number of Gd-enhancing T1-lesions as response variable; log-transformed number of scans as offset variable; treatment group, region of enrollment, baseline EDSS stratum and baseline number of Gd-enhancing T1-lesions as covariates)." (NCT00134563)
Timeframe: 108 weeks

Interventionlesions per scan (Number)
Placebo1.331
Teriflunomide 7 mg0.570
Teriflunomide 14 mg0.261

[back to top]

Cerebral MRI Assessment: Number of Gd-enhancing T1-lesions Per Scan (Poisson Regression Estimates)

"Number of Gd-enhancing T1-lesions per scan is obtained from the total number of Gd-enhancing T1-lesions observed during the study divided by the total number of scans performed during the study.~To account for the different number of scans among participants, a Poisson regression model with robust error variance was used (total number of Gd-enhancing T1-lesions as response variable; log-transformed number of scans as offset variable; treatment group, region of enrollment and baseline number of Gd-enhancing T1-lesions as covariates)." (NCT00475865)
Timeframe: 24 weeks

Interventionlesions per scan (Number)
Placebo + GA0.367
Teriflunomide 7 mg + GA0.109
Teriflunomide 14 mg + GA0.171

[back to top]

Overview of Adverse Events (AE]

AE are any unfavorable and unintended sign, symptom, syndrome, or illness observed by the investigator or reported by the participant during the study. (NCT00475865)
Timeframe: from first study drug intake up to 112 days after last intake or up to the first intake in the extension study LTS6047, whichever occured first (40 weeks max)

,,
Interventionparticipants (Number)
Any AE- serious AE- AE leading to death- AE leading to study drug discontinuation
Placebo + GA32300
Teriflunomide 14 mg + GA35104
Teriflunomide 7 mg + GA35303

[back to top]

Cerebral Magnetic Resonance Imaging [MRI] Assessment: Change From Baseline in Total Lesion Volume (Burden of Disease)

"Total lesion volume is the sum of the total volume of all T2-lesions and the total volume all T1-hypointense post-gadolinium lesions measured through T2/proton density scan analysis and gadolinium-enhanced T1 scan analysis.~Least-square means were estimated using a Mixed-effect model with repeated measures [MMRM] on cubic root transformed volume data (treatment group, region of enrollment, visit, treatment-by-visit interaction, baseline value (cubic root transformed), and baseline-by-visit interaction as factors)." (NCT00475865)
Timeframe: baseline (before randomization) and 24 weeks

Interventionmililiters (mL) (Least Squares Mean)
Placebo + GA-0.006
Teriflunomide 7 mg + GA-0.030
Teriflunomide 14 mg + GA-0.036

[back to top]

Annualized Relapse Rate [ARR]: Poisson Regression Estimates

"ARR is obtained from the total number of confirmed relapses that occured during the treatment period divided by the sum of the treatment durations.~Each episode of relapse - appearance, or worsening of a clinical symptom that was stable for at least 30 days, that persisted for a minimum of 24 hours in the absence of fever - was to be confirmed by an increase in Expanded Disability Status Scale [EDSS] score or Functional System scores.~To account for the different treatment durations among participants, a Poisson regression model with robust error variance was used (total number of confirmed relapses as response variable; log-transformed treatment duration as offset variable; treatment group and region of enrollment as covariates)." (NCT00475865)
Timeframe: 24 weeks

Interventionrelapses per year (Number)
Placebo + GA0.475
Teriflunomide 7 mg + GA0.311
Teriflunomide 14 mg + GA0.647

[back to top]

Overview of AE With Potential Risk of Occurrence

"AE with potential risk of occurrence were defined as follows:~Hepatic disorders;~Immune effects, mainly effects on bone marrow and infection;~Pancreatic disorders;~Malignancy;~Skin disorders, mainly hair loss and hair thinning;~Pulmonary disorders;~Hypertension;~Peripheral neuropathy;~Psychiatric disorders;~Hypersensitivity." (NCT00475865)
Timeframe: from first study drug intake up to 112 days after last intake or up to the first intake in the extension study LTS6047, whichever occured first (40 weeks max)

,,
Interventionparticipants (Number)
Any AE with potential risk of occurence- Hepatic disorder AE- Pancreatic disorder AE- Pulmonary disorder AE- Immune effects related AE- Hair loss / Hair thinning AE- Hypertension-related AE- Peripheral neuropathy AE- Hypersensitivity AE- Malignancy AE- Psychiatric disorder AE
Placebo + GA2446018102301
Teriflunomide 14 mg + GA2856015705801
Teriflunomide 7 mg + GA2825118511303

[back to top]

Pharmacokinetic [PK]: Teriflunomide Plasma Concentration

Plasma concentrations of teriflunomide were measured using validated liquid chromatography-tandem mass spectrometry methods. (NCT00475865)
Timeframe: 24 weeks

Interventionmicrograms/mililiter (μg/mL) (Mean)
Teriflunomide 7 mg + GA23.443
Teriflunomide 14 mg + GA46.992

[back to top]

Liver Function: Number of Participants With Potentially Clinically Significant Abnormalities (PCSA)

"PCSA values are abnormal values considered medically important by the Sponsor according to predefined criteria based on literature review.~Hepatic parameters thresholds were defined as follows:~Alanine Aminotransferase [ALT] >3, 5, 10 or 20 Upper Normal Limit [ULN];~Aspartate aminotransferase [AST] >3, 5, 10 or 20 ULN;~Alkaline Phosphatase >1.5 ULN;~Total Bilirubin [TB] >1.5 or 2 ULN;~ALT >3 ULN and TB >2 ULN." (NCT00475865)
Timeframe: from first study drug intake up to 112 days after last intake or up to the first intake in the extension study LTS6047, whichever occured first (40 weeks max)

,,
Interventionparticipants (Number)
ALT >3 ULN- ALT >5 ULN- ALT >10 ULNAST >3 ULN- AST >5 ULNAlkaline Phosphatase >1.5 ULNTB >1.5 ULNALT >3 ULN and TB >2 ULN
Placebo + GA11111000
Teriflunomide 14 mg + GA11000000
Teriflunomide 7 mg + GA00000000

[back to top]

Cerebral MRI Assessment: Volume of Gd-enhancing T1-lesions Per Scan

Total volume of Gd-enhancing T1-lesions per scan is obtained from the sum of the volumes of Gd-enhancing T1-lesions observed during the study divided by the total number of scans performed during the study. (NCT00475865)
Timeframe: 24 weeks

Interventionmililiters per scan (Number)
Placebo + GA0.063
Teriflunomide 7 mg + GA0.028
Teriflunomide 14 mg + GA0.017

[back to top]

Pharmacokinetic [PK]: Teriflunomide Plasma Concentration

Plasma concentrations of teriflunomide were measured using validated liquid chromatography-tandem mass spectrometry methods. (NCT00489489)
Timeframe: 24 weeks

Interventionmicrograms/mililiter (μg/mL) (Mean)
Teriflunomide 7 mg + IFN-β21.437
Teriflunomide 14 mg + IFN-β47.761

[back to top]

Cerebral MRI Assessment: Volume of Gd-enhancing T1-lesions Per Scan

Total volume of Gd-enhancing T1-lesions per scan is obtained from the sum of the volumes of Gd-enhancing T1-lesions observed during the study divided by the total number of scans performed during the study. (NCT00489489)
Timeframe: 24 weeks

Interventionmililiters per scan (Number)
Placebo + IFN-β0.068
Teriflunomide 7 mg + IFN-β0.022
Teriflunomide 14 mg + IFN-β0.024

[back to top]

Cerebral MRI Assessment: Number of Gd-enhancing T1-lesions Per Scan (Poisson Regression Estimates)

"Number of Gd-enhancing T1-lesions per scan is obtained from the total number of Gd-enhancing T1-lesions observed during the study divided by the total number of scans performed during the study.~To account for the different number of scans among participants, a Poisson regression model with robust error variance was used (total number of Gd-enhancing T1-lesions as response variable; log-transformed number of scans as offset variable; treatment group, region of enrollment, IFN-β dose level and baseline number of Gd-enhancing T1-lesions as covariates)." (NCT00489489)
Timeframe: 24 weeks

Interventionlesions per scan (Number)
Placebo + IFN-β0.570
Teriflunomide 7 mg + IFN-β0.099
Teriflunomide 14 mg + IFN-β0.089

[back to top]

Cerebral Magnetic Resonance Imaging [MRI] Assessment: Change From Baseline in Total Lesion Volume (Burden of Disease)

"Total lesion volume is the sum of the total volume of all T2-lesions and the total volume of all T1-hypointense post-gadolinium lesions measured through T2/proton density scan analysis and gadolinium-enhanced T1 scan analysis.~Least-square means were estimated using a Mixed-effect model with repeated measures [MMRM] on cubic root transformed volume data (treatment group, region of enrollment, IFN-β dose level, visit, treatment-by-visit interaction, baseline value (cubic root transformed), and baseline-by-visit interaction as factors)." (NCT00489489)
Timeframe: baseline (before randomization) and 24 weeks

Interventionmililiters (Least Squares Mean)
Placebo + IFN-β-0.001
Teriflunomide 7 mg + IFN-β0.002
Teriflunomide 14 mg + IFN-β-0.028

[back to top]

Annualized Relapse Rate [ARR]: Poisson Regression Estimates

"ARR is obtained from the total number of confirmed relapses that occured during the treatment period divided by the sum of the treatment durations.~Each episode of relapse - appearance, or worsening of a clinical symptom that was stable for at least 30 days, that persisted for a minimum of 24 hours in the absence of fever - was to be confirmed by an increase in Expanded Disability Status Scale [EDSS] score or Functional System scores.~To account for the different treatment durations among participants, a Poisson regression model with robust error variance was used (total number of confirmed relapses as response variable; log-transformed treatment duration as offset variable; treatment group, region of enrollment and IFN-β dose level as covariates)." (NCT00489489)
Timeframe: 24 weeks

Interventionrelapses per year (Number)
Placebo + IFN-β0.260
Teriflunomide 7 mg + IFN-β0.280
Teriflunomide 14 mg + IFN-β0.109

[back to top]

Overview of AE With Potential Risk of Occurrence

"AE with potential risk of occurrence were defined as follows:~Hepatic disorders;~Immune effects, mainly effects on bone marrow and infection;~Pancreatic disorders;~Malignancy;~Skin disorders, mainly Hair loss and Hair thinning;~Pulmonary disorders;~Hypertension;~Peripheral neuropathy;~Psychiatric disorders;~Hypersensitivity." (NCT00489489)
Timeframe: from first study drug intake up to 112 days after last intake or up to the first intake in the extension study LTS6047, whichever occured first (40 weeks max)

,,
Interventionparticipants (Number)
Any AE with potential risk of occurence- Hepatic disorder AE- Pancreatic disorder AE- Pulmonary disorder AE- Immune effects related AE- Hair loss / Hair thinning AE- Hypertension-related AE- Peripheral neuropathy AE- Hypersensitivity AE- Malignancy AE- Psychiatric disorder AE
Placebo + IFN-β2455013014500
Teriflunomide 14 mg + IFN-β26117019343300
Teriflunomide 7 mg + IFN-β2582018301300

[back to top]

Liver Function: Number of Participants With Potentially Clinically Significant Abnormalities (PCSA)

"PCSA values are abnormal values considered medically important by the Sponsor according to predefined criteria based on literature review.~Hepatic parameters thresholds were defined as follows:~Alanine Aminotransferase [ALT] >3, 5, 10 or 20 Upper Normal Limit [ULN];~Aspartate aminotransferase [AST] >3, 5, 10 or 20 ULN;~Alkaline Phosphatase >1.5 ULN;~Total Bilirubin [TB] >1.5 or 2 ULN;~ALT >3 ULN and TB >2 ULN;" (NCT00489489)
Timeframe: from first study drug intake up to 112 days after last intake or up to the first intake in the extension study LTS6047, whichever occured first (40 weeks max)

,,
Interventionparticipants (Number)
ALT >3 ULN- ALT >5 ULNAST >3 ULN- AST >5 ULNAlkaline Phosphatase >1.5 ULNTB >1.5 ULNALT >3 ULN and TB >2 ULN
Placebo + IFN-β2111100
Teriflunomide 14 mg + IFN-β2110000
Teriflunomide 7 mg + IFN-β0000000

[back to top]

Overview of Adverse Events [AE]

AE are any unfavorable and unintended sign, symptom, syndrome, or illness observed by the investigator or reported by the participant during the study. (NCT00489489)
Timeframe: from first study drug intake up to 112 days after last intake or up to the first intake in the extension study LTS6047, whichever occured first (40 weeks max)

,,
Interventionparticipants (Number)
Any AE- serious AE- AE leading to death- AE leading to study drug discontinuation
Placebo + IFN-β35101
Teriflunomide 14 mg + IFN-β32001
Teriflunomide 7 mg + IFN-β33201

[back to top]

Core Treatment Period: Time to 12-Week Sustained Disability Progression

The 12-week sustained disability progression was defined as increase from baseline of at least 1-point in EDSS score (at least 0.5-point for participants with baseline EDSS score of greater than [>] 5.5) that persisted for at least 12 weeks. Percent probability of participants free of 12-week sustained disability progression at 24, 48, and 108 weeks was estimated using Kaplan-Meier method. (NCT00622700)
Timeframe: Up to a maximum of 108 weeks depending on time of enrollment

,,
Interventionpercent probability (Number)
Percent Probability at 24 weeksPercent Probability at 48 weeksPercent Probability at 108 weeks
Placebo96.091.785.5
Teriflunomide 14 mg97.993.989.2
Teriflunomide 7 mg94.390.186.5

[back to top]

Core Treatment Period: Time to Conversion to Clinically Definite Multiple Sclerosis (CDMS)

Conversion to CDMS was defined by the occurrence of a relapse, which was defined as a new neurological abnormality separated by at least 30 days from onset of a preceding clinical event, presented for at least 24 hours and occurred in the absence of fever or known infection. Percent probability of conversion at 24, 48, and 108 weeks was estimated using Kaplan-Meier method. (NCT00622700)
Timeframe: Up to a maximum of 108 weeks depending on time of enrollment

,,
Interventionpercent probability (Number)
Percent Probability of Conversion at 24 weeksPercent Probability of Conversion at 48 weeksPercent Probability of Conversion at 108 weeks
Placebo14.326.035.9
Teriflunomide 14 mg9.013.724.0
Teriflunomide 7 mg8.714.227.6

[back to top]

Core Treatment Period: Brain Magnetic Resonance Imaging (MRI) Assessment: Change From Baseline in Total Lesion Volume at Week 108

The total lesion volume (burden of disease) is the total volumes of hyperintense on T2 plus hypointense on T1 as measured by MRI scan. Least-square means were estimated using a Mixed-effect model with repeated measures (MMRM) on cubic root transformed volume data with factors for treatment, baseline monofocal/multifocal status, region, visit, treatment-by-visit interaction, cubic root transformed baseline burden of disease, and baseline-by-visit interaction. (NCT00622700)
Timeframe: Baseline, Week 108

Interventionmilliliter (Least Squares Mean)
Placebo0.053
Teriflunomide 7 mg0.041
Teriflunomide 14 mg-0.038

[back to top]

Core Treatment Period: Brain MRI Assessment: Change From Baseline in Volume of Hypointense Post-Gadolinium T1 Lesion Component

Volume of hypointense post-gadolinium T1 lesion component was measured by MRI scan. Least-square means were estimated using a Mixed-effect model with repeated measures (MMRM) on cubic root transformed volume data adjusted for baseline monofocal/multifocal status, region, visit, treatment-by-visit interaction, cubic root transformed baseline value, and baseline-by-visit interaction (NCT00622700)
Timeframe: Baseline, Week 108

Interventionmilliliter (Least Squares Mean)
Placebo0.028
Teriflunomide 7 mg0.025
Teriflunomide 14 mg-0.033

[back to top]

Core Treatment Period: Annualized Relapse Rate (ARR)

ARR is the total number of confirmed relapses that occurred during the treatment period divided by the total number of patient-years treated. Each episode of relapse (appearance, or worsening of a clinical symptom that was stable for at least 30 days, that persisted for a minimum of 24 hours in the absence of fever) was to be confirmed by an increase in EDSS score or Functional System scores. ARR was assessed using Poisson regression model with robust error variance. To account for the different treatment durations among participants, a Poisson regression model with robust error variance was used (total number of confirmed relapses onset between randomization date and last dose date as the response variable, treatment, region and baseline monofocal/multifocal status as covariates, and log-transformed treatment duration as an offset variable). (NCT00622700)
Timeframe: Up to a maximum of 108 weeks depending on time of enrollment

Interventionrelapses per patient year (Number)
Placebo0.284
Teriflunomide 7 mg0.190
Teriflunomide 14 mg0.194

[back to top]

Core Treatment Period: Brain MRI Assessment: Change From Baseline in Volume of T2 Lesion Component

Volume of T2 lesion component was measured by MRI scan. Least-square means were estimated using a Mixed-effect model with repeated measures (MMRM) on cubic root transformed volume data adjusted for baseline monofocal/multifocal status, region, visit, treatment-by-visit interaction, cubic root transformed baseline value, and baseline-by-visit interaction. (NCT00622700)
Timeframe: Baseline, Week 108

Interventionmilliliter (Least Squares Mean)
Placebo0.052
Teriflunomide 7 mg0.036
Teriflunomide 14 mg-0.035

[back to top]

Core Treatment Period: Brain MRI Assessment: Number of Gadolinium Enhancing (Gd-enhancing) T1-lesions Per MRI Scan (Poisson Regression Estimates)

Number of Gd-enhancing T1-lesions per scan is the total number of Gd-enhancing T1-lesions that occurred during the treatment period divided by the total number of scans performed during the treatment period. To account for the different numbers of scans performed among the participants, a Poisson regression model with robust error variance was used (total number of Gd-enhancing T1-lesions as response variable, treatment, baseline monofocal/multifocal status, region and baseline number of Gd-enhancing T1-lesions as covariates, and log-transformed number of scans as an offset variable). (NCT00622700)
Timeframe: Up to a maximum of 108 weeks depending on time of enrollment

Interventionlesions per scan (Number)
Placebo0.953
Teriflunomide 7 mg0.749
Teriflunomide 14 mg0.395

[back to top]

Core Treatment Period: Brain MRI Assessment: Percent Change From Baseline in Atrophy

Atrophy was measured by MRI scan. (NCT00622700)
Timeframe: Baseline, Week 108

Interventionpercent change (Mean)
Placebo-0.386
Teriflunomide 7 mg-0.197
Teriflunomide 14 mg-0.366

[back to top]

Extension Treatment Period: Overview of Adverse Events (AEs)

AEs are any unfavorable and unintended sign, symptom, syndrome, or illness observed by the investigator or reported by the participant during the study. Safety population included all randomized population who actually received at least 1 dose of the IMP in extension and analyzed according to the treatment actually received in core study followed by treatment actually received in the extension treatment period. (NCT00622700)
Timeframe: From re-randomization up to 283 Weeks

,,,
Interventionparticipants (Number)
Any AEAny Serious AEAny AE Leading to DeathAny AE leading to Permanent Discontinuation
Placebo/ Teriflunomide 14 mg57805
Placebo/Teriflunomide 7 mg47805
Teriflunomide 14 mg/14 mg1202407
Teriflunomide 7 mg/ 7mg1101708

[back to top]

Core Treatment Period: Change From Baseline in EDSS at Week 108

EDSS is an ordinal scale in half-point increments that qualifies disability in participants with MS. It consists of 8 ordinal rating scales assessing seven functional systems (visual, brainstem, pyramidal, cerebellar, sensory, bowel/bladder and cerebral) as well as ambulation. EDSS total score ranges from 0 (normal neurological examination) to 10 (death due to MS). Least-square means were estimated using a Mixed-effect model with repeated measures (MMRM) on cubic root transformed volume data adjusted for baseline monofocal/multifocal status, region, visit, treatment-by-visit interaction, baseline value and baseline-by-visit interaction (NCT00622700)
Timeframe: Baseline, Week 108

Interventionunits on a scale (Least Squares Mean)
Placebo0.069
Teriflunomide 7 mg-0.191
Teriflunomide 14 mg-0.166

[back to top]

Extension Treatment Period: Time to Conversion to Clinically Definite Multiple Sclerosis (CDMS)

Conversion to CDMS was defined by the occurrence of a relapse, which was defined as a new neurological abnormality separated by at least 30 days from onset of a preceding clinical event, presented for at least 24 hours and occurred in the absence of fever or known infection. Percent probability of conversion was estimated using Kaplan-Meier method. (NCT00622700)
Timeframe: From randomization in the core period up to 390 Weeks (Extension treatment period [maximum exposure: 283 Weeks])

,,,
InterventionPercent probability (Number)
Percent Probability of Conversion at 24 WeeksPercent Probability of Conversion at 48 WeeksPercent Probability of Conversion at 72 WeeksPercent Probability of Conversion at 96 WeeksPercent Probability of Conversion at 120 WeeksPercent Probability of Conversion at 144 WeeksPercent Probability of Conversion at 168 WeeksPercent Probability of Conversion at 192 WeeksPercent Probability of Conversion at 216 WeeksPercent Probability of Conversion at 240 WeeksPercent Probability of Conversion at 264 WeeksPercent Probability of Conversion at 288 WeeksPercent Probability of Conversion at 312 WeeksPercent Probability of Conversion at 336 Weeks
Placebo/ Teriflunomide 14 mg13.521.224.327.432.233.933.935.939.339.339.339.349.449.4
Placebo/Teriflunomide 7 mg4.712.515.718.922.322.322.322.325.629.529.529.529.529.5
Teriflunomide 14 mg/14 mg4.78.714.816.218.322.022.824.824.826.326.326.326.326.3
Teriflunomide 7 mg/ 7mg3.59.917.123.927.729.432.237.538.940.843.043.043.048.7

[back to top]

Core Treatment Period: Brain MRI Assessment: Volume of Gadolinium Enhancing (Gd-enhancing) T1-lesions Per MRI Scan

Total volume of Gd-enhancing T1-lesions per scan is the sum of the volumes of Gd-enhancing T1-lesions observed during the treatment period divided by the total number of scans performed during the treatment period. To account for the different numbers of scans performed among the participants, a Poisson regression model with robust error variance was used (total number of Gd-enhancing T1-lesions as response variable, treatment, baseline monofocal/multifocal status, region and baseline number of Gd-enhancing T1-lesions as covariates, and log-transformed number of scans as an offset variable). (NCT00622700)
Timeframe: Up to a maximum of 108 weeks depending on time of enrollment

Interventionmilliliters per scan (Number)
Placebo0.079
Teriflunomide 7 mg0.058
Teriflunomide 14 mg0.034

[back to top]

Core Treatment Period: Change From Baseline in Fatigue Impact Scale (FIS) Total Score at Week 108

FIS is a participant-reported scale that qualifies the impact of fatigue on daily life in participants with MS. It consists of 40 statements that measure fatigue in three areas; physical, cognitive, and social. FIS total score ranges from 0 (no problem) to 160 (extreme problem). Least-square means were estimated using a Mixed-effect model with repeated measures [MMRM] on FIS total score data adjusted for or baseline monofocal/multifocal status, region, visit, treatment-by-visit interaction, baseline value and baseline-by-visit interaction. (NCT00622700)
Timeframe: Baseline, Week 108

Interventionunits on a scale (Least Squares Mean)
Placebo-2.537
Teriflunomide 7 mg-2.524
Teriflunomide 14 mg-1.827

[back to top]

Core Treatment Period: Time to Conversion to Definite Multiple Sclerosis (DMS)

Conversion to DMS was demonstrated by dissemination of MRI lesions in time (as per McDonald criteria) or a relapse, whichever occurs first. MRI Imaging criteria were detection of Gadolinium (Gd) enhancement at least 3 months after onset of initial clinical event, if not at site corresponding to initial event; detection of new T2 lesion if it appears at any time compared with reference scan (done at time of screening) done at least 30 days after onset of the initial clinical event. Occurrence of relapse was defined as new neurological abnormality separated by at least 30 days from onset of preceding clinical event, present for at least 24 hours and occurring in absence of fever or known infection. New clinical abnormality (neurological sign) that is consistent with participant's symptoms with increase in at least one Functional System (FS) or EDSS score compared to last EDSS assessment. Percent probability of conversion at 24, 48, and 108 weeks was estimated using Kaplan-Meier method. (NCT00622700)
Timeframe: Up to a maximum of 108 weeks depending on time of enrollment

,,
Interventionpercent probability (Number)
Percent Probability of Conversion at 24 weeksPercent Probability of Conversion at 48 weeksPercent Probability of Conversion at 108 weeks
Placebo58.272.487.0
Teriflunomide 14 mg46.057.871.5
Teriflunomide 7 mg45.757.373.3

[back to top]

Core Treatment Period: Overview of Adverse Events (AEs)

AEs are any unfavorable and unintended sign, symptom, syndrome, or illness observed by the investigator or reported by the participant during the study. (NCT00622700)
Timeframe: From first study drug intake up to 112 days after last intake in the placebo-controlled period or up to first intake in the extension treatment period, whichever occurred first

,,
Interventionparticipants (Number)
Any AEAny serious AEAny AE leading to deathAny AE leading to treatment discontinuation
Placebo15518119
Teriflunomide 14 mg18324018
Teriflunomide 7 mg16118025

[back to top]

Core Treatment Period: Liver Function: Number of Participants With Potentially Clinically Significant Abnormalities (PCSA)

"PCSA values are abnormal values considered medically important by the Sponsor according to predefined criteria based on literature review.~Hepatic parameters thresholds were defined as follows:~Alanine Aminotransferase (ALT) >3, 5, 10 or 20 upper limit of normal(ULN);~Aspartate aminotransferase (AST) >3, 5, 10 or 20 ULN;~Alkaline Phosphatase >1.5 ULN;~Total Bilirubin (TB) >1.5, 2, or 3 ULN;~ALT >3 ULN and TB >2 ULN." (NCT00622700)
Timeframe: From first study drug intake up to 112 days after last intake in the placebo-controlled period or up to first intake in the extension treatment period, whichever occurred first

,,
Interventionparticipants (Number)
ALT >3 ULN (n=190, 207, 216)ALT >5 ULN (n=190, 207, 216)ALT >10 ULN (n=190, 207, 216)ALT >20 ULN (n=190, 207, 216)AST >3 ULN (n=190, 207, 216)AST >5 ULN (n=190, 207, 216)AST >10 ULN (n=190, 207, 216)AST >20 ULN (n=190, 207, 216)Alkaline Phosphatase >1.5 ULN (n=190, 207, 216)TB >1.5 ULN (n=190, 207, 216)TB >2 ULN (n=190, 207, 216)TB >3 ULN (n=190, 207, 216)ALT >3 ULN and TB >2 ULN (n=190, 207, 216)
Placebo1812509110014802
Teriflunomide 14 mg2611311061118312
Teriflunomide 7 mg2510101240009000

[back to top]

Extension Treatment Period: Overview of Treatment Emergent Adverse Events (TEAE)

AEs were any unfavourable and unintended sign, symptom, syndrome, or illness observed by the investigator or reported by the participant during the study. (NCT00751881)
Timeframe: From first intake of study drug in extension treatment period up to 28 days after the last intake in the extension treatment period

,,
Interventionparticipants (Number)
Any TEAEAny serious TEAEAny TEAE leading to deathAny TEAE leading to treatment discontinuation
Placebo / Teriflunomide 14 mg20316117
Teriflunomide 14 mg / 14 mg18829120
Teriflunomide 7 mg / 14 mg20033317

[back to top]

Extension Treatment Period: Time to Disability Progression

"Probability of disability progression since the randomization of the core period was estimated using Kaplan-Meier method on the time to disability progression defined as the time from randomization to first 12 week sustained disability progression [i.e. increase from baseline of at least 1 point in EDSS score (at least 0.5 point for participants with baseline EDSS score >5.5) that persisted for at least 12 weeks].~Participants free of disability progression (no disability progression observed on treatment) were censored at the date of the last on-treatment EDSS evaluation.~Kaplan-Meier method consists in computing probabilities of non occurrence of event at any observed time of event and multiplying successive probabilities for time ≤t by any earlier computed probabilities to estimate the probability of being event free for the amount of time t. Probability of event at time t was 1 minus the probability of being event-free for the amount of time t." (NCT00751881)
Timeframe: Core treatment period (maximum: 173 weeks) and Extension treatment period (maximum: 174 weeks)

,,
Interventionpercent probability (Number)
1 year2 year3 year4 year5 year
Placebo / Teriflunomide 14 mg0.1300.1900.2450.3070.328
Teriflunomide 14 mg / 14 mg0.0770.1470.1900.2480.265
Teriflunomide 7 mg / 14 mg0.1170.1750.2330.2700.317

[back to top]

Core Treatment Period: Time to Disability Progression

"Probability of disability progression at 24, 48, 108 and 132 weeks was estimated using Kaplan-Meier method on the time to disability progression defined as the time from randomization to first 12-week sustained disability progression [i.e. increase from baseline of at least 1 point in EDSS score (at least 0.5 point for participants with baseline EDSS score >5.5) that persisted for at least 12 weeks].~Participants free of disability progression (no disability progression observed on treatment) were censored at the date of the last on-treatment EDSS evaluation.~Kaplan-Meier method consists in computing probabilities of non occurrence of event at any observed time of event and multiplying successive probabilities for time ≤t by any earlier computed probabilities to estimate the probability of being event-free for the amount of time t. Probability of event at time t is 1 minus the probability of being event-free for the amount of time t." (NCT00751881)
Timeframe: Core treatment period between 48 - 152 weeks depending on time of enrollment

,,
Interventionpercent probability (Number)
Probability of disability progression at 24 weeksProbability of disability progression at 48 weeksProbability of disability progression at 108 weeksProbability of disability progression at 132 weeks
Placebo8.014.219.721.0
Teriflunomide 14 mg2.77.815.815.8
Teriflunomide 7 mg5.312.121.122.2

[back to top]

Core Treatment Period: Change From Baseline to Week 48 in Fatigue Impact Scale (FIS) Total Score

"FIS is a participants-reported scale that qualifies the impact of fatigue on daily life in participants with MS. It consists of 40 statements that measure fatigue in 3 areas; physical, cognitive, and social.~FIS total score ranges from 0 (no problem) to 160 (extreme problem).~Baseline adjusted least-squares means at week 48 were estimated using a Mixed-effect model with repeated measures (MMRM) on FIS total score data (treatment group, region of enrollment, baseline EDSS stratum, visit, treatment-by-visit interaction, baseline value, and baseline-by-visit interaction as factors). All the timepoints from randomization up to Week 48 were included in the model." (NCT00751881)
Timeframe: Baseline (before randomization), Week 12, Week 24 and Week 48

Interventionunits on a scale (Least Squares Mean)
Placebo4.669
Teriflunomide 7 mg2.512
Teriflunomide 14 mg1.915

[back to top]

Extension Treatment Period: ARR: Poisson Regression Estimate

"ARR was obtained from the total number of confirmed relapses that occurred during the treatment period divided by the sum of treatment durations. A relapse is defined as the appearance of a new clinical sign/symptom or clinical worsening of a previous sign/symptom (one that had been stable for at least 30 days) that persists for a minimum of 24 hours in the absence of fever. Relapse was confirmed by an increase in EDSS score or Functional System scores. To account for the different treatment durations among participants, a Poisson regression model with robust error variance was used (total number of confirmed relapses as response variable; log-transformed treatment duration as offset variable; treatment group, region of enrolment and baseline EDSS stratum as covariates)." (NCT00751881)
Timeframe: Extension treatment period (Maximum: 174 weeks)

Interventionrelapses per year (Number)
Placebo / Teriflunomide 14 mg0.199
Teriflunomide 7 mg / 14 mg0.200
Teriflunomide 14 mg / 14 mg0.179

[back to top]

Core Treatment Period: Change From Baseline to Last Visit in Short Form Generic Health Survey - 36 Items (SF-36) Summary Scores

Baseline adjusted least-squares means at last visit were estimated using an analysis of covariance (ANCOVA) model on collected data for each summary score (treatment group, region of enrollment, baseline EDSS stratum, visit number for the last visit and baseline value as factors). (NCT00751881)
Timeframe: Baseline (before randomization) and up to Week 152

,,
Interventionunits on a scale (Least Squares Mean)
Physical Health componentMental Health component
Placebo-1.629-2.792
Teriflunomide 14 mg-0.638-1.087
Teriflunomide 7 mg-0.909-1.704

[back to top]

Core Treatment Period: Change From Baseline to Week 48 in Short Form Generic Health Survey - 36 Items (SF-36) Summary Scores

"SF-36 scale is a generic, self-administered, health-related quality-of-life (QOL) instrument. It is constructed such that the 36 questions represent 8 of the most important health concepts: physical functioning, role-physical, bodily pain, general health, vitality, social functioning, role-emotional, and mental health.~Two summary scores are obtained:~the physical health component summary score,~the mental health component summary score.~Both scores range from 0 to 100 and a high score indicates a more favorable health state.~Baseline adjusted least-squares means at week 48 were estimated using a Mixed-effect model with repeated measures [MMRM] on each summary score data (treatment group, region of enrollment, baseline EDSS stratum, visit, treatment-by-visit interaction, baseline value, and baseline-by-visit interaction as factors). All the timepoints from randomization up to Week 48 were included in the model." (NCT00751881)
Timeframe: Baseline (before randomization), Week 12, Week 24 and Week 48

,,
Interventionunits on a scale (Least Squares Mean)
Physical health componentMental health component
Placebo-1.082-2.913
Teriflunomide 14 mg-0.105-1.434
Teriflunomide 7 mg-0.397-2.031

[back to top]

Core Treatment Period: Liver Function: Number of Participants With Potentially Clinically Significant Abnormalities (PCSA)

"PCSA values are abnormal values considered medically important by the Sponsor according to predefined criteria based on literature review.~Hepatic parameters thresholds were defined as follows:~Alanine Aminotransferase (ALT) >3, 5, 10 or 20 upper limit of normal(ULN);~Aspartate aminotransferase (AST) >3, 5, 10 or 20 ULN;~Alkaline Phosphatase >1.5 ULN;~Total Bilirubin (TB) >1.5 or 2 ULN;~ALT >3 ULN and TB >2 ULN." (NCT00751881)
Timeframe: From first study drug intake up to 112 days after last intake in the core treatment period or up to first intake in the extension treatment period, whichever occurred first

,,
Interventionparticipants (Number)
ALT >3 ULN- ALT >5 ULN- ALT >10 ULNAST >3 ULN- AST >5 ULNAlkaline Phosphatase >1.5 ULNTB >1.5 ULNALT >3 ULN and TB >2 ULN
Placebo22145139592
Teriflunomide 14 mg2911393280
Teriflunomide 7 mg3110293462

[back to top]

Core Treatment Period: Overview of Adverse Events

Adverse Events (AE) are any unfavorable and unintended sign, symptom, syndrome, or illness observed by the investigator or reported by the participant during the study. (NCT00751881)
Timeframe: From first study drug intake up to 112 days after last intake in the core treatment period or up to first intake in the extension treatment period, whichever occurred first

,,
Interventionparticipants (Number)
Any AE- Any serious AE- Any AE leading to death- Any AE leading to treatment discontinuation
Placebo32047124
Teriflunomide 14 mg32044258
Teriflunomide 7 mg34452153

[back to top]

Core Treatment Period: Time Without Relapse

"Probability of no relapse at 24, 48, 108 and 132 weeks was estimated using Kaplan-Meier method on the time to relapse defined as the time from randomization to first EDSS confirmed relapse.~Participants free of confirmed relapse (no EDSS confirmed relapse observed on treatment) were censored at the date of the last study drug intake." (NCT00751881)
Timeframe: Core treatment period between 48 - 152 weeks depending on time of enrollment

,,
Interventionpercent probability (Number)
Probability of no relapse at 24 weeksProbability of no relapse at 48 weeksProbability of no relapse at 108 weeksProbability of no relapse at 132 weeks
Placebo76.460.646.837.7
Teriflunomide 14 mg85.576.357.151.5
Teriflunomide 7 mg81.571.958.255.4

[back to top]

Core Treatment Period: Change From Baseline to Week 48 in EDSS Total Score

"EDSS is an ordinal scale in half-point increments that qualifies disability in participants with MS. It consists of 8 ordinal rating scales assessing 7 functional systems (visual, brainstem, pyramidal, cerebellar, sensory, bowel/bladder and cerebral) as well as ambulation.~EDSS total score ranges from 0 (normal neurological examination) to 10 (death due to MS).~Baseline adjusted least-squares means at Week 48 were estimated using a Mixed-effect model with repeated measures (MMRM) on EDSS score data (treatment group, region of enrollment, baseline EDSS stratum, visit, treatment-by-visit interaction, baseline value, and baseline-by-visit interaction as factors). All the timepoints from randomization up to Week 48 were included in the model." (NCT00751881)
Timeframe: Baseline (before randomization), Week 12, Week 24, Week 36 and Week 48

Interventionunits on a scale (Least Squares Mean)
Placebo0.089
Teriflunomide 7 mg0.042
Teriflunomide 14 mg-0.050

[back to top]

Core Treatment Period: Annualized Relapse Rate (ARR): Poisson Regression Estimate

"ARR is obtained from the total number of confirmed relapses that occurred during the treatment period divided by the sum of treatment durations.~Each episode of relapse - appearance, or worsening of a clinical symptom that was stable for at least 30 days, that persisted for a minimum of 24 hours in the absence of fever - was to be confirmed by an increase in Expanded Disability Status Scale (EDSS) score or Functional System scores.~To account for the different treatment durations among participants, a Poisson regression model with robust error variance was used (total number of confirmed relapses as response variable; log-transformed treatment duration as offset variable; treatment group, region of enrollment and baseline EDSS stratum as covariates)." (NCT00751881)
Timeframe: Core treatment period between 48 - 152 weeks depending on time of enrollment

Interventionrelapses per year (Number)
Placebo0.501
Teriflunomide 7 mg0.389
Teriflunomide 14 mg0.319

[back to top]

Core Treatment Period: Change From Baseline to Last Visit in Fatigue Impact Scale (FIS) Total Score

Baseline adjusted least-squares means at last visit were estimated using an analysis of covariance (ANCOVA) model on collected data for FIS total score (treatment group, region of enrollment, baseline EDSS stratum, visit number for the last visit and baseline value as factors). (NCT00751881)
Timeframe: Baseline (before randomization) and up to Week 152

Interventionunits on a scale (Least Squares Mean)
Placebo6.311
Teriflunomide 7 mg4.464
Teriflunomide 14 mg2.043

[back to top]

Annualized MS Relapse Rate (ARR): Poisson Regression Estimates

"ARR was obtained from total number of confirmed relapses that occurred during treatment period divided by sum of treatment durations in LTS6050 study only. Each episode of relapse - appearance, or worsening of a clinical symptom that was stable for at least 30 days, that persisted for a minimum of 24 hours in the absence of fever was to be confirmed by an increase in EDSS score or Functional System (FS) scores. EDSS: an ordinal scale qualifies disability. EDSS total score range: 0 (normal neurological examination) to 10 (death due to MS). FSS: to assess the neurological function. Total score range: 0 (normal) - 6(worse), higher scores = worse neurological function. To account for the different treatment duration among participants, a Poisson regression model with robust error variance was used (total number of confirmed relapses as response variable; log transformed treatment duration as offset variable; treatment group, region of enrolment and baseline EDSS stratum as covariates)." (NCT00803049)
Timeframe: Up to 8 years since LTS6050 randomization

Interventionrelapses per participant-year (Number)
Placebo/Teriflunomide 7 mg0.216
Teriflunomide 7 mg/7 mg0.183
Placebo/Teriflunomide 14 mg0.176
Teriflunomide 14 mg/14 mg0.160

[back to top]

Magnetic Resonance Imaging (MRI) Assessment: Change From Baseline in Total Volume of Abnormal Lesions (Burden of Disease [BOD]) at Week 192 Since LTS6050 Randomization

BOD was assessed by cerebral MRI and defined as the total volume of all abnormal brain tissue (calculated as the sum of the total volume of T2-lesion component and T1-hypointense lesion component). (NCT00803049)
Timeframe: Baseline, Week 192

Interventionmillilitres (ml) (Mean)
Placebo/Teriflunomide 7 mg5.307
Teriflunomide 7 mg/7 mg3.969
Placebo/Teriflunomide 14 mg3.720
Teriflunomide 14 mg/14 mg3.943

[back to top]

Time to 12 Week Sustained Disability Progression (DP): Kaplan-Meier Estimates of the Rate of DP

Sustained DP defined as sustained increase of at least 1 point from baseline (EFC6049) expanded disability status scale (EDSS) score (0.5 point for participants with baseline EDSS>5.5) persisting for at least 12 weeks. EDSS: an ordinal scale qualifies disability in participants with MS. EDSS total score range: 0 (normal neurological examination) to 10 (death due to Multiple Sclerosis [MS]). Probability of DP at 12 weeks was estimated using Kaplan-Meier method on time to DP defined as date of first DP minus (-) date of randomization in EFC6049 study +1 day. Participants free of DP (no DP observed on treatment) were censored at the date of last on-treatment EDSS evaluation in LTS6050. Kaplan-Meier method consists in computing probabilities of non-occurrence of event at any observed time of event and multiplying successive probabilities for time ≤t by any earlier computed probabilities to estimate the probability of being event-free for the amount of time t. (NCT00803049)
Timeframe: Up to 10.8 years (EFC6049: 108 weeks + LTS6050: 450 weeks)

InterventionProbability (Number)
Placebo/Teriflunomide 7 mg0.544
Teriflunomide 7 mg/7 mg0.494
Placebo/Teriflunomide 14 mg0.627
Teriflunomide 14 mg/14 mg0.473

[back to top]

Time to 24 Week Sustained Disability Progression (DP): Kaplan-Meier Estimates of the Rate of DP

Sustained DP was defined as sustained increase of at least 1 point from baseline (EFC6049) EDSS score (0.5 point for participants with baseline EDSS>5.5) persisting for at least 24 weeks. EDSS: an ordinal scale qualifies disability in participants with MS. EDSS total score range: 0 (normal neurological examination) to 10 (death due to MS). Probability of DP at 24 weeks was estimated using Kaplan-Meier method on time to DP defined as date of first DP minus (-) date of randomization in EFC6049 study +1 day. Participants free of DP (no DP observed on treatment) were censored at the date of last on-treatment EDSS evaluation in LTS6050. Kaplan-Meier method consists in computing probabilities of non-occurrence of event at any observed time of event and multiplying successive probabilities for time ≤t by any earlier computed probabilities to estimate the probability of being event-free for the amount of time t. (NCT00803049)
Timeframe: Up to 10.8 years (EFC6049: 108 weeks + LTS6050: 450 weeks)

InterventionProbability (Number)
Placebo/Teriflunomide 7 mg0.444
Teriflunomide 7 mg/7 mg0.434
Placebo/Teriflunomide 14 mg0.518
Teriflunomide 14 mg/14 mg0.438

[back to top]

Percentage of Participants Free of Sustained Disability Progression (DP)

Sustained DP was defined as sustained increase of at least 1 point from baseline (EFC6049) EDSS score (0.5 point for participants with baseline EDSS>5.5) persisting for at least 12 weeks and 24 weeks. EDSS is an ordinal scale in half-point increments that qualifies disability in participants with MS. It consists of 8 ordinal rating scales assessing seven functional systems (visual, brainstem, pyramidal, cerebellar, sensory, bowel/bladder, cerebral) as well as ambulation. EDSS total score ranges from 0 (normal neurological examination) to 10 (death due to MS), where higher scores indicates worse neurological function. Percentage of participants who were considered as free of disability progression confirmed after 12 week sustained progression and 24 week sustained progression were reported. Analysis for this outcome measure was performed on combined data of EFC6049 and LTS6050 study, as pre-specified in protocol. (NCT00803049)
Timeframe: Up to 10.8 years since EFC6049 randomization (EFC6049: 108 weeks + LTS6050: 450 weeks)

,,,
Interventionpercentage of participants (Number)
Free of DP Sustained for 12 WeeksFree of DP Sustained for 24 Weeks
Placebo/Teriflunomide 14 mg37.348.2
Placebo/Teriflunomide 7 mg45.655.6
Teriflunomide 14 mg/14 mg52.756.2
Teriflunomide 7 mg/7 mg50.656.6

[back to top]

Percentage of Participants With Treatment-Emergent Adverse Events (TEAEs)

Adverse event (AE) was defined as any untoward medical occurrence in a participant who received investigational medicinal product (IMP) without regard to possibility of causal relationship with this treatment. TEAEs: AEs that developed or worsened or became serious during on-treatment period which was defined as the period from the time of first dose of study drug (in LTS6050) up to 4 weeks (28 days) after last dose of study drug. Serious adverse event (SAE) was defined as any untoward medical occurrence that resulted in any of the following outcomes: death, life-threatening, required initial or prolonged in-patient hospitalization, persistent or significant disability/incapacity, congenital anomaly/birth defect, or considered as medically important event. Any TEAE included both serious and non-serious AEs. (NCT00803049)
Timeframe: Baseline (LTS6050) up to 28 days after last dose of study drug up to 450 weeks

,,,
Interventionpercentage of participants (Number)
Any TEAEAny Treatment Emergent SAEAny TEAE Leading to DeathAny TEAE Leading to Permanent Discontinuation
Placebo/Teriflunomide 14 mg94.421.5012.1
Placebo/Teriflunomide 7 mg93.824.82.317.1
Teriflunomide 14 mg/14 mg91.224.80.810.8
Teriflunomide 7 mg/7 mg91.328.00.812.2

[back to top]

Overview of Adverse Events [AE]

AE are any unfavorable and unintended sign, symptom, syndrome, or illness observed by the investigator or reported by the participant during the study. (NCT00811395)
Timeframe: from first study drug intake in PDY6045/PDY6046 study up to 112 days after last intake in initial study or in the extension study, whichever occured last (64 weeks max)

,,,,,
Interventionparticipants (Number)
Any AE- serious AE- AE leading to death- AE leading to study drug discontinuation
Placebo + GA39602
Placebo + IFN-β35202
Teriflunomide 14 mg + GA38205
Teriflunomide 14 mg + IFN-β33103
Teriflunomide 7 mg + GA40503
Teriflunomide 7 mg + IFN-β35403

[back to top]

Overview of AE With Potential Risk of Occurence

"AE with potential risk of occurrence were defined as follows:~Hepatic disorders;~Immune effects, mainly effects on bone marrow and infection;~Pancreatic disorders;~Malignancy;~Skin disorders, mainly hair loss and hair thinning;~Pulmonary disorders;~Hypertension;~Peripheral neuropathy;~Psychiatric disorders;~Hypersensitivity." (NCT00811395)
Timeframe: from first study drug intake in PDY6045/PDY6046 study up to 112 days after last intake in initial study or in the extension study, whichever occured last (64 weeks max)

,,,,,
Interventionparticipants (Number)
Any AE with potential risk of occurence- Hepatic disorder AE- Immune effects related AE- Pancreatic disorder AE- Malignancy AE- Hair loss / hair thinning AE- Pulmonary disorder AE- Hypertension-related AE- Peripheral neuropathy AE- Psychiatric disorder AE- Hypersensitivity AE
Placebo + GA3452760100434
Placebo + IFN-β2871680101516
Teriflunomide 14 mg + GA3252111070210110
Teriflunomide 14 mg + IFN-β301320110406424
Teriflunomide 7 mg + GA3342260512536
Teriflunomide 7 mg + IFN-β30112150304314

[back to top]

Time to 12-week Sustained Disability Progression: Kaplan-Meier Estimates of the Rate of Disability Progression at Timepoints

"Probability of disability progression at 24 and 48 weeks was estimated using Kaplan-Meier method on the time to disability progression defined as the time from randomization to first EDSS increase. Participants free of disability progression were censored at the date of the last on-treatment EDSS evaluation.~Kaplan-Meier method consists in computing probabilities of non occurrence of event at any observed time of event and multiplying successive probabilities for time ≤t by any earlier computed probabilities to estimate the probability of being event-free for the amount of time t. Probability of event at time t is 1 minus the probability of being event-free for the amount of time t." (NCT00811395)
Timeframe: 48 weeks

,,,,,
Interventionpercent probability (Number)
Probability of disability progression at 24 weeksProbability of disability progression at 48 weeks
Placebo + GA2.510.6
Placebo + IFN-β0.00.0
Teriflunomide 14 mg + GA5.612.8
Teriflunomide 14 mg + IFN-β2.76.4
Teriflunomide 7 mg + GA0.03.3
Teriflunomide 7 mg + IFN-β3.011.1

[back to top]

Liver Function: Number of Participants With Potentially Clinically Significant Abnormalities [PCSA]

"PCSA values are abnormal values considered medically important by the Sponsor according to predefined criteria based on literature review.~Hepatic parameters thresholds were defined as follows:~Alanine Aminotransferase [ALT] >3, 5, 10 or 20 Upper Normal Limit [ULN];~Aspartate aminotransferase [AST] >3, 5, 10 or 20 ULN;~Alkaline Phosphatase >1.5 ULN;~Total Bilirubin [TB] >1.5 or 2 ULN;~ALT >3 ULN and TB >2 ULN;" (NCT00811395)
Timeframe: from first study drug intake in PDY6045/PDY6046 study up to 112 days after last intake in initial study or in the extension study, whichever occured last (64 weeks max)

,,,,,
Interventionparticipants (Number)
ALT >3 ULN- ALT >5 ULN- ALT >10 ULNAST >3 ULN- AST >5 ULNAlkaline Phosphatase >1.5 ULNTB >1.5 ULNALT >3 ULN and TB >2 ULN
Placebo + GA11110000
Placebo + IFN-β21011100
Teriflunomide 14 mg + GA11000000
Teriflunomide 14 mg + IFN-β31010000
Teriflunomide 7 mg + GA00000000
Teriflunomide 7 mg + IFN-β10000000

[back to top]

Cerebral MRI Assessment: Total Volume of Gd-enhancing T1-lesions Per Scan

Total volume of Gd-enhancing T1-lesions per scan is obtained from the sum of the volumes of Gd-enhancing T1-lesions observed during the study divided by the total number of scans performed during the study. (NCT00811395)
Timeframe: 48 weeks

Interventionmililiters per scan (Number)
Placebo + IFN-β0.068
Teriflunomide 7 mg + IFN-β0.019
Teriflunomide 14 mg + IFN-β0.020
Placebo + GA0.052
Teriflunomide 7 mg + GA0.031
Teriflunomide 14 mg + GA0.014

[back to top]

Cerebral MRI Assessment: Number of Gd-enhancing T1-lesions Per Scan (Poisson Regression Estimates)

"Number of Gd-enhancing T1-lesions per scan is obtained from the total number of Gd-enhancing T1-lesions observed during the study divided by the total number of scans performed during the study.~To account for the different number of scans among participants, two Poisson regression models with robust error variance were used (total number of Gd-enhancing T1-lesions as response variable, log-transformed number of scans as offset variable and:~Model 1 (IFN-β groups): Treatment group, region of enrollment, IFN-β dose level and baseline number of Gd-enhancing T1-lesions as covariates~Model 2 (GA groups): Treatment group, region of enrollment and baseline number of Gd-enhancing T1-lesions as covariates)" (NCT00811395)
Timeframe: 48 weeks

Interventionlesions per scan (Number)
Placebo + IFN-β0.521
Teriflunomide 7 mg + IFN-β0.080
Teriflunomide 14 mg + IFN-β0.090
Placebo + GA0.333
Teriflunomide 7 mg + GA0.120
Teriflunomide 14 mg + GA0.178

[back to top]

Cerebral Magnetic Resonance Imaging [MRI] Assessment: Change From Baseline in Total Lesion Volume (Burden of Disease)

"Total lesion volume is the sum of the total volume of all T2-lesions and the total volume all T1-hypointense post-gadolinium lesions measured through T2/proton density scan analysis and gadolinium-enhanced T1 scan analysis.~Least-square means were estimated using two Mixed-effect models with repeated measures [MMRM] on cubic root transformed volume data:~Model 1 (IFN-β groups): treatment group, region of enrollment, IFN-β dose level, visit, treatment-by-visit interaction, baseline value (cubic root transformed), and baseline-by-visit interaction as factors;~Model 2 (GA groups): treatment group, region of enrollment, visit, treatment-by-visit interaction, baseline value (cubic root transformed), and baseline-by-visit interaction as factors." (NCT00811395)
Timeframe: baseline (before randomization in PDY6045 or PDY6046) and 48 weeks

Interventionmililiters (mL) (Least Squares Mean)
Placebo + IFN-β-0.017
Teriflunomide 7 mg + IFN-β-0.011
Teriflunomide 14 mg + IFN-β-0.012
Placebo + GA0.016
Teriflunomide 7 mg + GA-0.010
Teriflunomide 14 mg + GA-0.063

[back to top]

Annualized Relapse Rate [ARR]: Poisson Regression Estimates

"ARR is obtained from the total number of confirmed relapses that occured during the treatment period divided by the sum of the treatment durations.~Each episode of relapse - appearance, or worsening of a clinical symptom that was stable for at least 30 days, that persisted for a minimum of 24 hours in the absence of fever - was to be confirmed by an increase in Expanded Disability Status Scale [EDSS] score or Functional System scores.~To account for the different treatment durations among participants, two Poisson regression models with robust error variance were used (total number of confirmed relapses as response variable, log-transformed treatment duration as offset variable and:~Model 1 (IFN-β groups): treatment group, region of enrollment and IFN-β dose level as covariates~Model 2 (GA groups): treatment group and region of enrollment as covariates)" (NCT00811395)
Timeframe: 48 weeks

Interventionrelapses per year (Number)
Placebo + IFN-β0.343
Teriflunomide 7 mg + IFN-β0.231
Teriflunomide 14 mg + IFN-β0.144
Placebo + GA0.420
Teriflunomide 7 mg + GA0.262
Teriflunomide 14 mg + GA0.497

[back to top]

Overview of 12-week Sustained Disability Progression

"12-week sustained disability progression was defined as an increase from baseline of at least 1-point in EDSS score (at least 0.5-point for participants with baseline EDSS score >5.5) that persisted for at least 12 weeks.~If no disability progression was observed on or before last EDSS evaluation before study drug discontinuation, then the participant was considered as free of disability progression." (NCT00811395)
Timeframe: 48 weeks

,,,,,
Interventionparticipants (Number)
Disability progressionFree of disability progression
Placebo + GA437
Placebo + IFN-β040
Teriflunomide 14 mg + GA436
Teriflunomide 14 mg + IFN-β236
Teriflunomide 7 mg + GA141
Teriflunomide 7 mg + IFN-β333

[back to top]

Core Treatment Period: Change From Baseline in Fatigue Impact Scale (FIS) Total Score

"FIS is a subject-reported scale that qualifies the impact of fatigue on daily life in patients with MS. It consists of 40 statements that measure fatigue in three areas; physical, cognitive, and social.~FIS total score ranges from 0 (no problem) to 160 (extreme problem).~Least-square means were estimated using a Mixed-effect model with repeated measures [MMRM] on FIS total score data (treatment group, region of enrollment, baseline EDSS stratum, visit, treatment-by-visit interaction, baseline value, and baseline-by-visit interaction as factors)." (NCT00883337)
Timeframe: Baseline (before randomization) and 48 weeks

Interventionunits on a scale (Least Squares Mean)
Teriflunomide 7 mg0.97
Teriflunomide 14 mg4.10
IFN-β-1a9.10

[back to top]

Extension Treatment Period: ARR Poisson Regression Estimates

"ARR was obtained from the total number of confirmed relapses that occurred during the treatment period divided by the sum of the standardized treatment durations.To account for the different treatment durations among participants, a Poisson Regression Model with robust error variance was used (total number of confirmed relapses as response variable; log-transformed treatment duration as offset variable; treatment group, region of enrolment and baseline EDSS stratum as covariates)." (NCT00883337)
Timeframe: Extension treatment period (Maximum: 197 weeks)

Interventionrelapses per year (Number)
Teriflunomide 7 mg / 14 mg0.236
Teriflunomide 14 mg / 14 mg0.193
IFN-β-1a / 14 mg0.252

[back to top]

Core Treatment Period: Overview of Adverse Events [AE]

AE are any unfavorable and unintended sign, symptom, syndrome, or illness observed by the investigator or reported by the participant during the study. (NCT00883337)
Timeframe: from first study drug intake up to 112 days after last intake in the core treatment period or up to first intake in the extension treatment period, whichever occurred first

,,
Interventionparticipants (Number)
Any AE- Any serious AE- Any AE leading to death- Any AE leading to treatment discontinuation
IFN-β-1a977022
Teriflunomide 14 mg1026012
Teriflunomide 7 mg1031209

[back to top]

Core Treatment Period: Overview of Failures

"Failure was defined as the first occurence of confirmed relapse or permanent treatment discontinuation (for any cause) which ever came first. If no events occurred, the participant was considered free of failure.~Each episode of relapse - appearance, or worsening of a clinical symptom that was stable for at least 30 days, that persisted for a minimum of 24 hours in the absence of fever - was to be confirmed by an increase in Expanded Disability Status Scale [EDSS] score or Functional System scores." (NCT00883337)
Timeframe: Core treatment period between 48 and 118 weeks depending on when the participant was enrolled

,,
Interventionparticipants (Number)
FailureFree of failure
IFN-β-1a4460
Teriflunomide 14 mg4269
Teriflunomide 7 mg5356

[back to top]

Core Treatment Period: Time to Failure: Kaplan-Meier Estimates of the Rate of Failure at Timepoints

"Probability of disability progression at 24, 48 and 96 weeks was estimated using Kaplan-Meier method on the time to failure defined as the time from randomization to failure. Participants free of failure were censored at the date of last treatment.~Kaplan-Meier method consists in computing probabilities of non occurrence of event at any observed time of event and multiplying successive probabilities for time ≤t by any earlier computed probabilities to estimate the probability of being event-free for the amount of time t. Probability of event at time t is 1 minus the probability of being event-free for the amount of time t." (NCT00883337)
Timeframe: Core treatment period between 48 and 118 weeks depending on when the participant was enrolled

,,
Interventionpercent probability (Number)
Probability of failure at 24 weeksProbability of failure at 48 weeksProbability of failure at 96 weeks
IFN-β-1a29.836.544.4
Teriflunomide 14 mg24.333.341.1
Teriflunomide 7 mg25.735.858.8

[back to top]

Core Treatment Period: Treatment Satisfaction Questionnaire for Medication [TSQM] Scores

TSQM version 1.4 is an instrument to assess patients' satisfaction with medication. It consists of 13 questions that cover three dimensions (effectiveness, side effects and convenience) plus a global satisfaction question. Four scores ranging from 0 to 100 (extremely satisfied) are obtained. Least-square means were estimated using a Mixed-effect model with repeated measures [MMRM] on TSQM score data (treatment group, region of enrollment, baseline EDSS stratum, visit, treatment-by-visit interaction as factors). (NCT00883337)
Timeframe: 48 weeks

,,
Interventionunits on a scale (Least Squares Mean)
Effectivness scoreSide effects scoreConvenience scoreGlobal satisfaction score
IFN-β-1a59.3071.3861.9060.98
Teriflunomide 14 mg63.1393.1589.8568.82
Teriflunomide 7 mg67.2595.2988.3068.29

[back to top]

Extension Treatment Period: Overview of AEs

AEs were any unfavourable and unintended sign, symptom, syndrome, or illness observed by the investigator or reported by the participant during the study. (NCT00883337)
Timeframe: From first intake of study drug in extension treatment period up to 28 days after the last intake in the extension treatment period

,,
Interventionparticipants (Number)
Any AEAny serious AEAny AE leading to deathAny AE leading to treatment discontinuation
IFN-β-1a / 14 mg481205
Teriflunomide 14 mg / 14 mg761306
Teriflunomide 7 mg / 14 mg83908

[back to top]

Core Treatment Period: Annualized Relapse Rate [ARR] - Poisson Regression Estimates

"ARR is obtained from the total number of confirmed relapses that occured during the treatment period divided by the sum of the treatment durations.~To account for the different treatment durations among participants, a Poisson regression model with robust error variance was used (total number of confirmed relapses as response variable; log-transformed treatment duration as offset variable; treatment group, region of enrollment and baseline EDSS stratum as covariates)." (NCT00883337)
Timeframe: Core treatment period between 48 and 118 weeks depending on when the participant was enrolled

Interventionrelapses per year (Number)
Teriflunomide 7 mg0.410
Teriflunomide 14 mg0.259
IFN-β-1a0.216

[back to top]

Brain MRI Assessment: Change From Baseline in Total Lesion Volume (Burden of Disease) at Week 24

The total lesion volume (burden of disease) is the total volumes of hyperintense on T2 plus hypointense on T1 as measured by MRI scan. Least-square means were estimated using a Mixed-effect model with repeated measures (MMRM) on cubic root transformed volume data with factors for treatment, region, IFN-beta dose stratum, visit, treatment-by-visit interaction, cubic root transformed baseline burden of disease, and baseline-by-visit interaction. (NCT01252355)
Timeframe: Baseline, Week 24

Interventionmilliliter (Least Squares Mean)
Placebo + IFN-beta-0.008
Teriflunomide 7 mg + IFN-beta-0.011
Teriflunomide 14 mg + IFN-beta-0.044

[back to top]

Brain MRI Assessment: Volume of Gd-enhancing T1-lesions Per MRI Scan

Total volume of Gd-enhancing T1-lesions per scan is the sum of the volumes of Gd-enhancing T1-lesions observed during the treatment period divided by the total number of scans performed during the treatment period. (NCT01252355)
Timeframe: Up to a maximum of 108 weeks depending on time of enrollment

Interventionmilliliters per scan (Number)
Placebo + IFN-beta0.045
Teriflunomide 7 mg + IFN-beta0.009
Teriflunomide 14 mg + IFN-beta0.01

[back to top]

Liver Function: Number of Participants With Potentially Clinically Significant Abnormalities (PCSA)

PCSA values are abnormal values considered medically important by the Sponsor according to predefined criteria based on literature review. Hepatic parameters thresholds were defined as follows: Alanine Aminotransferase (ALT) >3, 5 or 10 Upper Limit of Normal (ULN); Aspartate Aminotransferase (AST) >3, 5 or 10 ULN; Alkaline Phosphatase >1.5 ULN; Total Bilirubin (TB) >1.5 ULN; and ALT >3 ULN and TB >2 ULN. (NCT01252355)
Timeframe: First study drug intake up to 28 days after last study drug intake, for up to 112 weeks

,,
Interventionparticipants (Number)
ALT >3 ULNALT >5 ULNALT >10 ULNAST >3 ULNAST >5 ULNAST >10 ULNAlkaline Phosphatase >1.5 ULNTB >1.5 ULNALT >3 ULN and TB >2 ULN
Placebo + IFN-beta611321020
Teriflunomide 14 mg + IFN-beta952430020
Teriflunomide 7 mg + IFN-beta963432200

[back to top]

Overview of Adverse Events (AEs)

AEs are any unfavorable and unintended sign, symptom, syndrome, or illness observed by the investigator or reported by the participant during the study. (NCT01252355)
Timeframe: First study drug intake up to 28 days after last study drug intake, for up to 112 weeks

,,
Interventionparticipants (Number)
Any AEAny Serious AEAny AE Leading to DeathAny AE Leading to Study Drug Discontinuation
Placebo + IFN-beta119809
Teriflunomide 14 mg + IFN-beta14014022
Teriflunomide 7 mg + IFN-beta14013016

[back to top]

Time to Relapse: Kaplan-Meier Estimates of the Probability of no Relapse at Week 24, 48, and 72

Probability of no relapse at 24, 48 and 72 weeks was estimated using Kaplan-Meier method on the time to relapse defined as the time from randomization to first EDSS confirmed relapse. Participants free of confirmed relapse (no EDSS confirmed relapse observed on treatment) were censored at the date of the last study drug intake. Kaplan-Meier method consists in computing probabilities of non-occurrence of event at any observed time of event and multiplying successive probabilities for time <=t by any earlier computed probabilities to estimate the probability of being event-free for the amount of time t. (NCT01252355)
Timeframe: Up to a maximum of 108 weeks depending on time of enrollment

,,
Interventionpercent probability of no relapse (Number)
Percent probability of no relapse at Week 24Percent probability of no relapse at Week 48Percent probability of no relapse at Week 72
Placebo + IFN-beta81.967.358.3
Teriflunomide 14 mg + IFN-beta87.180.873.1
Teriflunomide 7 mg + IFN-beta86.880.678.2

[back to top]

Brain Magnetic Resonance Imaging (MRI) Assessment: Number of Gadolinium Enhancing (Gd-enhancing) T1-lesions Per Scan (Poisson Regression Estimates)

Number of Gd-enhancing T1-lesions per scan is the total number of Gd-enhancing T1-lesions that occurred during the treatment period divided by the total number of scans performed during the treatment period. To account for the different number of scans among participants, a Poisson regression model with robust error variance was used (total number of Gd-enhancing T1-lesions as response variable; log-transformed number of scans as offset variable; treatment group, region of enrollment, IFN-beta dose stratum and baseline number of Gd-enhancing T1-lesions as covariates). (NCT01252355)
Timeframe: Up to a maximum of 108 weeks depending on time of enrollment

Interventionlesions per scan (Number)
Placebo + IFN-beta0.542
Teriflunomide 7 mg + IFN-beta0.257
Teriflunomide 14 mg + IFN-beta0.158

[back to top]

Annualized Relapse Rate (ARR) (Poisson Regression Estimates)

"ARR is the total number of confirmed relapses that occurred during the treatment period divided by the total number of patient-years treated. Each episode of relapse (appearance, or worsening of a clinical symptom that was stable for at least 30 days, that persisted for a minimum of 24 hours in the absence of fever) was to be confirmed by an increase in Expanded Disability Status Scale (EDSS) score or Functional System scores. To account for the different treatment durations among participants, a Poisson regression model with robust error variance was used (total number of confirmed relapses as response variable; log-transformed treatment duration as offset variable; treatment group, region of enrollment and IFN-beta dose stratum, and number of relapses in the year prior to randomization as covariates)." (NCT01252355)
Timeframe: Up to a maximum of 108 weeks depending on time of enrollment

Interventionrelapses per patient-year (Number)
Placebo + IFN-beta0.298
Teriflunomide 7 mg + IFN-beta0.242
Teriflunomide 14 mg + IFN-beta0.238

[back to top]

Immunoglobulin Levels

(NCT01403376)
Timeframe: pre vaccination (baseline) and 28 days post vaccination

,,
Interventiong/L (Mean)
Immunoglobulin M - pre vaccinationImmunoglobulin M - 28-day post vaccinationImmunoglobulin G - pre vaccinationImmunoglobulin G - 28-day post vaccinationImmunoglobulin A - pre vaccinationImmunoglobulin A - 28-day post vaccination
IFN-β-11.141.1612.0312.252.272.33
Teriflunomide 14 mg1.231.279.129.091.511.50
Teriflunomide 7 mg1.281.309.359.571.651.63

[back to top]

Geometric Mean of Titers (GMT) Ratio Post/Pre Vaccination

(NCT01403376)
Timeframe: pre vaccination (baseline) and 28 days post vaccination

,,
Interventionratio post/pre vaccination (Geometric Mean)
H1N1 strainH3N2 strainB strain
IFN-β-13.44.14.7
Teriflunomide 14 mg2.32.63.0
Teriflunomide 7 mg2.52.63.1

[back to top]

Percentage of Participants With 2 Fold or More Increase in Antibody Titer at 28 Days Post Vaccination

Percentages of participants with an increase from baseline of 2-fold or more in antibody titers and 90% CIs using normal approximation were calculated for each strain and treatment group. (NCT01403376)
Timeframe: pre vaccination (baseline) and 28 days post vaccination

,,
Interventionpercentage of participants (Number)
H1N1 strainH3N2 strainB strain
IFN-β-146.551.258.1
Teriflunomide 14 mg30.841.051.3
Teriflunomide 7 mg35.035.047.5

[back to top]

Percentage of Participants With 4 Fold or More Increase in Antibody Titer at 28 Days Post Vaccination

Percentages of participants with an increase from baseline of 4-fold or more in antibody titers and 90% CIs using normal approximation were calculated for each strain and treatment group. (NCT01403376)
Timeframe: pre vaccination (baseline) and 28 days post vaccination

,,
Interventionpercentage of participants (Number)
H1N1 strainH3N2 strainB strain
IFN-β-139.541.951.2
Teriflunomide 14 mg20.523.130.8
Teriflunomide 7 mg22.525.037.5

[back to top]

Percentage of Participants With Antibody Titer ≥40 at 28 Days Post Vaccination

"For each viral strain (H1N1, H3N2, and B), the antibody titer, level of antibodies in blood sample when exposed to antigen, was calculated as the mean of two replicates. If the titer was below or above the limit of detection, the threshold value was used.~The percentage of participants achieving a titer of 40 or more, as well as the 90% confidence interval (CI) using normal approximation were calculated for each strain and treatment group." (NCT01403376)
Timeframe: 28 days post vaccination

,,
Interventionpercentage of participants (Number)
H1N1 strainH3N2 strainB strain
IFN-β-197.790.793.0
Teriflunomide 14 mg97.476.997.4
Teriflunomide 7 mg97.590.097.5

[back to top]

Time to Relapse: Kaplan-Meier Estimates of the Probability of Treated Relapse at Week 4, Week 24 and Week 48

A treated relapse was defined as a relapse treated by a systemic corticosteroid treatment or by another DMT. If a participant had no treated relapse before treatment discontinuation/completion, then the participant was considered as free of treated relapse until the date of treatment discontinuation/completion. Only treated relapse occurred during the treatment period (first drug administration to last drug administration) were considered for analysis. Kaplan-Meier method was used to estimate the probability of treated MS relapse at 4, 24 and 48 weeks. (NCT01895335)
Timeframe: Baseline up to end of treatment (up to Week 48)

Interventionpercent probability of treated relapse (Number)
Percent Probability of Treated Relapse at Week 4Percent Probability of Treated Relapse at Week 24Percent Probability of Treated Relapse at Week 48
Teriflunomide1.89.415.5

[back to top]

Annualized Treated Relapse Rate

Annualized treated relapse rate was defined as the total number of treated relapses during the study treatment period divided by the total number participants-years of treatment. Only events occurred during the treatment period (first drug administration to last drug administration) were considered for analysis. (NCT01895335)
Timeframe: Baseline up to end of treatment (up to Week 48)

Interventionrelapses per patient-year (Number)
Teriflunomide0.200

[back to top]

Change From Baseline in Cognition Measured by Symbol Digit Modalities Test (SDMT) Score at Week 48

SDMT measures the time to pair abstract symbols with specific numbers. It is a simple substitution task that gives the examinee 90 seconds to pair specific numbers with given geometric figures as a measure for screening cognitive impairment. The score is computed as a ratio of number of correct responses divided by the total number of responses. The test score range from 0 (worst outcome) to 1 (best outcome). Higher scores are indicative of better cognition function. (NCT01895335)
Timeframe: Baseline, Week 48

Interventionunits on a scale (Mean)
Teriflunomide0.00

[back to top]

Change From Baseline in Disease Progression Using Patient Determined Disease Steps (PDDS) Score at Week 48

PDDS scale developed to assess the disability in Multiple Sclerosis (MS) participants and in assessing disease progression that focuses mainly on how participants walk. PDDS scale consists of 0 = normal; 1 = mild disability; 2 = moderate disability; 3 = gait disability; 4 = early cane; 5 = late cane; 6 = bilateral support; 7 = wheelchair/scooter and 8 = bedridden. A higher score represented higher level of disability. (NCT01895335)
Timeframe: Baseline, Week 48

Interventionunits on a scale (Mean)
Teriflunomide-0.01

[back to top]

Change From Baseline in Multiple Sclerosis International Quality of Life (MusiQoL) Score at Week 48

The MusiQoL is a quality of life questionnaire that consists of 31 questions, divided into 9 dimensions: activities of daily living, physiological well-being, symptoms, relationship with friends, relationship with family, sentimental and sexual life, coping, rejection and relationship with healthcare system. All the 9 dimension scores and the global scores are linearly transformed and standardized on 0 (worst outcome) -100 (best outcome) scale. Higher scores represents higher quality of life. (NCT01895335)
Timeframe: Baseline, Week 48

Interventionunits on a scale (Mean)
Teriflunomide0.99

[back to top]

Change From Baseline in Stern Leisure Activity Scale at Week 48

The Stern Leisure Activity Scale is a self-reported scale that consists of 13 questions assessing the participant's participation in leisure activities during the preceding month. One point is given for participation in each of the 13 activities and an aggregate score (range from 0 to 13) is obtained. ≤ 6 score is considered as low leisure activity and > 6 score as high leisure activity. (NCT01895335)
Timeframe: Baseline, Week 48

Interventionunits on a scale (Mean)
Teriflunomide0.07

[back to top]

Duration of Teriflunomide Treatment Exposure

Duration of exposure was defined as last dose date - first dose date + 1 day, regardless of unplanned intermittent discontinuations and regardless of dosage administered (14 mg or 7 mg). (NCT01895335)
Timeframe: Baseline up to end of treatment (up to Week 48)

InterventionDays (Mean)
Teriflunomide301.6

[back to top]

Percentage of Participants With Treatment Compliance of ≥80% During the Study Treatment Period

Percentage of compliance for a participant was defined as the number of days that the participant was compliant (1 tablet/day) divided by the exposure duration in days (from the first dose administration to the last dose administration) times 100. (NCT01895335)
Timeframe: Baseline up to end of treatment (up to Week 48)

Interventionpercentage of participants (Number)
Teriflunomide98.2

[back to top]

Treatment Satisfaction Questionnaire for Medication (TSQM) Version 1.4 - Assessment of Global Satisfaction Subscale Score With Teriflunomide Treatment at Week 48

"TSQM version 1.4 is a global satisfaction scale used to assess the overall level of participant's satisfaction or dissatisfaction with their medications. It comprises of 14 items assessing the following 4 domains: effectiveness (questions: 1-3), side effects (questions: 4-8), convenience (questions: 9-11), global satisfaction (questions:12-14).~Primary outcome was the global satisfaction score. The score of the corresponding item was added based on the algorithm to create a score of 0 to 100. Higher score indicated greater satisfaction in that domain." (NCT01895335)
Timeframe: Week 48

Interventionunits on a scale (Mean)
Teriflunomide68.17

[back to top]

Change From Baseline in Multiple Sclerosis Performance Scale (MSPS) Score at Week 24 and Week 48

MSPS was a self-reported measure for MS associated disability in which participants were asked to indicate the category that best described their condition during the past month on the following 8 subscales: mobility, hand function, vision, fatigue, cognitive symptoms, bladder/bowel, sensory symptoms and spasticity symptoms. MSPS used a single question to assess each of 8 subscales. All of the subscales ranged from 0= normal to 5= total disability, except mobility subscale which ranged from 0= normal to 6=total disability. Total MSPS score ranged from 0 =normal to 41=greater disability, where higher score reflected greater disability. (NCT01895335)
Timeframe: Baseline, Week 24, Week 48

Interventionunits on a scale (Mean)
Change at Week 24 (n=854)Change at Week 48 (n=875)
Teriflunomide-0.61-0.06

[back to top]

Change From Baseline in TSQM Scores in Participants Switching From Another Disease Modifying Therapy (DMT) at Week 4 and Week 48

TSQM version 1.4 is a global satisfaction scale used to assess the overall level of participant's satisfaction or dissatisfaction with their medications. It comprises of 14 items assessing the following 4 domains: effectiveness (questions: 1-3), side effects (questions: 4-8), convenience (questions: 9-11), global satisfaction (questions: 12-14). For each of the 4 domains the scores of the corresponding items were added based on an algorithm to create a score of 0 to 100. Higher scores indicated greater satisfaction . (NCT01895335)
Timeframe: Baseline, Week 4, Week 48

Interventionunits on a scale (Mean)
Global Satisfaction Score Change at Week 4 (n=482)Global Satisfaction Score Change at Week 48(n=457)Effectiveness Score Change at Week 4 (n=477)Effectiveness Score Change at Week 48 (n=453)Side effects Score Change at Week 4 (n=479)Side effects Score Change at Week 48 (n=456)Convenience Score Change at Week 4 (n=487)Convenience Score Change at Week 48 (n=461)
Teriflunomide21.3516.5512.0210.1924.3119.9534.6432.21

[back to top]

Change From Week 4 in TSQM Scores in Naïve Participants to Week 48

TSQM version 1.4 is a global satisfaction scale used to assess the overall level of participant's satisfaction or dissatisfaction with their medications. It comprises of 14 items assessing the following 4 domains: effectiveness (questions: 1-3), side effects (questions: 4-8), convenience (questions: 9-11), global satisfaction (questions: 12-14). For each of the 4 domains the scores of the corresponding items were added based on an algorithm to create a score of 0 to 100. Higher scores indicated greater satisfaction. (NCT01895335)
Timeframe: Week 4, Week 48

Interventionunits on a scale (Mean)
Change in Global Satisfaction Score (n=234)Change in Effectiveness Score (n=231)Change in Side effects Score (n=234)Change in Convenience Score (n=235)
Teriflunomide-1.341.76-5.440.33

[back to top]

Expanded Disability Status Scale (EDSS) Score at Baseline and Week 48

EDSS is a method of quantifying disability in MS participants and monitoring changes in the level of disability over time. EDSS quantifies disability in 8 functional systems: pyramidal, cerebellar, brainstem, sensory, bowel and bladder, visual, cerebral, and other. EDSS scale ranges from 0 to 10 in 0.5 unit increments that represents higher levels of disability. EDSS score 1.0 to 4.5 refers to people with MS who are fully ambulatory; EDSS score 5.0 to 9.5 refers to impairment to ambulation; EDSS score 10 refers to death due to MS. (NCT01895335)
Timeframe: Baseline, Week 48

Interventionunits on a scale (Mean)
Baseline (n=981)Week 48 (n=886)
Teriflunomide3.053.05

[back to top]

Overview of Adverse Events (AEs)

Any untoward medical occurrence in a participant who received investigational medicinal product (IMP) was considered an AE without regard to possibility of causal relationship with this treatment. Treatment-emergent adverse events (TEAEs) were defined as AEs that developed or worsened or became serious during from first study drug intake up to 112 days after last intake for participant with no accelerated elimination procedure (AEP) or to last AEP follow up visit for participants with AEP. A serious adverse event (SAE) was defined as any untoward medical occurrence that resulted in any of the following outcomes: death, life-threatening, required initial or prolonged in-patient hospitalization, persistent or significant disability/incapacity, congenital anomaly/birth defect, or considered as medically important event. Any TEAE included participants with both serious and non-serious AEs. (NCT01895335)
Timeframe: From first study drug intake up to 112 days after last intake for participant with no AEP or to last AEP follow up visit for participants with AEP

Interventionpercentage of participants (Number)
Any TEAEAny treatment emergent SAEAny TEAE leading to deathAny TEAE leading to permanent discontinuation
Teriflunomide82.312.70.410.9

[back to top]

MAIN STUDY: Number of Participants Relapse Free at 24 Months

Number of patients relapses free by month 24. (NCT01970410)
Timeframe: 24 months

InterventionParticipants (Count of Participants)
Teriflunomide25

[back to top]

"MAIN STUDY: Time to Return of Radiological Evidence of Multiple Sclerosis Activity With New Gadolinium Enhancing (Gd+) Lesions on Cranial MRI."

"Mean time to first Gadolinium enhancing (GAD+) lesion in months." (NCT01970410)
Timeframe: 24 months

InterventionMonths (Mean)
Teriflunomide19.6

[back to top]

MAIN STUDY: Expanded Disability Status Scale (EDSS) Sustained Progression for 3 Months as Measured by at Least 0.5 Increase From Baseline or 1 in Any EDSS Set Score

Mean time to 3-month sustained disability worsening (SDW) in months. SDW is defined as an increase of ≥ 1 point for patients with EDSS of 1.0-5.0, and ≥ 0.5 points for patients with an EDSS of 5.5-6.0, sustained for 3 months. Patients with ≥ 1 point increase in EDSS in whom a second measure was not obtained 3 months later were not included as SDW. (NCT01970410)
Timeframe: 24 months

InterventionMonths (Mean)
Teriflunomide22

[back to top]

MAIN STUDY: Mean Time to New T2 or Enlarging T2 Hyperintensities on Monthly Sentinel Brain MRIs

Mean time of new T2 or enlarging T2 Lesions (NCT01970410)
Timeframe: 24 months

InterventionMonths (Mean)
Teriflunomide19.2

[back to top]

SUB-STUDY: Number of Patients With a Reduction of Anti-JCV Antibody Index (AJAI)

"Number of patients with a >= 20% reduction of AJAI.~The AJAI serostatus categories used in the study were negative (<0.2), positive (0.41-0.89) and high positive (≥0.90). The 0.2-0.4 range is frequently reported as indeterminate. We defined a change of 20% or greater as a meaningful increase or decrease in the AJAI." (NCT01970410)
Timeframe: 30 days

InterventionParticipants (Count of Participants)
Anti-JCV Antibody Status (Sub-Study)20

[back to top]

Cognitive Assessment: Change From Baseline in Trail Making Test- Part A (TMT-A) Test Scores (in Seconds) at Week 96 and 192

'Trail Making Test Part A' is a neuropsychological test of visual attention and task switching. The task requires a participant to 'connect-the-dots' of 25 consecutive numbers (1,2, 3, etc.) in sequential order on a sheet of paper or computer screen. The goal of the participant is to finish the test as quickly as possible, and the time taken to complete the test used as the primary performance metric (in seconds). This is a timed test and the number of seconds to complete the task is recorded. Maximum time allowed is 300 seconds. A lower score indicated better cognitive function. (NCT02201108)
Timeframe: Baseline, Weeks 96 and 192

,
Interventionseconds (Mean)
BaselineChange at Week 96Change at Week 192
Placebo/Teriflunomide43.48.46.3
Teriflunomide/ Teriflunomide47.1-3.1-6.6

[back to top]

Brain Magnetic Resonance Imaging Assessment: Change From Baseline in Volume of T2 Lesions at Weeks 24, 36, 48, 72, 96, 144 and 192

Volume of T2 lesions was measured by MRI scan. (NCT02201108)
Timeframe: Baseline, DB period: Weeks 24, 36, 48, 72 and 96; OL period: Weeks 48, 96, 144 and 192

,
Interventionmilliliters (Mean)
DB Period: Week 24DB Period: Week 36DB Period: Week 48DB Period: Week 72DB Period: Week 96OL Period: Week 48OL Period: Week 96OL Period: Week 144OL Period: Week 192
Placebo/Teriflunomide3.04.60.51.20.93.02.74.70.4
Teriflunomide/ Teriflunomide0.54.4-0.20.10.20.61.90.4-3.6

[back to top]

Probability of Participants Who Were Clinical Relapse Free at Weeks 24, 48, 72, 96, 120, 144, 168 and 192

Participant was considered free of clinical relapse if the participant had no confirmed clinical relapse before treatment discontinuation/completion in 192 weeks treatment period. Clinical relapses: new/recurrent neurological symptoms not associated with fever/infection, lasted at least 24 hours, and accompanied by new objective neurological findings upon neurological examination and documented by standardized, quantified FSSs which included 8 items: rated on different scales: brain stem, cerebellar and cerebral functions rated on scale of 0 to 5; visual, pyramidal, sensory and bowel/bladder rated on scale of 0 to 6 & ambulation on scale of 0 to 12, where higher score in each scale indicated worsened neurological function. New/recurrent symptoms occurred less than 30 days following onset of relapse were considered part of same relapse. Probability of participants who were clinical relapse free at specified weeks were estimated by Kaplan-Meier method and reported. (NCT02201108)
Timeframe: Weeks 24, 48, 72, 96, 120, 144, 168 and 192

,
Interventionprobability of relapse free participants (Number)
Week 24Week 48Week 72Week 96Week 120Week 144Week 168Week 192
Placebo/Teriflunomide0.7500.5960.5190.4420.4040.3650.3650.365
Teriflunomide/ Teriflunomide0.8200.7000.6300.6000.5700.5400.5180.518

[back to top]

Brain Magnetic Resonance Imaging (MRI) Assessment: Number of New or Enlarged T2 Lesions Per MRI Scan

Number of new or enlarged T2 lesions per scan was defined as the total number of new or enlarged T2 lesion that occurred during the 192 weeks treatment period divided by the total number of scans performed during 192 weeks. To account for the different numbers of scans performed among the participants, a negative binomial regression model with robust variance estimation was used. The model included the total number of new or enlarged T2-lesions as the response variable, with treatment group, region, pubertal status and age as covariates and log-transformed number of scans as an offset variable. (NCT02201108)
Timeframe: Baseline up to Week 192

Interventionlesions per scan (Number)
Placebo/Teriflunomide11.087
Teriflunomide/ Teriflunomide5.664

[back to top]

Brain Magnetic Resonance Imaging Assessment: Number of New T1 Hypointense Lesions Per MRI Scan

The number of new T1 hypointense lesions were obtained from MRI scans. (NCT02201108)
Timeframe: Baseline up to Week 192

Interventionlesions (Number)
Placebo/Teriflunomide1561
Teriflunomide/ Teriflunomide1910

[back to top]

Brain Magnetic Resonance Imaging Assessment: Number of T1 Gadolinium (Gd)-Enhancing T1 Lesions Per MRI Scan

The number of T1 Gd-Enhancing lesions per scan was defined as the total number of Gd-enhancing lesions that occurred during the 192 weeks treatment period divided by the total number of scans performed during 192 weeks. To account for the different number of scans performed among the participants, a negative binomial regression model with robust variance estimation was used. The model included the total number of T1-lesions as the response variable, with treatment group, region, pubertal status and age as covariates and log-transformed number of scans as an offset variable. (NCT02201108)
Timeframe: Baseline up to Week 192

Interventionlesions per scan (Number)
Placebo/Teriflunomide2.686
Teriflunomide/ Teriflunomide1.532

[back to top]

DB: Pharmacokinetics: Steady-state Trough Concentration (Ctrough) of Teriflunomide

Ctrough was defined as the concentration reached by the drug before the next dose administered. Data for this outcome measure was planned to be collected and analyzed separately for each dose of Teriflunomide. PK samples for teriflunomide 3.5 mg were collected during the first 8 weeks but all participants were switched to teriflunomide 7 mg after Week 8. Hence, plasma concentration of teriflunomide 7 mg and 14 mg were reported. (NCT02201108)
Timeframe: Predose on Week 36

Interventionmicrograms per milliliter (Mean)
Teriflunomide 7 mg53.1
Teriflunomide 14 mg67.8

[back to top]

OL: Pharmacokinetics: Steady-state Trough Concentration (Ctrough) of Teriflunomide

Ctrough was defined as the concentration reached by the drug before the next dose is administered. Data for this outcome measure was planned to be collected and analyzed separately for each dose of teriflunomide. PK samples for teriflunomide 3.5 mg were collected during the first 8 weeks but all participants were switched to teriflunomide 7 mg after Week 8. Hence, plasma concentration of teriflunomide 7 mg and 14 mg were reported. (NCT02201108)
Timeframe: Pre-dose at Week 36

Interventionmicrograms per milliliter (Mean)
Placebo / Teriflunomide 7 mg45.8
Placebo / Teriflunomide 14 mg50.4
Teriflunomide / Teriflunomide 7 mg33.7
Teriflunomide / Teriflunomide 14 mg63.6

[back to top]

OL: Time to First Confirmed Clinical Relapse

Time to first clinical relapse was defined as duration (in weeks) after enrollment in OL period and first confirmed clinical relapse. Clinical relapses were defined as new or recurrent neurological symptoms not associated with fever or infection, lasted at least 24 hours and accompanied by new objective neurological findings upon neurological examination and documented by standardized, quantified FSSs which included 8 items and items were rated on different scales: brain stem, cerebellar & cerebral functions rated on scale of 0 to 5; visual, pyramidal, sensory and bowel/bladder rated on scale of 0 to 6 and ambulation on scale of 0 to 12 where higher score in each scale indicated worsened neurological function. Confirmed clinical relapse were reviewed and confirmed by independent RAP. Participant without confirmed clinical relapse, was considered as clinical relapse free until end of Week 192. (NCT02201108)
Timeframe: Baseline up to Week 192

Interventionweeks (Median)
Placebo/Teriflunomide95.86
Teriflunomide/ Teriflunomide96.00

[back to top]

Time to First Confirmed Clinical Relapse

Time to first clinical relapse was defined as the duration (in weeks) between randomization and first confirmed clinical relapse. Clinical relapses were defined as new or recurrent neurological symptoms not associated with fever or infection, lasting at least 24 hours, and accompanied by new objective neurological findings upon neurological examination and documented by a standardized, quantified functional system score (FSSs) which included 8 items and items were rated on different scales: brain stem, cerebellar and cerebral functions rated on a scale of 0 to 5; visual, pyramidal, sensory and bowel/bladder rated on a scale of 0 to 6 and ambulation on a scale of 0 to 12, where higher score in each scale indicated worsened neurological function. Confirmed clinical relapse were reviewed and confirmed by an independent Relapse Adjudication Panel (RAP). A participant without confirmed clinical relapse, was considered as clinical relapse free until the end of Week 96. (NCT02201108)
Timeframe: Baseline up to Week 96

Interventionweeks (Median)
Double-Blind Treatment Period: Placebo39.14
Double-Blind Treatment Period: Teriflunomide75.29

[back to top]

Brain Magnetic Resonance Imaging Assessment: Change From Baseline in Volume of T1 Hypointense Lesions

Volume of T1 hypointense lesions was measured by MRI scan. (NCT02201108)
Timeframe: Baseline, DB period: Weeks 24, 36, 48, 72 and 96; OL period: Weeks 48, 96, 144 and 192

,
Interventionmilliliters (Mean)
DB Period: Week 24DB Period: Week 36DB Period: Week 48DB Period: Week 72DB Period: Week 96OL Period: Week 48OL Period: Week 96OL Period: Week 144OL Period: Week 192
Placebo/Teriflunomide0.30.80.20.10.10.71.02.04.0
Teriflunomide/ Teriflunomide0.00.20.40.10.10.50.61.92.5

[back to top]

Brain Magnetic Resonance Imaging Assessment: Percent Change From Baseline in Brain Volume at Weeks 24, 36, 48, 72, 96, 144 and 192

Percent change from baseline in brain volume (assessed using MRI scans of the Brain) at Weeks 24, 36, 48,72, 96, 144 and 192 was reported. (NCT02201108)
Timeframe: Baseline, DB period: Weeks 24, 36, 48, 72 and 96; OL period: Weeks 48, 96, 144 and 192

,
Interventionpercent change (Mean)
DB Period: Week 24DB Period: Week 36DB Period: Week 48DB Period: Week 72DB Period: Week 96OL Period: Week 48OL Period: Week 96OL Period: Week 144OL Period: Week 192
Placebo/Teriflunomide-0.3-0.7-0.6-0.9-0.9-1.4-1.8-3.1-2.2
Teriflunomide/ Teriflunomide-0.2-0.4-0.5-0.6-0.8-1.1-1.4-2.0-3.0

[back to top]

Brain Magnetic Resonance Imaging Assessment: Percentage of Participants Free of New or Enlarged MRI T2-Lesions

Percentage of participants who were free of new or enlarged T2 lesions at Weeks 24, 48, 72, 96, 144 and 192 were reported. (NCT02201108)
Timeframe: Baseline, Weeks 24, 48, 72, 96, 144 and 192

,
Interventionpercentage of participants (Number)
Week 24Week 48Week 72Week 96Week 144Week 192
Placebo/Teriflunomide86.532.715.415.411.57.7
Teriflunomide/ Teriflunomide86.025.017.016.014.09.0

[back to top]

Cognitive Assessment - Selective Reminding Test (SRT): Change From Baseline in Total Number of Words on Delayed Recall at Weeks 96 and 192

SRT is a test to assess verbal learning and memory. During the administration of the SRT only the examiner and the participant should be in the testing room. A list of twelve words was read aloud by the examiner at a rate of one word per two seconds. The participant is asked to recall all twelve words after a 30 minute delay. Only the words that were missed on the preceding trial were given in the consecutive trial. The total score represented a sum score of total 6 trials, therefore the score range was from 0 to 72. The lower the score the worse the outcome, higher score indicated better recall. (NCT02201108)
Timeframe: Baseline, Weeks 96 and 192

,
Interventionscore on a scale (Mean)
BaselineChange at Week 96Change at Week 192
Placebo/Teriflunomide3.51.00.0
Teriflunomide/ Teriflunomide10.0-0.50.0

[back to top]

Cognitive Assessment: Change From Baseline in Beery Visual-motor Integration (BVMI) Scores at Weeks 96 and 192

The Beery VMI is a non-verbal assessment that assessed the extent to which individuals can integrate their visual and motor abilities. The participants were provided with geometric designs ranging from simple line drawings to more complex figures and were asked to copy the designs. The test consisted of 24 figures. One point was scored for each successful copy of drawings and no scoring was given when the participant failed to copy the drawings properly. Each successful copying of drawings was summed up and the total was scored on a scale ranged from 0 to 24, where higher score indicated better visual construction skills/better visual and motor abilities and lower score indicated poor visual construction skills/poor visual and motor abilities. (NCT02201108)
Timeframe: Baseline, Weeks 96 and 192

,
Interventionscore on a scale (Mean)
BaselineChange at Week 96Change at Week 192
Placebo/Teriflunomide26.10.60.1
Teriflunomide/ Teriflunomide25.9-0.40.4

[back to top]

Cognitive Assessment: Change From Baseline in Brief Visuospatial Memory Test-Revised (BVMT-R) Scores at Weeks 96 and 192

The BVMT consists of three trials in which participants must recall shapes by drawing figures on a blank page (response booklet) after being given the opportunity to memorize the figures (given in BMVT-R form) for 10 seconds. BMVT-R form consists of six figures. Points are awarded based on the accuracy of the drawn figure and by correct placement on the blank page. A minimum of 0 to 12 points/scores are awarded per trial, so a participant can score between 0 and 36 points for all three trials (by adding the points/score from each trial), where higher score indicates better outcome. (NCT02201108)
Timeframe: Baseline, Weeks 96 and 192

,
Interventionscore on a scale (Mean)
BaselineChange at Week 96Change at Week 192
Placebo/Teriflunomide23.8-0.81.0
Teriflunomide/ Teriflunomide24.81.61.2

[back to top]

Cognitive Assessment: Change From Baseline in Delis-Kaplan Executive Function System (D-KEFS) Letter Fluency Total Correct Raw Score at Weeks 96 and 192

Letter Fluency is a condition measured in the D-KEFS. Participants were asked to name as many words as they can, starting with a specified letter for 60 seconds. The words cannot be names, places, numbers or grammatical variants of previous answers. Repeated answers were not scored as a correct response. There were 3 trials, with 3 different letters. The total number of correct responses was totaled for all 3 trials and a letter fluency score was given. A higher score was considered better. There was no set range as the score depends on how many correct words the participant relays in the given time period. (NCT02201108)
Timeframe: Baseline, Weeks 96 and 192

,
Interventionscore on a scale (Mean)
BaselineChange at Week 96Change at Week 192
Placebo/Teriflunomide32.5-3.05.0
Teriflunomide/ Teriflunomide21.04.0-6.5

[back to top]

Cognitive Assessment: Change From Baseline in Delis-Kaplan Executive Function System Category Fluency Total Correct Raw Score at Weeks 96 and 192

Category Fluency is a condition measured in the D-KEFS. It measured participant's ability to generate words from three different categories (e.g., fruits, vegetables and animals), within a minute for each category. Total score was number of correct words for each category with no points for repetitions or non-words. Score ranged from 0 to unlimited, where 0 = low score, higher score indicated better performance. (NCT02201108)
Timeframe: Baseline, Weeks 96 and 192

,
Interventionscore on a scale (Mean)
BaselineChange at Week 96Change at Week 192
Placebo/Teriflunomide28.06.0-2.0
Teriflunomide/ Teriflunomide27.55.0-13.5

[back to top]

Cognitive Assessment: Change From Baseline in Number of Completed Items Measured by Symbol Digit Modalities Test at Weeks 24, 48, 72, 96, 120, 144, 168 and 192

SDMT measures the time to pair abstract symbols with specific numbers. It is a simple substitution task that gives the examinee 90 seconds to pair specific numbers with given geometric figures as a measure for screening cognitive impairment. The SDMT score is the number of completed items and ranged from 0 (worst outcome) to 110 (best outcome), where higher score indicated better cognitive function. (NCT02201108)
Timeframe: Baseline, DB period: Weeks 24, 48, 72 and 96; OL period: Weeks 24, 48, 72, 96, 120, 144, 168 and 192

,
Interventionscore on a scale (Mean)
DB Period: Week 24DB Period: Week 48DB Period: Week 72DB Period: Week 96OL Period: Week 24OL Period: Week 48OL Period: Week 72OL Period: Week 96OL Period: Week 120OL Period: Week 144OL Period: Week 168OL Period: Week 192
Placebo/Teriflunomide3.86.35.17.14.85.56.46.33.74.82.712.0
Teriflunomide/ Teriflunomide3.64.74.56.97.16.48.17.66.33.7-0.3-1.5

[back to top]

Cognitive Assessment: Change From Baseline in Total Number of Correct Substitutions Measured by Symbol Digit Modalities Test (SDMT) at Weeks 24, 48, 72, 96, 120, 144, 168 and 192

SDMT measures the time to pair abstract symbols with specific numbers. It is a simple substitution task that gives the examinee 90 seconds to pair specific numbers with given geometric figures as a measure for screening cognitive impairment. The SDMT score is the number of correct substitution and ranged from 0 (worst outcome) to 110 (best outcome), where higher score indicated better cognitive function. (NCT02201108)
Timeframe: Baseline, DB period: Weeks 24, 48, 72 and 96; OL period: Weeks 24, 48, 72, 96, 120, 144, 168 and 192

,
Interventionscore on a scale (Mean)
DB Period: Week 24DB Period: Week 48DB Period: Week 72DB Period: Week 96OL Period: Week 24OL Period: Week 48OL Period: Week 72OL Period: Week 96OL Period: Week 120OL Period: Week 144OL Period: Week 168OL Period: Week 192
Placebo/Teriflunomide5.17.36.48.86.36.78.07.63.76.63.512.0
Teriflunomide/ Teriflunomide4.65.75.68.18.37.69.28.07.25.21.7-0.3

[back to top]

Cognitive Assessment: Change From Baseline in Trail Making Test B (TMT-B) Test Scores (in Seconds) at Weeks 96 and 192

TMT-B is a cognitive test that gives a measure of various aspects of cognitive performance. It is used to measure cognitive fatigue. The test consisted of 25 circles containing 13 sequential numbers (1 to 13) and 12 sequential letters (A to L) positioned. The test evaluates the time (in seconds) to correctly order letters and numbers in alternate order (1, A, 2, B etc.). Maximum time allowed is 300 seconds, where less time/lower score indicated better cognitive function/performance. (NCT02201108)
Timeframe: Baseline, Weeks 96 and 192

,
Interventionseconds (Mean)
BaselineChange at Week 96Change at Week 192
Placebo/Teriflunomide113.8-19.8-37.0
Teriflunomide/ Teriflunomide115.0-29.5-18.8

[back to top]

Cognitive Assessment: Change From Baseline in Wechsler Abbreviated Scale of Intelligence-II (WASI-II) Vocabulary Total Raw Scores at Weeks 96 and 192

The WASI-II: Vocabulary test is a quick estimate of an individual's level of intellectual functioning which comprised of 31 total items that required the participant to orally define 3 images and 28 words presented both orally and visually. Items 1 to 3 rated on a score of 0 or 1, items 4 and 5 rated on a score of 0 or 2, items 6 to 31 rated on a scale of 0 to 2. Each item score was summed up to derive the total score which ranged from 0 (minimum score) to 59 (maximum score), where higher score indicated better level of intellectual functioning/higher level of intelligence. (NCT02201108)
Timeframe: Baseline, Weeks 96 and 192

,
Interventionscore on a scale (Mean)
BaselineChange at Week 96Change at Week 192
Placebo/Teriflunomide39.05.03.0
Teriflunomide/ Teriflunomide34.34.05.0

[back to top]

Secondary Outcome Measure: Mean Percentage Change of Serum Teriflunomide Levels Percentage Change of Teriflunomide Concentrations at Day 8, Day 14, Day 26 ad Day 36 Following Administration of Colestipol Hydrochloride Tablets.

The last blood draw will be about 50 days from the start of the study (NCT02263547)
Timeframe: duration of study about 50 days

Interventionpercentage change in teriflunomide level (Mean)
8 days after colestipol hcl administered14 days after colestipol hcl administered26 days after colestipol hcl administered36 days after colestipol hcl administered
Teriflunomide Elimination With Colestipol-56.4-74.9-94.9-96.9

[back to top]

Primary Outcome Measures: Teriflunomide Concentrations at Day 28

After receiving 14 days of teriflunomide, participants will take 11 days of colestipol to wash out the teriflunomide (measuring the levels of teriflunomide in the blood at each visit) (NCT02263547)
Timeframe: 28 days after the start in the study

Interventionteriflunomide level (mcg/ml) (Mean)
Teriflunomide Elimination With Colestipol9.36

[back to top]

Cumulative Number of Combined Unique Active Lesions (CUAL) Per Year From Baseline to Week 108

CUALs was calculated as sum of new Gadolinium-enhanced (Gd+) T1 lesions plus new or enlarging T2 lesions (without double-counting of lesions) from baseline based on the Magnetic resonance imaging (MRI) scans up to Week 108. Average number of lesions per year were reported. (NCT02425644)
Timeframe: Baseline to Week 108

Interventionlesions per year (Mean)
Ponesimod 20 mg1.405
Teriflunomide 14 mg3.164

[back to top]

24-Week Confirmed Disability Accumulation (CDA) Assessed From Baseline to EOS

A 24-week CDA was defined as an increase of at least 1.5 in EDSS for participants with a baseline EDSS score of 0.0 or an increase of at least 1.0 in EDSS for participants with a baseline EDSS score of 1.0 to 5.0, or an increase of at least 0.5 in EDSS for participants with a baseline EDSS score >= 5.5, which was confirmed after 24 weeks. Baseline EDSS was defined as the last EDSS score recorded prior to randomization. The EDSS is an ordinal scale ranging from 0 (normal neurological exam) to 10 (death to MS). (NCT02425644)
Timeframe: Baseline to 60 Weeks and 108 Weeks

,
InterventionPercentage of Participants (Number)
60 Weeks- from Kaplan Meier estimates108 Weeks- from Kaplan Meier estimates
Ponesimod 20 mg6.38.7
Teriflunomide 14 mg6.910.5

[back to top]

12-Week Confirmed Disability Accumulation (CDA) Assessed From Baseline to EOS

A 12-week CDA was defined as an increase of at least 1.5 in Expanded Disability Status Scale (EDSS) for participants with a baseline EDSS score of 0.0 or an increase of at least 1.0 in EDSS for participants with a baseline EDSS score of 1.0 to 5.0, or an increase of at least 0.5 in EDSS for participants with a baseline EDSS score greater than or equal to (>=) 5.5, which was confirmed after 12 weeks. Baseline EDSS was defined as the last EDSS score recorded prior to randomization. EDSS is an ordinal clinical rating scale ranging from 0 (normal neurological examination) to 10 (death due to MS). (NCT02425644)
Timeframe: Baseline to Week 60 and 108 Weeks

,
InterventionPercentage of Participants (Number)
60 Weeks- from Kaplan Meier estimates108 Weeks- from Kaplan Meier estimates
Ponesimod 20 mg7.610.8
Teriflunomide 14 mg8.313.2

[back to top] [back to top]

Annualized Confirmed Relapse Rate

Relapse: occurrence of acute episode of one or more new symptoms or worsened symptoms of Multiple sclerosis (MS), not associated with fever/infection and lasting 24 hours after stable 30 days period. Confirmed relapse: increase from baseline at least 0.5 point Expanded Disability Status Scale (EDSS) score or increase of one point in one, two or three Functional Systems (FS), excluding bowel/bladder and cerebral/mental FS. EDSS and FS scores are based on neurological examination for assessing its impairment in MS. Among eight FS, seven are ordinal clinical rating scales ranging from 0-5 or 6 with higher scale indicates overall functional impairment assessing Visual,Brain Stem,Pyramidal,Cerebellar,Sensory, Bowel/Bladder and Cerebral functions. Rating individual FS scores is used to rate EDSS in conjunction with observations and information concerning gait and use of assistance. EDSS is ordinal clinical rating scale ranging from 0 (normal neurological examination) to 10(death due to MS). (NCT02425644)
Timeframe: From randomization to end of study (Week 108)

Interventionrelapses per year (Mean)
Ponesimod 20 mg0.202
Teriflunomide 14 mg0.290

[back to top]

Annualized Relapse Rates (ARR) by NfL High-low Subgroups - Pooled Data

ARR was the number of confirmed relapses in a year, calculated as the total number of relapses for all participants in the treatment group divided by the total participant-years of time in study. A confirmed MS relapse was defined as one accompanied by a clinically-relevant change in the EDSS performed by the Independent EDSS rater, i.e. an increase of at least 0.5 points on the EDSS score, or an increase of 1 point on two functional scores or 2 points on one functional score (excluding changes involving bowel/bladder or cerebral functional system). (NCT02792218)
Timeframe: Baseline up to 2.5 years

,
Interventionnumber of relapses in a year (Mean)
High > median n= 443,410Low <= median n=428,431
OMB 20 mg0.080.12
TER 14 mg0.210.23

[back to top]

Annual Rate of Percent Change in Brain Volume Loss by NfL High-low Subgroups - Pooled Data

Percent change from baseline in brain volume loss (BVL) on all MRI scans adjusted for different time of scan versus baseline due to variable follow up time in study. (NCT02792218)
Timeframe: Baseline, Months 12 and 24

,
Interventionpercentage of brain volume loss (Mean)
NfL High: > 9.3 median n=416,387NfL Low: <=9.3 median n=403,407
OMB 20 mg-0.32-0.24
TER 14 mg-0.43-0.29

[back to top]

6-month Confirmed Worsening of at Least 20% in the Timed 25-Foot Walk (T25FW) - Pooled Data

The patient is directed to walk 25 feet quickly and safely as possible from one marked end to the other. The time is calculated from the initiation of the patient instructed to begin, until the patient has reached the 25-foot mark. This study was not powered for the analysis of this endpoint individually. It was pre-specified in the study protocol to combine the data from this study with study COMB157G2302 to address this endpoint. (NCT02792218)
Timeframe: Baseline, every 3 months up to 2.5 years

,
Interventionpercentage of participants (Number)
Month 18 - from Kaplan Meier estimatesMonth 24 - from Kaplan Meier estimates
OMB 20 mg11.011.4
TER 14 mg10.410.6

[back to top]

6-month Confirmed Worsening of at Least 20% in the 9-Hole Peg Test (9HPT) - Pooled Data

9 Hole Peg Test is a test of upper limb function. Participants place 9 pegs on pegboard and remove pegs and this is timed for each hand. Time recorded in seconds. Longer time indicates poorer upper limb function. 20% improvement is defined as 20% shorter time in seconds. This study was not powered for the analysis of this endpoint individually. It was pre-specified in the study protocol to combine the data from this study with study COMB157G2302 to address this endpoint. (NCT02792218)
Timeframe: Baseline, every 6 months up to 2.5 years

,
Interventionpercentage of participants (Number)
Month 18 - from Kaplan Meier estimatesMonth 24 - from Kaplan Meier estimates
OMB 20 mg2.92.9
TER 14 mg3.33.3

[back to top]

6-month Confirmed Disability Worsening (6mCDW) or 6-month Confirmed Cognitive Decline (6mCCD) - Pooled Data

A 6-month confirmed disability worsening (6mCDW) was defined as an increase from baseline in Expanded Disability Status Scale (EDSS) score sustained for at least 6 months. For patients with a baseline EDSS of 0, the criterion for disability worsening was an increase in EDSS of ≥1.5, for patients with a baseline EDSS of 1 to 5 or ≥5.5, the criterion for disability worsening was an increase in EDSS of ≥1 or ≥0.5, respectively. A 6-month confirmed cognitive decline (6mCCD) was defined as a 4-point worsening on Symbol Digit Modalities Test (SDMT) sustained for at least 6 months. This study was not powered for the analysis of this endpoint individually. It was pre-specified in the study protocol to combine the data from this study with study COMB157G2302 to address this endpoint. (NCT02792218)
Timeframe: Baseline up to 2.5 years

,
Interventionpercentage of participants (Number)
Month 18 - from Kaplan Meier estimatesMonth 24 - from Kaplan Meier estimates
OMB 20 mg20.521.4
TER 14 mg21.722.6

[back to top]

6-month Confirmed Disability Worsening (6mCDW) Based on EDSS > 8 Weeks After Onset of Treatment - Pooled Data

A 6-month confirmed disability worsening (6mCDW) was defined as an increase from baseline in Expanded Disability Status Scale (EDSS) score sustained for at least 6 months. For patients with a baseline EDSS of 0, the criterion for disability worsening was an increase in EDSS of ≥1.5, for patients with a baseline EDSS of 1 to 5 or ≥5.5, the criterion for disability worsening was an increase in EDSS of ≥1 or ≥0.5, respectively. This study was not powered for the analysis of this endpoint individually. It was pre-specified in the study protocol to combine the data from this study with study COMB157G2302 to address this endpoint. (NCT02792218)
Timeframe: Baseline up to 2.5 years

,
Interventionpercentage of participants (Mean)
Month 18 - from Kaplan Meier estimatesMonth 24 - from Kaplan Meier estimates
OMB 20 mg7.88.1
TER 14 mg10.712.0

[back to top]

6-month Confirmed Disability Worsening (6mCDW) Based on EDSS - Study COMB157G2301

A 6-month confirmed disability worsening (6mCDW) was defined as an increase from baseline in Expanded Disability Status Scale (EDSS) score sustained for at least 6 months. For patients with a baseline EDSS of 0, the criterion for disability worsening was an increase in EDSS of ≥1.5, for patients with a baseline EDSS of 1 to 5 or ≥5.5, the criterion for disability worsening was an increase in EDSS of ≥1 or ≥0.5, respectively. This study was not powered for the analysis of this endpoint individually. It was pre-specified in the study protocol to combine the data from this study with study COMB157G2302 to address this endpoint. (NCT02792218)
Timeframe: Baseline, every 3 months up to 2.5 years

,
Interventionpercentage of participants (Number)
Month 18- from Kaplan Meier estimatesMonth 24 - from Kaplan Meier estimates
OMB 20 mg7.58.2
TER 14 mg11.513.0

[back to top]

3-month Confirmed Disability Worsening (3mCDW) Based on EDSS > 8 Weeks After Onset of Treatment - Pooled Data

A 3-month confirmed disability worsening (3mCDW) was defined as an increase from baseline in Expanded Disability Status Scale (EDSS) score sustained for at least 3 months. For patients with a baseline EDSS of 0, the criterion for disability worsening was an increase in EDSS of ≥1.5, for patients with a baseline EDSS of 1 to 5 or ≥5.5, the criterion for disability worsening was an increase in EDSS of ≥1 or ≥0.5, respectively. This study was not powered for the analysis of this endpoint individually. It was pre-specified in the study protocol to combine the data from this study with study COMB157G2302 to address this endpoint. (NCT02792218)
Timeframe: Baseline up to 2.5 years

,
Interventionpercentage of participants (Mean)
Month 18 - from Kaplan Meier estimatesMonth 24 - from Kaplan Meier estimates
OMB 20 mg9.410.9
TER 14 mg13.515.0

[back to top]

6-month Confirmed Disability Worsening (6mCDW) Based on EDSS - Pooled Data

A 6-month confirmed disability worsening (6mCDW) was defined as an increase from baseline in Expanded Disability Status Scale (EDSS) score sustained for at least 6 months. For patients with a baseline EDSS of 0, the criterion for disability worsening was an increase in EDSS of ≥1.5, for patients with a baseline EDSS of 1 to 5 or ≥5.5, the criterion for disability worsening was an increase in EDSS of ≥1 or ≥0.5, respectively. This study was not powered for the analysis of this endpoint individually. It was pre-specified in the study protocol to combine the data from this study with study COMB157G2302 to address this endpoint. (NCT02792218)
Timeframe: Baseline, every 3 months up to 2.5 years

,
Interventionpercentage of participants (Number)
Month 18- from Kaplan Meier estimatesMonth 24 - from Kaplan Meier estimates
OMB 20 mg7.88.1
TER 14 mg10.712.0

[back to top]

6-month Confirmed Disability Improvement (6mCDI) Sustained Until End of Study (EOS) as Measured by EDSS - Pooled Data

A 6-month confirmed disability improvement (6mCDI) sustained until EOS was defined as a decrease from baseline EDSS sustained until EOS. For patients with a baseline EDSS of 0 to 1.5, no disability improvement was possible based on the protocol definition of an improvement; for patients with a baseline EDSS of ≥2 to 6 or ≥6.5 to 9.5, the criterion for disability improvement was a decrease in EDSS of ≤1 or ≤0.5, respectively. This study was not powered for the analysis of this endpoint individually. It was pre-specified in the study protocol to combine the data from this study with study COMB157G2302 to address this endpoint. (NCT02792218)
Timeframe: Baseline, every 3 months up to 2.5 years

,
Interventionpercentage of participants (Number)
Month 18 - from Kaplan Meier estimatesMonth 24 - from Kaplan Meier estimates
OMB 20 mg5.45.8
TER 14 mg4.64.6

[back to top]

6-month Confirmed Disability Improvement (6mCDI ) Based on EDSS - Study COMB157G2301

A 6-month confirmed disability improvement (6mCDI) was defined as a decrease from baseline EDSS sustained for at least 6 months. For patients with a baseline EDSS of 0 to 1.5, no disability improvement was possible based on the protocol definition of an improvement; for patients with a baseline EDSS of ≥2 to 6 or ≥6.5 to 9.5, the criterion for disability improvement was a decrease in EDSS of ≤1 or ≤0.5, respectively. This study was not powered for the analysis of this endpoint individually. It was pre-specified in the study protocol to combine the data from this study with study COMB157G2302 to address this endpoint. (NCT02792218)
Timeframe: Baseline, every 3 months up to 2.5 years

,
Interventionpercentage of participants (Mean)
Month 18 - from Kaplan Meier estimatesMonth 24 - from Kaplan Meier estimates
OMB 20 mg9.19.7
TER 14 mg7.18.2

[back to top]

6-month Confirmed Disability Improvement (6mCDI ) Based on EDSS - Pooled Data

A 6-month confirmed disability improvement (6mCDI) was defined as a decrease from baseline EDSS sustained for at least 6 months. For patients with a baseline EDSS of 0 to 1.5, no disability improvement was possible based on the protocol definition of an improvement; for patients with a baseline EDSS of ≥2 to 6 or ≥6.5 to 9.5, the criterion for disability improvement was a decrease in EDSS of ≤1 or ≤0.5, respectively. This study was not powered for the analysis of this endpoint individually. It was pre-specified in the study protocol to combine the data from this study with study COMB157G2302 to address this endpoint. (NCT02792218)
Timeframe: Baseline, every 3 months up to 2.5 years

,
Interventionpercentage of participants (Mean)
Month 18 - from Kaplan Meier estimatesMonth 24 - from Kaplan Meier estimates
OMB 20 mg10.111.0
TER 14 mg7.68.2

[back to top]

Pharmacokinetic (PK) Concentrations of Ofatumumab

Summary statistics of pharmacokinetic (PK) concentrations from trough samples collected within a 7-day window prior or at day of dosing. (NCT02792218)
Timeframe: Baseline, Weeks 4, 12, 24, 48, 96

Interventionug/mL (Mean)
Baseline n=304Week 4 n=347Week 12 n=323Week 24 n=304Week 48 n=270Week 96 n=336
OMB 20 mg0.002671.257460.226450.369910.512801.05314

[back to top]

Multiple Sclerosis Impact Scale (MSIS-29) Physical Impact Score Change From Baseline

"MSIS-29 is a 29-item, self-administered questionnaire that includes 2 domains, physical and psychological. Responses are captured on a 4-point scale ranging from not at all (1) to extremely (4), where higher scores reflect greater impact on day to day life." (NCT02792218)
Timeframe: Baseline, every 6 months up to 2.5 years

,
Interventionscores on a scale (Mean)
Month 6 n=455,451Month 12 n=438,428Month 18 n=428,406Month 24 n=262,233Month 30 n=86,65
OMB 20 mg-2.75-2.43-2.37-2.60-3.16
TER 14 mg-0.440.170.670.590.57

[back to top]

Change in Cognitive Performance Measured by the Symbol Digit Modalities Test (SDMT) - Pooled Data

Processing speed is being measured by the Symbol Digit Modalities Test (SDMT) score. SDMT measures the time to pair abstract symbols with specific numbers. The test requires visuoperceptual processing, working memory, and psychomotor speed. The score is the number of correctly coded items in 90 seconds. (max=110, min=0). Higher scores indicate improvement. Lower scores indicate worsening. (NCT02792218)
Timeframe: Baseline up to 2.5 years

,
Interventionscores (Mean)
Month 6 n=921,909Month 12 n=879, 863Month 18 n=849 ,808Month 24 n=492,468Month 30 n=156,117
OMB 20 mg1.021.822.843.502.97
TER 14 mg0.641.702.052.392.97

[back to top]

6-month Confirmed Cognitive Decline on Symbol Digit Modalities Test (SDMT) - Pooled Data

A 6-month confirmed cognitive decline was defined as a decrease from baseline of at least 4 points in SDMT score sustained for at least 6 months. Processing speed was measured by the Symbol Digit Modalities Test (SDMT) score. SDMT measures the time to pair abstract symbols with specific numbers. The test requires visuoperceptual processing, working memory, and psychomotor speed. The score is the number of correctly coded items in 90 seconds. (max=110, min=0). Higher scores indicate improvement. Lower scores indicate worsening. This study was not powered for the analysis of this endpoint individually. It was pre-specified in the study protocol to combine the data from this study with study COMB157G2302 to address this endpoint (NCT02792218)
Timeframe: Baseline, every 6 months up to 2.5 years

,
Interventionpercentage of participants (Number)
Month 18 - from Kaplan Meier estimatesMonth 24 - from Kaplan Meier estimates
OMB 20 mg14.315.4
TER 14 mg13.712.4

[back to top]

Percent Change in T2 Lesion Volume Relative to Baseline

Percent change from baseline in total T2 lesion volume (NCT02792218)
Timeframe: Baseline, Month 12, Month 24

,
Interventionpercentage change in lesion volume (Mean)
Month 12 n=436,430Month 24 n=332,324
OMB 20 mg-1.4-2.6
TER 14 mg9.713.5

[back to top]

Number of New or Enlarging T2 Lesions Per Year by NfL High-low Subgroups - Pooled Data

Number of new or enlarging T2 lesions on MRI per year (annualized lesion rate). (NCT02792218)
Timeframe: Baseline, yearly up to 2.5 years

,
InterventionT2 lesions per year (Mean)
NfL High: > 9.3 median n=432,402NfL Low: <=9.3 median n=418,421
OMB 20 mg0.950.39
TER 14 mg5.283.02

[back to top]

No Evidence of Disease Activity (NEDA-4)

NEDA-4 was defined as no 3-month confirmed disability worsening, no confirmed MS relapse, no new or enlarging T2 lesions compared to baseline, and the annualized rate of brain atrophy >-0.04%. (NCT02792218)
Timeframe: Baseline, Month 12, Month 24

,
Interventionpercentage of participants (Number)
Month 12 n=428,413Month 24 n=104,95
OMB 20 mg23.414.4
TER 14 mg14.83.2

[back to top]

Neurofilament Light Chain (NfL) Concentration in Serum

The NfL concentration (geometric mean concentration) was estimated by treatment and time point with using a repeated measures model on the basis of all evaluable log-transformed NfL values. (NCT02792218)
Timeframe: Month 3, 12 and 24

,
Interventionpg/mL (Geometric Mean)
Month 3 n=430,403Month 12 n=414,398Month 24 n=371,349
OMB 20 mg8.807.026.90
TER 14 mg9.419.638.99

[back to top]

Multiple Sclerosis Impact Scale (MSIS-29) Psychological Impact Score Change From Baseline

"MSIS-29 is a 29-item, self-administered questionnaire that includes 2 domains, physical and psychological. Responses are captured on a 4-point scale ranging from not at all (1) to extremely (4), where higher scores reflect greater impact on day to day life." (NCT02792218)
Timeframe: Baseline, every 6 months up to 2.5 years

,
Interventionscores on a scale (Mean)
Month 6 n=454,449Month 12 n=438,427Month 18 n=428,406Month 24 n=262,232Month 30 n=86,65
OMB 20 mg-5.20-5.22-5.72-6.14-7.78
TER 14 mg-3.07-2.57-3.94-3.93-0.85

[back to top]

3-month Confirmed Disability Worsening (3mCDW) Based on EDSS - Study COMB157G2301

A 3-month confirmed disability worsening (3mCDW) was defined as an increase from baseline in Expanded Disability Status Scale (EDSS) score sustained for at least 3 months. For patients with a baseline EDSS of 0, the criterion for disability worsening was an increase in EDSS of ≥1.5, for patients with a baseline EDSS of 1 to 5 or ≥5.5, the criterion for disability worsening was an increase in EDSS of ≥1 or ≥0.5, respectively. This study was not powered for the analysis of this endpoint individually. It was pre-specified in the study protocol to combine the data from this study with study COMB157G2302 to address this endpoint. (NCT02792218)
Timeframe: Baseline, every 3 months up to 2.5 years

,
Interventionpercentage of participants (Number)
Month 18 - from Kaplan Meier estimatesMonth 24 - from Kaplan Meier estimates
OMB 20 mg9.411.3
TER 14 mg13.915.4

[back to top]

3-month Confirmed Disability Worsening (3mCDW) Based on EDSS - Pooled Data

A 3-month confirmed disability worsening (3mCDW) was defined as an increase from baseline in Expanded Disability Status Scale (EDSS) score sustained for at least 3 months. For patients with a baseline EDSS of 0, the criterion for disability worsening was an increase in EDSS of ≥1.5, for patients with a baseline EDSS of 1 to 5 or ≥5.5, the criterion for disability worsening was an increase in EDSS of ≥1 or ≥0.5, respectively. This study was not powered for the analysis of this endpoint individually. It was pre-specified in the study protocol to combine the data from this study with study COMB157G2302 to address this endpoint. (NCT02792218)
Timeframe: Baseline, every 3 months up to 2.5 years

,
Interventionpercentage of participants (Number)
Month 18 - from Kaplan Meier estimatesMonth 24 - from Kaplan Meier estimates
OMB 20 mg9.410.9
TER 14 mg13.515.0

[back to top]

Percentage of Participants With Confirmed Relapse

A confirmed MS relapse was defined as one accompanied by a clinically-relevant change in the EDSS performed by the Independent EDSS rater, i.e. an increase of at least 0.5 points on the EDSS score, or an increase of 1 point on two functional scores or 2 points on one functional score (excluding changes involving bowel/bladder or cerebral functional system). (NCT02792218)
Timeframe: Baseline up to 2.5 years

Interventionpercentage of participants (Number)
OMB 20 mg18.82
TER 14 mg32.73

[back to top]

Number of New or Enlarging T2 Lesions on MRI Per Year From Month 12 Until End of Study (EOS)

Number of new/enlarging T2 lesions on the last available MRI scan compared to Month 12 adjusted for different time of scans versus Month 12 due to variable follow up time in study. (NCT02792218)
Timeframe: Month 12 up to 2.5 years

InterventionT2 lesions per year (Mean)
OMB 20 mg0.05
TER 14 mg3.73

[back to top]

Number of Gd-enhancing T1 Lesions Per MRI Scan

Total number of Gd-enhancing T1 lesions across all scans per patient adjusted for different number of scans due to variable follow-up time in study. (NCT02792218)
Timeframe: Baseline, yearly up to 2.5 years

Interventionlesions per scan (Mean)
OMB 20 mg0.0115
TER 14 mg0.4555

[back to top]

Annualized Relapse Rate (ARR) >8 Weeks After Onset of Treatment

ARR was the number of confirmed relapses in a year, calculated as the total number of relapses for all participants in the treatment group divided by the total participant-years of time in study. A confirmed MS relapse was defined as one accompanied by a clinically-relevant change in the EDSS performed by the Independent EDSS rater, i.e. an increase of at least 0.5 points on the EDSS score, or an increase of 1 point on two functional scores or 2 points on one functional score (excluding changes involving bowel/bladder or cerebral functional system). Comparisons were made to the previous rating (the last EDSS rating that did not occur during a relapse). (NCT02792218)
Timeframe: Baseline up to 2.5 years

Interventionnumber of relapses in a year (Mean)
OMB 20 mg0.096
TER 14 mg0.242

[back to top]

Number of New or Enlarging T2 Lesions on MRI Per Year (Annualized Lesion Rate)

Number of new/enlarging T2 lesions on last available MRI scan compared to baseline adjusted for different time of scans versus baseline due to variable follow up time in study (NCT02792218)
Timeframe: Baseline, yearly up to 2.5 years

,
InterventionT2 lesions per year (Mean)
Month 12 n=420,407Month 24 n=103,93EOS n=440,431
OMB 20 mg1.130.720.72
TER 14 mg4.303.214.00

[back to top]

Annualized Relapse Rate (ARR)

ARR was the number of confirmed relapses in a year, calculated as the total number of relapses for all participants in the treatment group divided by the total participant-years of time in study. A confirmed MS relapse was defined as one accompanied by a clinically-relevant change in the EDSS performed by the Independent EDSS rater, i.e. an increase of at least 0.5 points on the EDSS score, or an increase of 1 point on two functional scores or 2 points on one functional score (excluding changes involving bowel/bladder or cerebral functional system). Comparisons were made to the previous rating (the last EDSS rating that did not occur during a relapse). (NCT02792218)
Timeframe: Baseline up to 2.5 years

Interventionnumber of relapses in a year (Mean)
OMB 20 mg0.11
TER 14 mg0.22

[back to top]

Annualized Rate of Brain Volume Loss Based on Assessments of Percent Brain Volume Change From Baseline

Percent change from baseline in brain volume loss (BVL) on all MRI scans adjusted for different time of scan versus baseline due to variable follow up time in study (NCT02792218)
Timeframe: Baseline, months 12 and 24

Interventionpercentage of brain volume loss (Mean)
OMB 20 mg-0.28
TER 14 mg-0.35

[back to top]

Number of Gadolinium-enhancing T1 Lesions Per MRI Scan

Total number of Gd-enhancing T1 lesions across all scans per patient adjusted for different number of scans due to variable follow-up time in study. (NCT02792231)
Timeframe: Baseline, yearly up to 2.5 years

Interventionlesions per scan (Mean)
OMB 20 mg0.0317
TER 14 mg0.5172

[back to top]

Annualized Relapse Rate (ARR) >8 Weeks After Onset of Treatment

ARR was the number of confirmed relapses in a year, calculated as the total number of relapses for all participants in the treatment group divided by the total participant-years of time in study. A confirmed MS relapse was defined as one accompanied by a clinically-relevant change in the EDSS performed by the Independent EDSS rater, i.e. an increase of at least 0.5 points on the EDSS score, or an increase of 1 point on two functional scores or 2 points on one functional score (excluding changes involving bowel/bladder or cerebral functional system). Comparisons were made to the previous rating (the last EDSS rating that did not occur during a relapse). (NCT02792231)
Timeframe: Baseline up to 2.5 years

Interventionnumber of relapses in a year (Mean)
OMB 20 mg0.096
TER 14 mg0.241

[back to top]

Annualized Relapse Rate (ARR)

ARR was the number of confirmed relapses in a year, calculated as the total number of relapses for all participants in the treatment group divided by the total participant-years of time in study. A confirmed MS relapse was defined as one accompanied by a clinically-relevant change in the EDSS performed by the Independent EDSS rater, i.e. an increase of at least 0.5 points on the EDSS score, or an increase of 1 point on two functional scores or 2 points on one functional score (excluding changes involving bowel/bladder or cerebral functional system). Comparisons were made to the previous rating (the last EDSS rating that did not occur during a relapse). (NCT02792231)
Timeframe: Baseline up to 2.5 years

Interventionnumber of relapses in a year (Mean)
OMB 20 mg0.10
TER 14 mg0.25

[back to top]

Annualized Rate of Brain Volume Loss Based on Assessments of Percent Brain Volume Change From Baseline

Percent change from baseline in brain volume loss (BVL) on all MRI scans adjusted for different time of scan versus baseline due to variable follow up time in study (NCT02792231)
Timeframe: Baseline, Months 12 and 24

Interventionpercentage of brain volume loss (Mean)
OMB 20 mg-0.29
TER 14 mg-0.35

[back to top]

Number of New or Enlarging T2 Lesions on MRI Per Year From Month 12 Until End of Study (EOS)

Number of new/enlarging T2 lesions on the last available MRI scan compared to Month 12 adjusted for different time of scans versus Month 12 due to variable follow up time in study. (NCT02792231)
Timeframe: Month 12 up to 2.5 years

InterventionT2 lesions per year (Mean)
OMB 20 mg0.13
TER 14 mg3.84

[back to top]

6-month Confirmed Disability Worsening (6mCDW) Based on EDSS - Study COMB157G2302

A 6-month confirmed disability worsening (6mCDW) was defined as an increase from baseline in Expanded Disability Status Scale (EDSS) score sustained for at least 6 months. For patients with a baseline EDSS of 0, the criterion for disability worsening was an increase in EDSS of ≥1.5, for patients with a baseline EDSS of 1 to 5 or ≥5.5, the criterion for disability worsening was an increase in EDSS of ≥1 or ≥0.5, respectively. This study was not powered for the analysis of this endpoint individually. It was pre-specified in the study protocol to combine the data from this study with study COMB157G2301 to address this endpoint. (NCT02792231)
Timeframe: Baseline, every 3 months up to 2.5 years

,
Interventionpercentage of participants (Number)
Month 18- from Kaplan Meier estimatesMonth 24 - from Kaplan Meier estimates
OMB 20 mg8.08.0
TER 14 mg10.010.9

[back to top]

6-month Confirmed Disability Worsening (6mCDW) Based on EDSS - Pooled Data

A 6-month confirmed disability worsening (6mCDW) was defined as an increase from baseline in Expanded Disability Status Scale (EDSS) score sustained for at least 6 months. For patients with a baseline EDSS of 0, the criterion for disability worsening was an increase in EDSS of ≥1.5, for patients with a baseline EDSS of 1 to 5 or ≥5.5, the criterion for disability worsening was an increase in EDSS of ≥1 or ≥0.5, respectively. This study was not powered for the analysis of this endpoint individually. It was pre-specified in the study protocol to combine the data from this study with study COMB157G2301 to address this endpoint. (NCT02792231)
Timeframe: Baseline, every 3 months up to 2.5 years

,
Interventionpercentage of participants (Number)
Month 18- from Kaplan Meier estimatesMonth 24 - from Kaplan Meier estimates
OMB 20 mg7.88.1
TER 14 mg10.712.0

[back to top]

6-month Confirmed Disability Improvement (6mCDI) Sustained Until End of Study (EOS) as Measured by EDSS - Pooled Data

A 6-month confirmed disability improvement (6mCDI) sustained until EOS was defined as a decrease from baseline EDSS sustained until EOS. For patients with a baseline EDSS of 0 to 1.5, no disability improvement was possible based on the protocol definition of an improvement; for patients with a baseline EDSS of ≥2 to 6 or ≥6.5 to 9.5, the criterion for disability improvement was a decrease in EDSS of ≤1 or ≤0.5, respectively. This study was not powered for the analysis of this endpoint individually. It was pre-specified in the study protocol to combine the data from this study with study COMB157G2301 to address this endpoint. (NCT02792231)
Timeframe: Baseline, every 3 months up to 2.5 years

,
Interventionpercentage of participants (Number)
Month 18 - from Kaplan Meier estimatesMonth 24 - from Kaplan Meier estimates
OMB 20 mg5.45.8
TER 14 mg4.64.6

[back to top]

6-month Confirmed Disability Improvement (6mCDI ) Based on EDSS - Study COMB157G2302

A 6-month confirmed disability improvement (6mCDI) was defined as a decrease from baseline EDSS sustained for at least 6 months. For patients with a baseline EDSS of 0 to 1.5, no disability improvement was possible based on the protocol definition of an improvement; for patients with a baseline EDSS of ≥2 to 6 or ≥6.5 to 9.5, the criterion for disability improvement was a decrease in EDSS of ≤1 or ≤0.5, respectively. This study was not powered for the analysis of this endpoint individually. It was pre-specified in the study protocol to combine the data from this study with study COMB157G2301 to address this endpoint. (NCT02792231)
Timeframe: Baseline, every 3 months up to 2.5 years

,
Interventionpercentage of participants (Number)
Month 18 - from Kaplan Meier estimatesMonth 24 - from Kaplan Meier estimates
OMB 20 mg11.112.3
TER 14 mg8.18.1

[back to top]

6-month Confirmed Disability Improvement (6mCDI ) Based on EDSS - Pooled Data

A 6-month confirmed disability improvement (6mCDI) was defined as a decrease from baseline EDSS sustained for at least 6 months. For patients with a baseline EDSS of 0 to 1.5, no disability improvement was possible based on the protocol definition of an improvement; for patients with a baseline EDSS of ≥2 to 6 or ≥6.5 to 9.5, the criterion for disability improvement was a decrease in EDSS of ≤1 or ≤0.5, respectively. This study was not powered for the analysis of this endpoint individually. It was pre-specified in the study protocol to combine the data from this study with study COMB157G2301 to address this endpoint. (NCT02792231)
Timeframe: Baseline, every 3 months up to 2.5 years

,
Interventionpercentage of participants (Number)
Month 18 - from Kaplan Meier estimatesMonth 24 - from Kaplan Meier estimates
OMB 20 mg10.111.0
TER 14 mg7.68.2

[back to top]

6-month Confirmed Cognitive Decline on Symbol Digit Modalities Test (SDMT) - Pooled Data

A 6-month confirmed cognitive decline was defined as a decrease from baseline of at least 4 points in SDMT score sustained for at least 6 months. Processing speed was measured by the Symbol Digit Modalities Test (SDMT) score. SDMT measures the time to pair abstract symbols with specific numbers. The test requires visuoperceptual processing, working memory, and psychomotor speed. The score is the number of correctly coded items in 90 seconds. (max=110, min=0). Higher scores indicate improvement. Lower scores indicate worsening. This study was not powered for the analysis of this endpoint individually. It was pre-specified in the study protocol to combine the data from this study with study COMB157G2301 to address this endpoint (NCT02792231)
Timeframe: Baseline, every 6 months up to 2.5 years

,
Interventionpercentage of participants (Number)
Month 18 - from Kaplan Meier estimatesMonth 24 - from Kaplan Meier estimates
OMB 20 mg14.315.4
TER 14 mg13.714.0

[back to top]

3-month Confirmed Disability Worsening (3mCDW) Based on EDSS > 8 Weeks After Onset of Treatment - Pooled Data

A 3-month confirmed disability worsening (3mCDW) was defined as an increase from baseline in Expanded Disability Status Scale (EDSS) score sustained for at least 3 months. For patients with a baseline EDSS of 0, the criterion for disability worsening was an increase in EDSS of ≥1.5, for patients with a baseline EDSS of 1 to 5 or ≥5.5, the criterion for disability worsening was an increase in EDSS of ≥1 or ≥0.5, respectively. This study was not powered for the analysis of this endpoint individually. It was pre-specified in the study protocol to combine the data from this study with study COMB157G2301 to address this endpoint. (NCT02792231)
Timeframe: Baseline up to 2.5 years

,
Interventionpercentage of participants (Number)
Month 18 - from Kaplan Meier estimatesMonth 24 - from Kaplan Meier estimates
OMB 20 mg9.410.9
TER 14 mg13.515.0

[back to top]

3-month Confirmed Disability Worsening (3mCDW) Based on EDSS - Study COMB157G2302

A 3-month confirmed disability worsening (3mCDW) was defined as an increase from baseline in Expanded Disability Status Scale (EDSS) score sustained for at least 3 months. For patients with a baseline EDSS of 0, the criterion for disability worsening was an increase in EDSS of ≥1.5, for patients with a baseline EDSS of 1 to 5 or ≥5.5, the criterion for disability worsening was an increase in EDSS of ≥1 or ≥0.5, respectively. This study was not powered for the analysis of this endpoint individually. It was pre-specified in the study protocol to combine the data from this study with study COMB157G2301 to address this endpoint. (NCT02792231)
Timeframe: Baseline, every 3 months up to 2.5 years

,
Interventionpercentage of participants (Number)
Month 18 - from Kaplan Meier estimatesMonth 24 - from Kaplan Meier estimates
OMB 20 mg9.310.5
TER 14 mg13.214.6

[back to top]

3-month Confirmed Disability Worsening (3mCDW) Based on EDSS - Pooled Data

A 3-month confirmed disability worsening (3mCDW) was defined as an increase from baseline in Expanded Disability Status Scale (EDSS) score sustained for at least 3 months. For patients with a baseline EDSS of 0, the criterion for disability worsening was an increase in EDSS of ≥1.5, for patients with a baseline EDSS of 1 to 5 or ≥5.5, the criterion for disability worsening was an increase in EDSS of ≥1 or ≥0.5, respectively. This study was not powered for the analysis of this endpoint individually. It was pre-specified in the study protocol to combine the data from this study with study COMB157G2301 to address this endpoint. (NCT02792231)
Timeframe: Baseline, every 3 months up to 2.5 years

,
Interventionpercentage of participants (Number)
Month 18 - from Kaplan Meier estimatesMonth 24 - from Kaplan Meier estimates
OMB 20 mg9.410.9
TER 14 mg13.515.0

[back to top]

Participants With Confirmed Relapse

A confirmed MS relapse was defined as one accompanied by a clinically-relevant change in the EDSS performed by the Independent EDSS rater, i.e. an increase of at least 0.5 points on the EDSS score, or an increase of 1 point on two functional scores or 2 points on one functional score (excluding changes involving bowel/bladder or cerebral functional system). (NCT02792231)
Timeframe: Baseline up to 2.5 years

Interventionpercentage of participants (Number)
OMB 20 mg16.51
TER 14 mg32.68

[back to top]

Pharmacokinetic (PK) Concentrations of Ofatumumab

Summary statistics of pharmacokinetic (PK) concentrations from trough samples collected within a 7-day window prior or at day of dosing. (NCT02792231)
Timeframe: Baseline, Weeks 4, 12, 24, 48, 96

Interventionug/mL (Mean)
Baseline n=325Week 4 n=346Week 12 n=257Week 24 n=243Week 48 n=240Week 96 n=304
OMB 20 mg0.003251.265120.209320.382030.590871.13218

[back to top]

Percent Change in T2 Lesion Volume Relative to Baseline

Percent change from baseline in total T2 lesion volume (NCT02792231)
Timeframe: Baseline, Month 12, Month 24

,
Interventionpercentage change in lesion volume (Mean)
Month 12 n=447,437Month 24 n=330,320
OMB 20 mg-2.4-2.6
TER 14 mg10.117.8

[back to top]

Number of New or Enlarging T2 Lesions Per Year by NfL High-low Subgroups - Pooled Data

Number of new or enlarging T2 lesions on MRI per year (annualized lesion rate). (NCT02792231)
Timeframe: Baseline, yearly up to 2.5 years

,
InterventionT2 lesions per year (Mean)
High > median n=432,402Low <= median n=418,421
OMB 20 mg0.950.39
TER 14 mg5.283.02

[back to top]

Number of New or Enlarging T2 Lesions on MRI Per Year (Annualized Lesion Rate)

Number of new/enlarging T2 lesions on last available MRI scan compared to baseline adjusted for different time of scans versus baseline due to variable follow up time in study (NCT02792231)
Timeframe: Baseline, yearly up to 2.5 years

,
InterventionT2 lesions per year (Mean)
Month 12 n=422,410Month 24 n=90,76EOS n=448,442
OMB 20 mg0.940.720.64
TER 14 mg4.413.724.16

[back to top]

No Evidence of Disease Activity (NEDA-4)

NEDA-4 was defined as no 3-month confirmed disability worsening, no confirmed MS relapse, no new or enlarging T2 lesions compared to baseline, and the annualized rate of brain atrophy >-0.04%. (NCT02792231)
Timeframe: Baseline, Month 12, Month 24

,
Interventionpercentage of participants (Number)
Month 12 n=433,427Month 24 n=92,78
OMB 20 mg23.89.8
TER 14 mg17.85.1

[back to top]

Neurofilament Light Chain (NfL) Concentration in Serum

The NfL concentration (geometric mean concentration) was estimated by treatment and time point with using a repeated measures model on the basis of all evaluable log-transformed NfL values. (NCT02792231)
Timeframe: Month 3, 12 and 24

,
Interventionpg/mL (Geometric Mean)
Month 3 n=425,423Month 12 n=406,406Month 24 n=345,349
OMB 20 mg8.927.066.80
TER 14 mg10.029.538.99

[back to top]

Multiple Sclerosis Impact Scale (MSIS-29) Psychological Impact Score Change From Baseline

"MSIS-29 is a 29-item, self-administered questionnaire that includes 2 domains, physical and psychological. Responses are captured on a 4-point scale ranging from not at all (1) to extremely (4), where higher scores reflect greater impact on day to day life." (NCT02792231)
Timeframe: Baseline, every 6 months up to 2.5 years

,
Interventionscores on a scale (Mean)
Month 6 n=473,461Month 12 n=448,436Month 18 n=423,409Month 24 n=234,238Month 30 n=70,54
OMB 20 mg-5.96-5.42-6.23-6.10-6.25
TER 14 mg-3.77-3.88-2.51-3.12-4.75

[back to top]

Multiple Sclerosis Impact Scale (MSIS-29) Physical Impact Score Change From Baseline

"MSIS-29 is a 29-item, self-administered questionnaire that includes 2 domains, physical and psychological. Responses are captured on a 4-point scale ranging from not at all (1) to extremely (4), where higher scores reflect greater impact on day to day life." (NCT02792231)
Timeframe: Baseline, every 6 months up to 2.5 years

,
Interventionscores on a scale (Mean)
Month 6 n=473,461Month 12 n=448,438Month 18 n=425,409Month 24 n=235,238Month 30 n=70,54
OMB 20 mg-2.20-2.47-2.29-2.93-2.49
TER 14 mg-0.46-0.491.530.621.44

[back to top]

Change in Cognitive Performance Measured by the Symbol Digit Modalities Test (SDMT) - Pooled Data

Processing speed is being measured by the Symbol Digit Modalities Test (SDMT) score. SDMT measures the time to pair abstract symbols with specific numbers. The test requires visuoperceptual processing, working memory, and psychomotor speed. The score is the number of correctly coded items in 90 seconds. (max=110, min=0). Higher scores indicate improvement. Lower scores indicate worsening. This study was not powered for the analysis of this endpoint individually. It was pre-specified in the study protocol to combine the data from this study with study COMB157G2301 to address this endpoint. (NCT02792231)
Timeframe: Baseline up to 2.5 years

,
Interventionscores (Mean)
Month 6 n=921,909Month 12 n=879,863Month 18 n=849,808Month 24 n=492,468Month 30 n=156,117
OMB 20 mg1.021.822.843.503.53
TER 14 mg0.641.702.052.392.97

[back to top]

Brain Volume Loss by NfL High-low Subgroups - Pooled Data

Percent change from baseline in brain volume loss (BVL) on all MRI scans adjusted for different time of scan versus baseline due to variable follow up time in study. (NCT02792231)
Timeframe: Baseline, Months 12 and 24

,
Interventionpercentage of brain volume loss (Mean)
High > median n=416,387Low <= median n=403,407
OMB 20 mg-0.32-0.24
TER 14 mg-0.43-0.29

[back to top]

Annualized Relapse Rates (ARR) by NfL High-low Subgroups - Pooled Data

ARR was the number of confirmed relapses in a year, calculated as the total number of relapses for all participants in the treatment group divided by the total participant-years of time in study. A confirmed MS relapse was defined as one accompanied by a clinically-relevant change in the EDSS performed by the Independent EDSS rater, i.e. an increase of at least 0.5 points on the EDSS score, or an increase of 1 point on two functional scores or 2 points on one functional score (excluding changes involving bowel/bladder or cerebral functional system). (NCT02792231)
Timeframe: Baseline up to 2.5 years

,
Interventionnumber of relapses in a year (Mean)
High > median n=443,410Low <= median n=428,431
OMB 20 mg0.080.12
TER 14 mg0.210.23

[back to top]

6-month Confirmed Worsening of at Least 20% in the Timed 25-Foot Walk (T25FW) - Pooled Data

The patient is directed to walk 25 feet quickly and safely as possible from one marked end to the other. The time is calculated from the initiation of the patient instructed to begin, until the patient has reached the 25-foot mark. This study was not powered for the analysis of this endpoint individually. It was pre-specified in the study protocol to combine the data from this study with study COMB157G2301 to address this endpoint. (NCT02792231)
Timeframe: Baseline, every 3 months up to 2.5 years

,
Interventionpercentage of participants (Number)
Month 18 - from Kaplan Meier estimatesMonth 24 - from Kaplan Meier estimates
OMB 20 mg11.011.4
TER 14 mg10.410.6

[back to top]

6-month Confirmed Worsening of at Least 20% in the 9-Hole Peg Test (9HPT) - Pooled Data

9-Hole Peg Test is a test of upper limb function. Participants place 9 pegs on pegboard and remove pegs and this is timed for each hand. Time recorded in seconds. Longer time indicates poorer upper limb function. 20% improvement is defined as 20% shorter time in seconds. This study was not powered for the analysis of this endpoint individually. It was pre-specified in the study protocol to combine the data from this study with study COMB157G2301 to address this endpoint. (NCT02792231)
Timeframe: Baseline, every 6 months up to 2.5 years

,
Interventionpercentage of participants (Number)
Month 18 - from Kaplan Meier estimatesMonth 24 - from Kaplan Meier estimates
OMB 20 mg2.92.9
TER 14 mg3.33.3

[back to top]

6-month Confirmed Disability Worsening (6mCDW) or 6-month Confirmed Cognitive Decline (6mCCD) - Pooled Data

A 6-month confirmed disability worsening (6mCDW) was defined as an increase from baseline in Expanded Disability Status Scale (EDSS) score sustained for at least 6 months. For patients with a baseline EDSS of 0, the criterion for disability worsening was an increase in EDSS of ≥1.5, for patients with a baseline EDSS of 1 to 5 or ≥5.5, the criterion for disability worsening was an increase in EDSS of ≥1 or ≥0.5, respectively. A 6-month confirmed cognitive decline (6mCCD) was defined as a 4-point worsening on Symbol Digit Modalities Test (SDMT) sustained for at least 6 months. This study was not powered for the analysis of this endpoint individually. It was pre-specified in the study protocol to combine the data from this study with study COMB157G2301 to address this endpoint. (NCT02792231)
Timeframe: Baseline up to 2.5 years

,
Interventionpercentage of participants (Number)
Month 18 - from Kaplan Meier estimatesMonth 24 - from Kaplan Meier estimates
OMB 20 mg20.521.4
TER 14 mg21.722.6

[back to top]

6-month Confirmed Disability Worsening (6mCDW) Based on EDSS > 8 Weeks After Onset of Treatment - Pooled Data

A 6-month confirmed disability worsening (6mCDW) was defined as an increase from baseline in Expanded Disability Status Scale (EDSS) score sustained for at least 6 months. For patients with a baseline EDSS of 0, the criterion for disability worsening was an increase in EDSS of ≥1.5, for patients with a baseline EDSS of 1 to 5 or ≥5.5, the criterion for disability worsening was an increase in EDSS of ≥1 or ≥0.5, respectively. This study was not powered for the analysis of this endpoint individually. It was pre-specified in the study protocol to combine the data from this study with study COMB157G2301 to address this endpoint. (NCT02792231)
Timeframe: Baseline up to 2.5 years

,
Interventionpercentage of participants (Number)
Month 18- from Kaplan Meier estimatesMonth 24 - from Kaplan Meier estimates
OMB 20 mg7.88.1
TER 14 mg10.712.0

[back to top]

Annualized Relapse Rate (ARR)

ARR is defined as the number of Independent Relapse Adjudication Committee (IRAP)-confirmed relapses per participant year. The estimate of ARR for a treatment group is the total number of relapses for participants in the respective treatment group divided by the sum of treatment duration for participants in that specific treatment group. (NCT03277248)
Timeframe: Up to 96 weeks

Interventionrelapses per participant-years (Least Squares Mean)
Ublituximab + Oral Placebo0.091
Teriflunomide + IV Placebo0.178

[back to top]

Percent Change From Baseline in Brain Volume

(NCT03277248)
Timeframe: Baseline to Week 96

Interventionpercent change (Least Squares Mean)
Ublituximab + Oral Placebo-0.194
Teriflunomide + IV Placebo-0.176

[back to top]

Percentage of Participants With Impaired Symbol Digit Modalities Test (SDMT)

The SDMT involves a simple substitution task using a reference key, the examinee has 90 seconds to pair specific numbers with given geometric figures. Responses are done verbally. The administration time is approximately 5 minutes. The total SDMT score for each visit ranging from 0-110 is defined as the total number of correct answers reported in the case report form (CRF), where high scores indicate better outcome. Impaired SDMT is defined as a decrease of at least 4 points from baseline at any post-baseline assessment up to the Week 96 visit. (NCT03277248)
Timeframe: Baseline to Week 96

Interventionpercentage of participants (Number)
Ublituximab + Oral Placebo29.0
Teriflunomide + IV Placebo31.6

[back to top]

Percentage of Participants With No Evidence of Disease Activity (NEDA)

A participant with NEDA is defined as a participant without relapses confirmed by the IRAP, without MRI activities (no T1 Gd+ lesions and no new/enlarging T2 lesions), and no 12-week CDP. Any evidence of disease activity from Week 24 to Week 96 was counted as not reaching NEDA. Any evidence of disease activity before Week 24 was not counted. (NCT03277248)
Timeframe: From Week 24 to Week 96

Interventionpercentage of participants (Number)
Ublituximab + Oral Placebo43.0
Teriflunomide + IV Placebo11.4

[back to top]

Time to Confirmed Disability Progression (CDP) for at Least 12 Weeks

12-week CDP is defined as an increase in EDSS at least 1 point higher than the baseline EDSS if the baseline EDSS is ≤5.5 or at least 0.5 higher than the baseline EDSS if the baseline EDSS is >5.5. The EDSS is based on a standard neurological examination, (pyramidal, cerebellar, brainstem, sensory, bowel and bladder, visual, and cerebral) and ambulation function system assessments. The EDSS disability scale ranges in 0.5-point steps from 0 (normal) to 10 (death) where higher scores indicate disability. The time to onset of 12-week CDP is the time to progression to the EDSS change defined above. (NCT03277248)
Timeframe: Up to Week 96

Interventionweeks (Median)
Ublituximab + Oral PlaceboNA
Teriflunomide + IV PlaceboNA

[back to top]

Total Number of Gadolinium (Gd)-Enhancing T1-Lesions Per Magnetic Resonance Imaging (MRI) Scan Per Participant

The total number of Gd-enhancing T1-lesions were calculated as the sum of the individual number of lesions at Weeks 12, 24, 48, and 96, divided by the total number of MRI scans of the brain. (NCT03277248)
Timeframe: Weeks 12, 24, 48, and 96

Interventionlesions per scan per participant (Least Squares Mean)
Ublituximab + Oral Placebo0.009
Teriflunomide + IV Placebo0.250

[back to top]

Total Number of New and Enlarging T2 Hyperintense Lesions (NELs) Per MRI Scan Per Participant

The total number of NELs were calculated as the sum of the individual number of lesions at Weeks 24, 48, and 96, divided by the total number of MRI scans of the brain. (NCT03277248)
Timeframe: Weeks 24, 48, and 96

Interventionlesions per scan per participant (Least Squares Mean)
Ublituximab + Oral Placebo0.282
Teriflunomide + IV Placebo2.831

[back to top]

Percentage of Participants With Treatment-Emergent Adverse Events (TEAEs) and Treatment-Emergent Serious Adverse Events (TESAEs)

An adverse event (AE) is any unfavorable and unintended sign (including an abnormal laboratory finding, for example), symptom, or disease temporarily associated with the use of a medicinal product, whether or not considered related to the medicinal product. A serious AE is defined as any untoward medical occurrence that: results in death, is immediately life-threatening, requires in-patient hospitalization or prolongation of existing hospitalization, results in persistent or significant disability/incapacity, and/or causes a congenital anomaly/birth defect. TEAEs are AEs that start or worsen after receiving the study drug. (NCT03277248)
Timeframe: From the first dose of study drug through the end of the study (up to approximately 116 weeks)

,
Interventionpercentage of participants (Number)
TEAEsTESAEs
Teriflunomide + IV Placebo93.87.7
Ublituximab + Oral Placebo92.310.3

[back to top]

Time to Confirmed Disability Progression (CDP) for at Least 12 Weeks

12-week CDP is defined as an increase in EDSS at least 1 point higher than the baseline EDSS if the baseline EDSS is ≤5.5 or at least 0.5 higher than the baseline EDSS if the baseline EDSS is >5.5. The EDSS is based on a standard neurological examination, (pyramidal, cerebellar, brainstem, sensory, bowel and bladder, visual, and cerebral) and ambulation function system assessments. The EDSS disability scale ranges in 0.5-point steps from 0 (normal) to 10 (death) where higher scores indicate disability. The time to onset of 12-week CDP is the time to progression to the EDSS change defined above. (NCT03277261)
Timeframe: Up to Week 96

Interventionweeks (Median)
Ublituximab + Oral PlaceboNA
Teriflunomide + IV PlaceboNA

[back to top]

Total Number of Gadolinium (Gd)-Enhancing T1-Lesions Per Magnetic Resonance Imaging (MRI) Scan Per Participant

The total number of Gd-enhancing T1-lesions were calculated as the sum of the individual number of lesions at Weeks 12, 24, 48, and 96, divided by the total number of MRI scans of the brain. (NCT03277261)
Timeframe: Weeks 12, 24, 48, and 96

Interventionlesions per scan per participant (Least Squares Mean)
Ublituximab + Oral Placebo0.016
Teriflunomide + IV Placebo0.491

[back to top]

Total Number of New and Enlarging T2 Hyperintense Lesions (NELs) Per MRI Scan Per Participant

The total number of NELs were calculated as the sum of the individual number of lesions at Weeks 24, 48, and 96, divided by the total number of MRI scans of the brain. (NCT03277261)
Timeframe: Weeks 24, 48, and 96

Interventionlesions per scan per participant (Least Squares Mean)
Ublituximab + Oral Placebo0.213
Teriflunomide + IV Placebo2.789

[back to top]

Annualized Relapse Rate (ARR)

ARR is defined as the number of Independent Relapse Adjudication Panel (IRAP)-confirmed relapses per participant year. The estimate of ARR for a treatment group is the total number of relapses for participants in the respective treatment group divided by the sum of treatment duration for participants in that specific treatment group. (NCT03277261)
Timeframe: Up to 96 weeks

Interventionrelapses per participant-years (Least Squares Mean)
Ublituximab + Oral Placebo0.076
Teriflunomide + IV Placebo0.188

[back to top]

Percent Change From Baseline in Brain Volume

(NCT03277261)
Timeframe: Baseline up to Week 96

Interventionpercent change (Least Squares Mean)
Ublituximab + Oral Placebo-0.197
Teriflunomide + IV Placebo-0.125

[back to top]

Percentage of Participants With Impaired Symbol Digit Modalities Test (SDMT)

The SDMT involves a simple substitution task using a reference key, the examinee has 90 seconds to pair specific numbers with given geometric figures. Responses are done verbally. The administration time is approximately 5 minutes. The total SDMT score for each visit ranging from 0-110 is defined as the total number of correct answers reported in the case report form (CRF), where high scores indicate better outcome. Impaired SDMT is defined as a decrease from baseline of at least 4 points at any post-baseline assessment up to the Week 96 visit. (NCT03277261)
Timeframe: Baseline up to Week 96

Interventionpercentage of participants (Number)
Ublituximab + Oral Placebo29.2
Teriflunomide + IV Placebo31.8

[back to top]

Percentage of Participants With No Evidence of Disease Activity (NEDA)

A participant with NEDA is defined as a participant without relapses confirmed by the IRAP, without MRI activities (no T1 Gd+ lesions and no new/enlarging T2 lesions), and no 12-week CDP. Any evidence of disease activity from Week 24 to Week 96 was counted as not reaching NEDA. Any evidence of disease activity before Week 24 was not counted. (NCT03277261)
Timeframe: Week 24 up to Week 96

Interventionpercentage of participants (Number)
Ublituximab + Oral Placebo44.6
Teriflunomide + IV Placebo15.0

[back to top]

Percentage of Participants With Treatment Emergent Adverse Events (TEAEs) and Treatment Emergent Serious Adverse Events (TESAEs)

An adverse event (AE) is any unfavorable and unintended sign (including an abnormal laboratory finding, for example), symptom, or disease temporarily associated with the use of a medicinal product, whether or not considered related to the medicinal product. A Serious AE is defined as any untoward medical occurrence that: results in death, is immediately life-threatening, requires in-patient hospitalization or prolongation of existing hospitalization, results in persistent or significant disability/incapacity, and/or causes a congenital anomaly/birth defect. A TEAE is an AE that starts or worsens after receiving study drug. (NCT03277261)
Timeframe: From the first dose of study drug through the end of the study (up to approximately 116 weeks)

,
Interventionpercentage of participants (Number)
TEAEsTESAEs
Teriflunomide + IV Placebo89.16.9
Ublituximab + Oral Placebo86.111.4

[back to top]