Page last updated: 2024-09-24

fingolimod hydrochloride

Description

Fingolimod Hydrochloride: A sphingosine-derivative and IMMUNOSUPPRESSIVE AGENT that blocks the migration and homing of LYMPHOCYTES to the CENTRAL NERVOUS SYSTEM through its action on SPHINGOSINE 1-PHOSPHATE RECEPTORS. It is used in the treatment of MULTIPLE SCLEROSIS. [Medical Subject Headings (MeSH), National Library of Medicine, extracted Dec-2023]

fingolimod hydrochloride : The hydrochloride salt of 2-amino-2-[2-(4-octylphenyl) ethyl]-1,3-propanediol (fingolimod). [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]

Cross-References

ID SourceID
PubMed CID107969
CHEMBL ID544665
CHEBI ID63112
SCHEMBL ID81362
MeSH IDM0259274

Synonyms (94)

Synonym
AC-1929
fingolimod (hydrochloride)
HY-12005
fingolimod hydrochloride
fty-720
tdi-132
gilenia
gilenya
fty-720a
imusera
fingolimod hydrochloride (jan/usan)
gilenya (tn)
D04187
162359-56-0
1,3-propanediol, 2-amino-2-(2-(4-octylphenyl)ethyl)-, hydrochloride
2-amino-2-(2-(4-octylphenyl)ethyl)-1,3-propanediol hydrochloride
fty 720
fingolimod hydrochloride [usan]
fty720 ,
fingolimod hcl
chebi:63112 ,
CHEMBL544665
fty-720 hydrochloride
fingolimod hydrochlorid
fty720 hydrochloride
fingolimod (as hydrochloride)
AKOS005145784
EC-000.2314
2-amino-2-[2-(4-octylphenyl)ethyl]propane-1,3-diol hydrochloride
A25158
2-(4-octylphenethyl)-2-aminopropane-1,3-diol hydrochloride
FTY720 - FINGOLIMOD
2-amino-2-[2-(4-octyl-phenyl)-ethyl]-propane -1,3-diol hydrochloride
2-amino-2-(2-(4-octylphenyl)ethyl)propane-1,3-diol hydrochloride
2-amino-2-[2-(4-octylphenyl) ethyl]-1,3-propanediol hydrochloride
BCP9000705
FT-0643569
g926ec510t ,
unii-g926ec510t
BCPP000225
fty720,fingolimod
EPITOPE ID:156573
AM84549
CS-0114
S5002
fty720-d4 hydrochloride
fingolimod hydrochloride [who-dd]
fingolimod hydrochloride [ep monograph]
fingolimod hydrochloride [mi]
fingolimod hydrochloride [jan]
fingolimod hydrochloride [usp monograph]
fingolimod hydrochloride [orange book]
fingolimod hydrochloride [usp-rs]
SCHEMBL81362
smr004701287
MLS006010179
KS-1172
SWZTYAVBMYWFGS-UHFFFAOYSA-N
2-amino-2-[2-(4-octylphenyl)ethyl]-1,3-propanediol hydrochloride
2-amino-2-(4-octylphenethyl)propane-1,3-diol hydrochloride
mfcd00939512
2-amino-2-[2-(4-octylphenyl)-ethyl]-1,3-propanediol hydrochloride
fingolimod (fty720) hcl
2-amino-2-[2-(4-octyl-phenyl)-ethyl]-propane-1,3-diol hydrochloride
Q-101363
1,3-propanediol, 2-amino-2-[2-(4-octylphenyl)ethyl]-, hydrochloride
F1018
2-amino-2-[2-(4-n-octylphenyl)ethyl]propane-1,3-diol hydrochloride
2-amino-2-[2-(4-octylphenyl)ethyl]-1,3-propanediol, hydrochloride
DTXSID00167364 ,
2-amino-2-[2-(4-octylphenyl)ethyl]propane-1,3-diol;hydrochloride
EX-A960
fingolimod hclfty-720
fingolimod, hcl
SW219384-1
fty720 free base
Q27132395
SR-01000942237-2
sr-01000942237
BCP01808
[1-hydroxy-2-(hydroxymethyl)-4-(4-octylphenyl)butan-2-yl]azanium;chloride
CCG-265016
1,3-propanediol, 2-amino-2-[2-(4-octylphenyl)ethyl]-, hydrochloride (1:1)
SY057854
fingolimodhydrochloride
BF164458
fingolimod hydrochloride- bio-x
fingolimod hydrochloride (ep monograph)
dtxcid5089855
2-amino-2-(2-(4-octylphenyl)ethyl)propan-1,3-diol hydrochloride
fingolimod hydrochloride (usp-rs)
fingolimod hydrochloride (usp monograph)
2-amino-2-(2-(4-octylphenyl) ethyl)-1,3-propanediol hydrochloride
2-amino-2-(2-(4-octylphenyl)ethyl)propane-1,3-diol monohydrochloride

Research Excerpts

Overview

ExcerptReference
"Fingolimod hydrochloride is an effective immunomodulatory drug in improving relapsing-remitting multiple sclerosis (RRMS). "( Achiron, A; Gurevich, M; Stone, E; Waknin, R, 2018)

Roles (3)

RoleDescription
sphingosine-1-phosphate receptor agonistAn agonist that binds to and activates sphingosine 1-phosphate receptors.
immunosuppressive agentAn agent that suppresses immune function by one of several mechanisms of action. Classical cytotoxic immunosuppressants act by inhibiting DNA synthesis. Others may act through activation of T-cells or by inhibiting the activation of helper cells. In addition, an immunosuppressive agent is a role played by a compound which is exhibited by a capability to diminish the extent and/or voracity of an immune response.
prodrugA compound that, on administration, must undergo chemical conversion by metabolic processes before becoming the pharmacologically active drug for which it is a prodrug.
[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 (1)

ClassDescription
hydrochlorideA salt formally resulting from the reaction of hydrochloric acid with an organic base.
[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]

Protein Targets (5)

Activation Measurements

ProteinTaxonomyMeasurementAverageMin (ref.)Avg (ref.)Max (ref.)Bioassay(s)
Sphingosine 1-phosphate receptor 1Homo sapiens (human)EC50 (µMol)3.00000.00000.17597.8700AID639285
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Other Measurements

ProteinTaxonomyMeasurementAverageMin (ref.)Avg (ref.)Max (ref.)Bioassay(s)
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Biological Processes (85)

Processvia Protein(s)Taxonomy
blood vessel maturationSphingosine 1-phosphate receptor 1Homo sapiens (human)
cardiac muscle tissue growth involved in heart morphogenesisSphingosine 1-phosphate receptor 1Homo sapiens (human)
sphingosine-1-phosphate receptor signaling pathwaySphingosine 1-phosphate receptor 1Homo sapiens (human)
chemotaxisSphingosine 1-phosphate receptor 1Homo sapiens (human)
cell adhesionSphingosine 1-phosphate receptor 1Homo sapiens (human)
G protein-coupled receptor signaling pathwaySphingosine 1-phosphate receptor 1Homo sapiens (human)
adenylate cyclase-inhibiting G protein-coupled receptor signaling pathwaySphingosine 1-phosphate receptor 1Homo sapiens (human)
phospholipase C-activating G protein-coupled receptor signaling pathwaySphingosine 1-phosphate receptor 1Homo sapiens (human)
brain developmentSphingosine 1-phosphate receptor 1Homo sapiens (human)
cell population proliferationSphingosine 1-phosphate receptor 1Homo sapiens (human)
cell migrationSphingosine 1-phosphate receptor 1Homo sapiens (human)
transmission of nerve impulseSphingosine 1-phosphate receptor 1Homo sapiens (human)
lamellipodium assemblySphingosine 1-phosphate receptor 1Homo sapiens (human)
actin cytoskeleton organizationSphingosine 1-phosphate receptor 1Homo sapiens (human)
regulation of cell adhesionSphingosine 1-phosphate receptor 1Homo sapiens (human)
neuron differentiationSphingosine 1-phosphate receptor 1Homo sapiens (human)
positive regulation of cell migrationSphingosine 1-phosphate receptor 1Homo sapiens (human)
regulation of bone mineralizationSphingosine 1-phosphate receptor 1Homo sapiens (human)
leukocyte chemotaxisSphingosine 1-phosphate receptor 1Homo sapiens (human)
regulation of bone resorptionSphingosine 1-phosphate receptor 1Homo sapiens (human)
endothelial cell differentiationSphingosine 1-phosphate receptor 1Homo sapiens (human)
positive regulation of transcription by RNA polymerase IISphingosine 1-phosphate receptor 1Homo sapiens (human)
positive regulation of smooth muscle cell proliferationSphingosine 1-phosphate receptor 1Homo sapiens (human)
positive regulation of positive chemotaxisSphingosine 1-phosphate receptor 1Homo sapiens (human)
negative regulation of stress fiber assemblySphingosine 1-phosphate receptor 1Homo sapiens (human)
heart trabecula morphogenesisSphingosine 1-phosphate receptor 1Homo sapiens (human)
T cell migrationSphingosine 1-phosphate receptor 1Homo sapiens (human)
angiogenesisSphingosine 1-phosphate receptor 1Homo sapiens (human)
regulation of metabolic processSphingosine 1-phosphate receptor 1Homo sapiens (human)
adenylate cyclase-activating G protein-coupled receptor signaling pathwaySphingosine 1-phosphate receptor 1Homo sapiens (human)
blood vessel developmentSphingosine kinase 2Homo sapiens (human)
positive regulation of cytokine production involved in immune responseSphingosine kinase 2Homo sapiens (human)
sphingosine-1-phosphate receptor signaling pathwaySphingosine kinase 2Homo sapiens (human)
sphinganine-1-phosphate biosynthetic processSphingosine kinase 2Homo sapiens (human)
sphingosine metabolic processSphingosine kinase 2Homo sapiens (human)
brain developmentSphingosine kinase 2Homo sapiens (human)
cell population proliferationSphingosine kinase 2Homo sapiens (human)
positive regulation of cell population proliferationSphingosine kinase 2Homo sapiens (human)
sphingolipid biosynthetic processSphingosine kinase 2Homo sapiens (human)
negative regulation of cell growthSphingosine kinase 2Homo sapiens (human)
positive regulation of interleukin-13 productionSphingosine kinase 2Homo sapiens (human)
positive regulation of interleukin-6 productionSphingosine kinase 2Homo sapiens (human)
positive regulation of tumor necrosis factor productionSphingosine kinase 2Homo sapiens (human)
positive regulation of mast cell activation involved in immune responseSphingosine kinase 2Homo sapiens (human)
positive regulation of apoptotic processSphingosine kinase 2Homo sapiens (human)
regulation of canonical NF-kappaB signal transductionSphingosine kinase 2Homo sapiens (human)
positive regulation of mast cell degranulationSphingosine kinase 2Homo sapiens (human)
transcription initiation-coupled chromatin remodelingSphingosine kinase 2Homo sapiens (human)
sphingosine biosynthetic processSphingosine kinase 2Homo sapiens (human)
positive regulation of protein kinase C signalingSphingosine kinase 2Homo sapiens (human)
positive regulation of calcium ion importSphingosine kinase 2Homo sapiens (human)
regulation of reactive oxygen species biosynthetic processSphingosine kinase 2Homo sapiens (human)
cellular response to phorbol 13-acetate 12-myristateSphingosine kinase 2Homo sapiens (human)
regulation of cytochrome-c oxidase activitySphingosine kinase 2Homo sapiens (human)
positive regulation of ceramide biosynthetic processSphingosine kinase 2Homo sapiens (human)
regulation of ATP biosynthetic processSphingosine kinase 2Homo sapiens (human)
phosphorylationSphingosine kinase 2Homo sapiens (human)
negative regulation of apoptotic processSphingosine kinase 1Homo sapiens (human)
positive regulation of fibroblast proliferationSphingosine kinase 1Homo sapiens (human)
blood vessel developmentSphingosine kinase 1Homo sapiens (human)
sphingosine-1-phosphate receptor signaling pathwaySphingosine kinase 1Homo sapiens (human)
protein acetylationSphingosine kinase 1Homo sapiens (human)
sphingosine metabolic processSphingosine kinase 1Homo sapiens (human)
inflammatory responseSphingosine kinase 1Homo sapiens (human)
brain developmentSphingosine kinase 1Homo sapiens (human)
cell population proliferationSphingosine kinase 1Homo sapiens (human)
positive regulation of peptidyl-threonine phosphorylationSphingosine kinase 1Homo sapiens (human)
regulation of tumor necrosis factor-mediated signaling pathwaySphingosine kinase 1Homo sapiens (human)
phosphorylationSphingosine kinase 1Homo sapiens (human)
calcium-mediated signalingSphingosine kinase 1Homo sapiens (human)
regulation of endocytosisSphingosine kinase 1Homo sapiens (human)
sphingolipid biosynthetic processSphingosine kinase 1Homo sapiens (human)
positive regulation of cell growthSphingosine kinase 1Homo sapiens (human)
positive regulation of cell migrationSphingosine kinase 1Homo sapiens (human)
positive regulation of protein ubiquitinationSphingosine kinase 1Homo sapiens (human)
regulation of interleukin-1 beta productionSphingosine kinase 1Homo sapiens (human)
positive regulation of interleukin-17 productionSphingosine kinase 1Homo sapiens (human)
response to tumor necrosis factorSphingosine kinase 1Homo sapiens (human)
intracellular signal transductionSphingosine kinase 1Homo sapiens (human)
cellular response to vascular endothelial growth factor stimulusSphingosine kinase 1Homo sapiens (human)
negative regulation of apoptotic processSphingosine kinase 1Homo sapiens (human)
positive regulation of angiogenesisSphingosine kinase 1Homo sapiens (human)
positive regulation of mitotic nuclear divisionSphingosine kinase 1Homo sapiens (human)
positive regulation of mitotic cell cycleSphingosine kinase 1Homo sapiens (human)
positive regulation of smooth muscle contractionSphingosine kinase 1Homo sapiens (human)
sphingosine biosynthetic processSphingosine kinase 1Homo sapiens (human)
sphingoid catabolic processSphingosine kinase 1Homo sapiens (human)
regulation of phagocytosisSphingosine kinase 1Homo sapiens (human)
positive regulation of NF-kappaB transcription factor activitySphingosine kinase 1Homo sapiens (human)
cellular response to hydrogen peroxideSphingosine kinase 1Homo sapiens (human)
DNA biosynthetic processSphingosine kinase 1Homo sapiens (human)
regulation of neuroinflammatory responseSphingosine kinase 1Homo sapiens (human)
negative regulation of ceramide biosynthetic processSphingosine kinase 1Homo sapiens (human)
positive regulation of p38MAPK cascadeSphingosine kinase 1Homo sapiens (human)
positive regulation of non-canonical NF-kappaB signal transductionSphingosine kinase 1Homo sapiens (human)
regulation of microglial cell activationSphingosine kinase 1Homo sapiens (human)
regulation of endosomal vesicle fusionSphingosine kinase 1Homo sapiens (human)
cellular response to growth factor stimulusSphingosine kinase 1Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Molecular Functions (17)

Processvia Protein(s)Taxonomy
G protein-coupled receptor bindingSphingosine 1-phosphate receptor 1Homo sapiens (human)
G protein-coupled receptor activitySphingosine 1-phosphate receptor 1Homo sapiens (human)
protein bindingSphingosine 1-phosphate receptor 1Homo sapiens (human)
sphingosine-1-phosphate receptor activitySphingosine 1-phosphate receptor 1Homo sapiens (human)
sphingolipid bindingSphingosine 1-phosphate receptor 1Homo sapiens (human)
lipid kinase activitySphingosine kinase 2Homo sapiens (human)
protein bindingSphingosine kinase 2Homo sapiens (human)
ATP bindingSphingosine kinase 2Homo sapiens (human)
sphinganine kinase activitySphingosine kinase 2Homo sapiens (human)
D-erythro-sphingosine kinase activitySphingosine kinase 2Homo sapiens (human)
small GTPase bindingSphingosine kinase 2Homo sapiens (human)
sphingosine-1-phosphate receptor activitySphingosine kinase 2Homo sapiens (human)
histone bindingSphingosine kinase 2Homo sapiens (human)
magnesium ion bindingSphingosine kinase 1Homo sapiens (human)
lipid kinase activitySphingosine kinase 1Homo sapiens (human)
DNA bindingSphingosine kinase 1Homo sapiens (human)
protein bindingSphingosine kinase 1Homo sapiens (human)
calmodulin bindingSphingosine kinase 1Homo sapiens (human)
ATP bindingSphingosine kinase 1Homo sapiens (human)
lipid bindingSphingosine kinase 1Homo sapiens (human)
sphinganine kinase activitySphingosine kinase 1Homo sapiens (human)
acetyltransferase activitySphingosine kinase 1Homo sapiens (human)
D-erythro-sphingosine kinase activitySphingosine kinase 1Homo sapiens (human)
sphingosine-1-phosphate receptor activitySphingosine kinase 1Homo sapiens (human)
protein phosphatase 2A bindingSphingosine kinase 1Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Ceullar Components (19)

Processvia Protein(s)Taxonomy
nucleoplasmSphingosine 1-phosphate receptor 1Homo sapiens (human)
endosomeSphingosine 1-phosphate receptor 1Homo sapiens (human)
plasma membraneSphingosine 1-phosphate receptor 1Homo sapiens (human)
external side of plasma membraneSphingosine 1-phosphate receptor 1Homo sapiens (human)
intracellular membrane-bounded organelleSphingosine 1-phosphate receptor 1Homo sapiens (human)
membrane raftSphingosine 1-phosphate receptor 1Homo sapiens (human)
plasma membraneSphingosine 1-phosphate receptor 1Homo sapiens (human)
cytoplasmSphingosine 1-phosphate receptor 1Homo sapiens (human)
nucleusSphingosine kinase 2Homo sapiens (human)
nucleoplasmSphingosine kinase 2Homo sapiens (human)
cytoplasmSphingosine kinase 2Homo sapiens (human)
mitochondrionSphingosine kinase 2Homo sapiens (human)
mitochondrial inner membraneSphingosine kinase 2Homo sapiens (human)
lysosomal membraneSphingosine kinase 2Homo sapiens (human)
endoplasmic reticulumSphingosine kinase 2Homo sapiens (human)
cytosolSphingosine kinase 2Homo sapiens (human)
membraneSphingosine kinase 2Homo sapiens (human)
nucleosomeSphingosine kinase 2Homo sapiens (human)
intracellular membrane-bounded organelleSphingosine kinase 2Homo sapiens (human)
membraneSphingosine kinase 2Homo sapiens (human)
cytoplasmSphingosine kinase 2Homo sapiens (human)
presynapseSphingosine kinase 1Homo sapiens (human)
nucleusSphingosine kinase 1Homo sapiens (human)
nucleoplasmSphingosine kinase 1Homo sapiens (human)
cytoplasmSphingosine kinase 1Homo sapiens (human)
cytosolSphingosine kinase 1Homo sapiens (human)
plasma membraneSphingosine kinase 1Homo sapiens (human)
clathrin-coated pitSphingosine kinase 1Homo sapiens (human)
endocytic vesicleSphingosine kinase 1Homo sapiens (human)
early endosome membraneSphingosine kinase 1Homo sapiens (human)
presynapseSphingosine kinase 1Homo sapiens (human)
intracellular membrane-bounded organelleSphingosine kinase 1Homo sapiens (human)
membraneSphingosine kinase 1Homo sapiens (human)
cytoplasmSphingosine kinase 1Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Bioassays (99)

Assay IDTitleYearJournalArticle
AID1660287Anti-colitis activity against DSS-induced C57BL/6 mouse model of chronic relapsing colitis assessed as reduction in DSS-induced spleen swelling at 0.3 mg/kg, po administered once daily for 30 days starting from day 13 post DSS-challenge2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660273Anti-colitis activity against DSS-induced C57BL/6 mouse model of chronic relapsing colitis assessed as reduction in stool bleeding at 0.3 mg/kg, po administered once daily for 30 days starting from day 13 post DSS-challenge measured on day 432020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID196253Immunosuppressive activity against skin allograft in F344 recipient (rat) from LEW donor (rat) at a concentration of 0.1 mg/kg2000Journal of medicinal chemistry, Jul-27, Volume: 43, Issue:15
Synthesis and immunosuppressive activity of 2-substituted 2-aminopropane-1,3-diols and 2-aminoethanols.
AID1660247Anti-colitis activity against DSS-induced C57BL/6 mouse model of subchronic relapsing colitis assessed as reduction in stool consistency score at 0.15 mg/kg, po administered once daily for 14 days starting from day 5 post DSS-challenge cotreated with 20 m2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660298Anti-colitis activity against DSS-induced C57BL/6 mouse model of chronic relapsing colitis assessed as reduction in DSS-induced mononuclear cell infiltration in spleen at 0.3 mg/kg, po administered once daily for 30 days starting from day 13 post DSS-chal2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID183610Lymphocyte-decreasing effect against F344 rats.2000Journal of medicinal chemistry, Jul-27, Volume: 43, Issue:15
Synthesis and immunosuppressive activity of 2-substituted 2-aminopropane-1,3-diols and 2-aminoethanols.
AID1660326Antiinflammatory activity against DSS-induced C57BL/6 mouse model of subchronic relapsing colitis assessed as reduction in S1PR1 mRNA level at 0.15 mg/kg, po administered once daily for 14 days starting from day 5 post DSS-challenge cotreated with 20 mg/k2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660337Antiinflammatory activity in human THP1 cells assessed as reduction in LPS-induced TNFalpha mRNA level at 0.5 uM cotreated with 0.5 uM berberine incubated for 1 hr followed by LPS challenge and measured after 4 to 6 hrs by q-PCR method2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660286Anti-colitis activity against DSS-induced C57BL/6 mouse model of subchronic relapsing colitis assessed as reduction in DSS-induced colon swelling at 0.15 mg/kg, po administered once daily for 30 days starting from day 13 post DSS-challenge cotreated with 2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660334Antiinflammatory activity in human THP1 cells assessed as reduction in LPS-induced IL6 mRNA level at 1 uM incubated for 1 hr followed by LPS challenge and measured after 4 to 6 hrs by q-PCR method2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660275Anti-colitis activity against DSS-induced C57BL/6 mouse model of chronic relapsing colitis assessed as reduction in stool consistency score at 0.15 mg/kg, po administered once daily for 30 days starting from day 13 post DSS-challenge cotreated with 20 mg/2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660299Anti-colitis activity against DSS-induced C57BL/6 mouse model of chronic relapsing colitis assessed as reduction in DSS-induced mononuclear cell infiltration in spleen at 0.15 mg/kg, po administered once daily for 30 days starting from day 13 post DSS-cha2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID662675Lymphopenic activity in C57BL/6 mouse assessed as reduction in blood lymphocytes after 4 to 8 weeks relative to control2012Bioorganic & medicinal chemistry letters, Jun-15, Volume: 22, Issue:12
Identification of benzoxazole analogs as novel, S1P(3) sparing S1P(1) agonists.
AID1660335Antiinflammatory activity in human THP1 cells assessed as reduction in LPS-induced TNFalpha mRNA level at 1 uM incubated for 1 hr followed by LPS challenge and measured after 4 to 6 hrs by q-PCR method2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1677875Prodrug conversion in rat blood assessed as SPHK1/SPHK2-mediated compound phosphorylation by measuring intrinsic clearance by LC-MS/MS analysis2020Bioorganic & medicinal chemistry, 11-01, Volume: 28, Issue:21
Design and synthesis of analogues of the sphingosine-1-phosphate receptor 1 agonist IMMH001 with improved phosphorylation rate in human blood.
AID1660243Anti-colitis activity against DSS-induced C57BL/6 mouse model of subchronic relapsing colitis assessed as effect on body weight at 0.3 mg/kg, po administered once daily for 14 days starting from day 5 post DSS-challenge and measured after day 12 to day 182020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID628836Half life in human2011Bioorganic & medicinal chemistry letters, Oct-01, Volume: 21, Issue:19
Discovery and characterization of potent and selective 4-oxo-4-(5-(5-phenyl-1,2,4-oxadiazol-3-yl)indolin-1-yl)butanoic acids as S1P₁ agonists.
AID183445T cell-decreasing effect in spleen cells of WKAH rat which were injected into foot-pad of LEW rat.2000Journal of medicinal chemistry, Jul-27, Volume: 43, Issue:15
Synthesis and immunosuppressive activity of 2-substituted 2-aminopropane-1,3-diols and 2-aminoethanols.
AID1660341Anti-colitis activity against DSS-induced C57BL/6 mouse model of subchronic relapsing colitis assessed as reduction in phosphorylated STAT3 level at 0.3 mg/kg, po administered once daily for 14 days starting from day 5 post DSS-challenge cotreated with 202020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660253Anti-colitis activity against DSS-induced C57BL/6 mouse model of subchronic relapsing colitis assessed as reduction in disease activity index at 0.15 mg/kg, po administered once daily for 14 days starting from day 5 post DSS-challenge cotreated with 20 mg2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660331Effect on total S1PR1 expression in DSS-induced C57BL/6 mouse model of subchronic relapsing colitis at 0.3 mg/kg, po administered once daily for 14 days starting from day 5 post DSS-challenge by Western blot analysis2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660282Anti-colitis activity against DSS-induced C57BL/6 mouse model of chronic relapsing colitis assessed as reversal of DSS-induced colon shortening at 0.3 mg/kg, po administered once daily for 30 days starting from day 13 post DSS-challenge2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID639286Agonist activity at EGFP-tagged S1P1 receptor expressed in human UOS2 cells assessed as receptor internalization in endosomes at 3 uM after 60 mins relative to S1P2012Bioorganic & medicinal chemistry letters, Jan-01, Volume: 22, Issue:1
Novel immunomodulators based on an oxazolin-2-one-4-carboxamide scaffold.
AID1660255Anti-colitis activity against DSS-induced C57BL/6 mouse model of subchronic relapsing colitis assessed as reduction in DSS-induced colon shortening at 0.3 mg/kg, po administered once daily for 14 days starting from day 5 post DSS-challenge2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660249Anti-colitis activity against DSS-induced C57BL/6 mouse model of subchronic relapsing colitis assessed as reduction in stool bleeding scores at 0.3 mg/kg, po administered once daily for 14 days starting from day 5 post DSS challenge and measured on day 192020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660288Anti-colitis activity against DSS-induced C57BL/6 mouse model of chronic relapsing colitis assessed as reduction in DSS-induced spleen swelling at 0.15 mg/kg, po administered once daily for 30 days starting from day 13 post DSS-challenge cotreated with 202020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660259Anti-colitis activity against DSS-induced C57BL/6 mouse model of subchronic relapsing colitis assessed as reduction in colon swelling at 0.15 mg/kg, po administered once daily for 14 days starting from day 5 post DSS-challenge cotreated with 20 mg/kg berb2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660256Anti-colitis activity against DSS-induced C57BL/6 mouse model of subchronic relapsing colitis assessed as reduction in DSS-induced colon shortening at 0.15 mg/kg, po administered once daily for 14 days starting from day 5 post DSS-challenge cotreated with2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660330Effect on total STAT3 expression in DSS-induced C57BL/6 mouse model of subchronic relapsing colitis at 0.3 mg/kg, po administered once daily for 14 days starting from day 5 post DSS-challenge by Western blot analysis2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1677873Prodrug conversion in rat blood assessed as SPHK1/SPHK2-mediated compound phosphorylation by measuring Vmax by LC-MS/MS analysis2020Bioorganic & medicinal chemistry, 11-01, Volume: 28, Issue:21
Design and synthesis of analogues of the sphingosine-1-phosphate receptor 1 agonist IMMH001 with improved phosphorylation rate in human blood.
AID1660297Anti-colitis activity against DSS-induced C57BL/6 mouse model of chronic relapsing colitis assessed as reduction in DSS-induced inflammatory cell infiltration in colon at 0.15 mg/kg, po administered once daily for 30 days starting from day 13 post DSS-cha2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1677872Prodrug conversion in human blood assessed as SPHK1/SPHK2-mediated compound phosphorylation by measuring Km by LC-MS/MS analysis2020Bioorganic & medicinal chemistry, 11-01, Volume: 28, Issue:21
Design and synthesis of analogues of the sphingosine-1-phosphate receptor 1 agonist IMMH001 with improved phosphorylation rate in human blood.
AID196255Immunosuppressive activity against skin allograft in F344 recipient (rat) from LEW donor (rat) at a concentration of 1 mg/kg2000Journal of medicinal chemistry, Jul-27, Volume: 43, Issue:15
Synthesis and immunosuppressive activity of 2-substituted 2-aminopropane-1,3-diols and 2-aminoethanols.
AID1660310Toxicity in DSS-induced C57BL/6 mouse model of chronic relapsing colitis assessed as induction of abnormalities in heart at 0.15 mg/kg, po administered once daily for 30 days starting from day 13 post DSS-challenge cotreated with 20 mg/kg berberine by hem2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID182934In vivo immunosuppressive effect of compound was determined on host versus graft reaction (HvGR) using popliteal lymph node gain assay in rat1998Bioorganic & medicinal chemistry letters, Jan-06, Volume: 8, Issue:1
Synthesis and biological evaluation of 2,2-disubstituted 2-aminoethanols: analogues of FTY720.
AID1660285Anti-colitis activity against DSS-induced C57BL/6 mouse model of subchronic relapsing colitis assessed as reduction in DSS-induced colon swelling at 0.3 mg/kg, po administered once daily for 30 days starting from day 13 post DSS-challenge2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID637900Immunosuppressant activity in Balb/C mouse assessed as decrease in circulating CD3+ T cells at 10 mg/kg, po measured after 30 mins by flow cytometric analysis2012Bioorganic & medicinal chemistry letters, Jan-01, Volume: 22, Issue:1
Novel immunomodulators based on an oxazolin-2-one-4-carboxamide scaffold.
AID1660305Toxicity in DSS-induced C57BL/6 mouse model of chronic relapsing colitis assessed as induction of abnormalities in heart at 0.3 mg/kg, po administered once daily for 30 days starting from day 13 post DSS-challenge by hematoxylin and eosin staining based l2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID639287Agonist activity at EGFP-tagged S1P1 receptor expressed in human UOS2 cells assessed as receptor internalization in endosomes at 10 uM after 60 mins relative to S1P2012Bioorganic & medicinal chemistry letters, Jan-01, Volume: 22, Issue:1
Novel immunomodulators based on an oxazolin-2-one-4-carboxamide scaffold.
AID182789Decrease in number of T cells in peripheral blood after administration for 4 days in rat1998Bioorganic & medicinal chemistry letters, Jan-06, Volume: 8, Issue:1
Synthesis and biological evaluation of 2,2-disubstituted 2-aminoethanols: analogues of FTY720.
AID196257Immunosuppressive activity against skin allograft in F344 recipient (rat) from LEW donor (rat) at a concentration of 10 mg/kg; Toxic2000Journal of medicinal chemistry, Jul-27, Volume: 43, Issue:15
Synthesis and immunosuppressive activity of 2-substituted 2-aminopropane-1,3-diols and 2-aminoethanols.
AID1660283Anti-colitis activity against DSS-induced C57BL/6 mouse model of subchronic relapsing colitis assessed as reversal of DSS-induced colon shortening at 0.15 mg/kg, po administered once daily for 30 days starting from day 13 post DSS-challenge cotreated with2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660278Anti-colitis activity against DSS-induced C57BL/6 mouse model of chronic relapsing colitis assessed as reduction in stool consistency score at 0.15 mg/kg, po administered once daily for 30 days starting from day 13 post DSS-challenge cotreated with 20 mg/2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660269Anti-colitis activity against DSS-induced C57BL/6 mouse model of chronic relapsing colitis assessed as reduction in stool consistency score at 0.3 mg/kg, po administered once daily for 30 days starting from day 13 post DSS-challenge measured on day 242020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID639291Immunosuppressant activity in Balb/C mouse assessed as decrease in circulating CD3+ T cells at 0.1 mg/kg, po measured after 30 mins by flow cytometric analysis2012Bioorganic & medicinal chemistry letters, Jan-01, Volume: 22, Issue:1
Novel immunomodulators based on an oxazolin-2-one-4-carboxamide scaffold.
AID639292Immunosuppressant activity in Balb/C mouse assessed as decrease in circulating CD3+ T cells at 0.3 mg/kg, po measured after 30 mins by flow cytometric analysis2012Bioorganic & medicinal chemistry letters, Jan-01, Volume: 22, Issue:1
Novel immunomodulators based on an oxazolin-2-one-4-carboxamide scaffold.
AID1660314Toxicity in DSS-induced C57BL/6 mouse model of chronic relapsing colitis assessed as induction of abnormalities in lung at 0.15 mg/kg, po administered once daily for 30 days starting from day 13 post DSS-challenge cotreated with 20 mg/kg berberine by hema2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660313Toxicity in DSS-induced C57BL/6 mouse model of chronic relapsing colitis assessed as induction of abnormalities in brain at 0.15 mg/kg, po administered once daily for 30 days starting from day 13 post DSS-challenge cotreated with 20 mg/kg berberine by hem2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660294Anti-colitis activity against DSS-induced C57BL/6 mouse model of chronic relapsing colitis assessed as reduction in DSS-induced inflammatory cell infiltration in colon at 0.3 mg/kg, po administered once daily for 30 days starting from day 13 post DSS-chal2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660339Inhibition of STAT3 activation in human THP1 cells assessed as reduction in LPS-induced phosphorylated STAT3 expression at 0.5 uM cotreated with 0.5 uM berberine incubated for 1 hr followed by LPS challenge and measured after 4 to 6 hrs by Western blot an2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID639288Cytotoxicity against HLF cells2012Bioorganic & medicinal chemistry letters, Jan-01, Volume: 22, Issue:1
Novel immunomodulators based on an oxazolin-2-one-4-carboxamide scaffold.
AID1660340Anti-colitis activity against DSS-induced C57BL/6 mouse model of subchronic relapsing colitis assessed as reduction in phosphorylated STAT3 level at 0.3 mg/kg, po administered once daily for 14 days starting from day 5 post DSS-challenge by Western blot a2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660270Anti-colitis activity against DSS-induced C57BL/6 mouse model of chronic relapsing colitis assessed as reduction in stool bleeding at 0.3 mg/kg, po administered once daily for 30 days starting from day 13 post DSS-challenge measured on day 382020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660320Antiinflammatory activity against DSS-induced C57BL/6 mouse model of subchronic relapsing colitis assessed as reduction in TNFalpha mRNA level at 0.3 mg/kg, po administered once daily for 14 days starting from day 5 post DSS-challenge by RT-qPCR analysis2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660321Antiinflammatory activity against DSS-induced C57BL/6 mouse model of subchronic relapsing colitis assessed as reduction in IL6 mRNA level at 0.3 mg/kg, po administered once daily for 14 days starting from day 5 post DSS-challenge by RT-qPCR analysis2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID639285Agonist activity at EGFP-tagged S1P1 receptor expressed in human UOS2 cells assessed as receptor internalization in endosomes after 60 mins2012Bioorganic & medicinal chemistry letters, Jan-01, Volume: 22, Issue:1
Novel immunomodulators based on an oxazolin-2-one-4-carboxamide scaffold.
AID183137Lymphocyte-decreasing effect against F344 rats.2000Journal of medicinal chemistry, Jul-27, Volume: 43, Issue:15
Synthesis and immunosuppressive activity of 2-substituted 2-aminopropane-1,3-diols and 2-aminoethanols.
AID1660332Effect on total STAT3 expression in DSS-induced C57BL/6 mouse model of subchronic relapsing colitis at 0.15 mg/kg, po administered once daily for 14 days starting from day 5 post DSS-challenge cotreated with 20 mg/kg berberine by Western blot analysis2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1677870Prodrug conversion in human blood assessed as SPHK1/SPHK2-mediated compound phosphorylation by measuring intrinsic clearance by LC-MS/MS analysis2020Bioorganic & medicinal chemistry, 11-01, Volume: 28, Issue:21
Design and synthesis of analogues of the sphingosine-1-phosphate receptor 1 agonist IMMH001 with improved phosphorylation rate in human blood.
AID1660336Antiinflammatory activity in human THP1 cells assessed as reduction in LPS-induced IL6 mRNA level at 0.5 uM cotreated with 0.5 uM berberine incubated for 1 hr followed by LPS challenge and measured after 4 to 6 hrs by q-PCR method2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660280Anti-colitis activity against DSS-induced C57BL/6 mouse model of chronic relapsing colitis assessed as reduction in disease activity index at 0.15 mg/kg, po administered once daily for 30 days starting from day 13 post DSS-challenge cotreated with 20 mg/k2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660250Anti-colitis activity against DSS-induced C57BL/6 mouse model of subchronic relapsing colitis assessed as reduction in stool bleeding score at 0.15 mg/kg, po administered once daily for 14 days starting from day 5 post DSS-challenge cotreated with 20 mg/k2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660319Antiinflammatory activity against DSS-induced C57BL/6 mouse model of subchronic relapsing colitis assessed as reduction in IL17 mRNA level at 0.3 mg/kg, po administered once daily for 14 days starting from day 5 post DSS-challenge by RT-qPCR analysis2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660323Antiinflammatory activity against DSS-induced C57BL/6 mouse model of subchronic relapsing colitis assessed as reduction in IL17 mRNA level at 0.15 mg/kg, po administered once daily for 14 days starting from day 5 post DSS-challenge cotreated with 20 mg/kg2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660261Anti-colitis activity against DSS-induced C57BL/6 mouse model of subchronic relapsing colitis assessed as reduction in spleen swelling at 0.3 mg/kg, po administered once daily for 14 days starting from day 5 post DSS-challenge2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID196259Immunosuppressive activity against skin allograft in F344 recipient (rat) from LEW donor (rat) at a concentration of 3 mg/kg2000Journal of medicinal chemistry, Jul-27, Volume: 43, Issue:15
Synthesis and immunosuppressive activity of 2-substituted 2-aminopropane-1,3-diols and 2-aminoethanols.
AID1660252Anti-colitis activity against DSS-induced C57BL/6 mouse model of subchronic relapsing colitis assessed as reduction in disease activity index at 0.3 mg/kg, po administered once daily for 14 days starting from day 5 post DSS challenge and measured on day 12020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID196254Immunosuppressive activity against skin allograft in F344 recipient (rat) from LEW donor (rat) at a concentration of 0.3 mg/kg2000Journal of medicinal chemistry, Jul-27, Volume: 43, Issue:15
Synthesis and immunosuppressive activity of 2-substituted 2-aminopropane-1,3-diols and 2-aminoethanols.
AID1660311Toxicity in DSS-induced C57BL/6 mouse model of chronic relapsing colitis assessed as induction of abnormalities in liver at 0.15 mg/kg, po administered once daily for 30 days starting from day 13 post DSS-challenge cotreated with 20 mg/kg berberine by hem2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660272Anti-colitis activity against DSS-induced C57BL/6 mouse model of chronic relapsing colitis assessed as reduction in stool consistency score at 0.3 mg/kg, po administered once daily for 30 days starting from day 13 post DSS-challenge measured on day 432020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID183454Immunosuppressive activity against LEW rats transfected with spleen cells of WKAH rat in Popliteal Lymph Node Gain Assay.2000Journal of medicinal chemistry, Jul-27, Volume: 43, Issue:15
Synthesis and immunosuppressive activity of 2-substituted 2-aminopropane-1,3-diols and 2-aminoethanols.
AID662674Lymphopenic activity in C57BL/6 mouse assessed as reduction in blood lymphocytes relative to control2012Bioorganic & medicinal chemistry letters, Jun-15, Volume: 22, Issue:12
Identification of benzoxazole analogs as novel, S1P(3) sparing S1P(1) agonists.
AID1660295Anti-colitis activity against DSS-induced C57BL/6 mouse model of chronic relapsing colitis assessed as reduction in DSS-induced multiple erosive lesions in colon at 0.15 mg/kg, po administered once daily for 30 days starting from day 13 post DSS-challenge2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660244Anti-colitis activity against DSS-induced C57BL/6 mouse model of subchronic relapsing colitis assessed as effect on body weight at 0.15 mg/kg, po administered once daily for 14 days starting from day 5 post DSS-challenge cotreated with 20 mg/kg berberine 2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660277Anti-colitis activity against DSS-induced C57BL/6 mouse model of chronic relapsing colitis assessed as reduction in disease activity index at 0.15 mg/kg, po administered once daily for 30 days starting from day 13 post DSS-challenge cotreated with 20 mg/k2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660292Anti-colitis activity against DSS-induced C57BL/6 mouse model of chronic relapsing colitis assessed as reduction in DSS-induced multiple erosive lesions in colon at 0.3 mg/kg, po administered once daily for 30 days starting from day 13 post DSS-challenge2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660333Effect on total S1PR1 expression in DSS-induced C57BL/6 mouse model of subchronic relapsing colitis at 0.15 mg/kg, po administered once daily for 14 days starting from day 5 post DSS-challenge cotreated with 20 mg/kg berberine by Western blot analysis2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660325Antiinflammatory activity against DSS-induced C57BL/6 mouse model of subchronic relapsing colitis assessed as reduction in IL6 mRNA level at 0.15 mg/kg, po administered once daily for 14 days starting from day 5 post DSS-challenge cotreated with 20 mg/kg 2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660274Anti-colitis activity against DSS-induced C57BL/6 mouse model of chronic relapsing colitis assessed as reduction in disease activity index at 0.3 mg/kg, po administered once daily for 30 days starting from day 13 post DSS-challenge and measured on day 432020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660312Toxicity in DSS-induced C57BL/6 mouse model of chronic relapsing colitis assessed as induction of abnormalities in kidney at 0.15 mg/kg, po administered once daily for 30 days starting from day 13 post DSS-challenge cotreated with 20 mg/kg berberine by he2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660246Anti-colitis activity against DSS-induced C57BL/6 mouse model of subchronic relapsing colitis assessed as reduction in stool consistency scores at 0.3 mg/kg, po administered once daily for 14 days starting from day 5 post DSS challenge and measured on day2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1677874Prodrug conversion in rat blood assessed as SPHK1/SPHK2-mediated compound phosphorylation by measuring Km by LC-MS/MS analysis2020Bioorganic & medicinal chemistry, 11-01, Volume: 28, Issue:21
Design and synthesis of analogues of the sphingosine-1-phosphate receptor 1 agonist IMMH001 with improved phosphorylation rate in human blood.
AID1660271Anti-colitis activity against DSS-induced C57BL/6 mouse model of chronic relapsing colitis assessed as reduction in disease activity index at 0.3 mg/kg, po administered once daily for 30 days starting from day 13 post DSS-challenge and measured on day 242020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660322Antiinflammatory activity against DSS-induced C57BL/6 mouse model of subchronic relapsing colitis assessed as reduction in S1PR1 mRNA level at 0.3 mg/kg, po administered once daily for 14 days starting from day 5 post DSS-challenge by RT-qPCR analysis2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660293Anti-colitis activity against DSS-induced C57BL/6 mouse model of chronic relapsing colitis assessed as reduction in DSS-induced loss of colon crypt and columnar epithelium at 0.3 mg/kg, po administered once daily for 30 days starting from day 13 post DSS-2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660262Anti-colitis activity against DSS-induced C57BL/6 mouse model of subchronic relapsing colitis assessed as reduction in spleen swelling at 0.15 mg/kg, po administered once daily for 14 days starting from day 5 post DSS-challenge cotreated with 20 mg/kg ber2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660309Toxicity in DSS-induced C57BL/6 mouse model of chronic relapsing colitis assessed as induction of abnormalities in lung at 0.3 mg/kg, po administered once daily for 30 days starting from day 13 post DSS-challenge by hematoxylin and eosin staining based li2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660276Anti-colitis activity against DSS-induced C57BL/6 mouse model of chronic relapsing colitis assessed as reduction in stool bleeding at 0.15 mg/kg, po administered once daily for 30 days starting from day 13 post DSS-challenge cotreated with 20 mg/kg berber2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660308Toxicity in DSS-induced C57BL/6 mouse model of chronic relapsing colitis assessed as induction of abnormalities in brain at 0.3 mg/kg, po administered once daily for 30 days starting from day 13 post DSS-challenge by hematoxylin and eosin staining based l2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID637899Immunosuppressant activity in Balb/C mouse assessed as decrease in circulating CD3+ T cells at 3 mg/kg, po measured after 30 mins by flow cytometric analysis2012Bioorganic & medicinal chemistry letters, Jan-01, Volume: 22, Issue:1
Novel immunomodulators based on an oxazolin-2-one-4-carboxamide scaffold.
AID1660338Inhibition of STAT3 activation in human THP1 cells assessed as reduction in LPS-induced phosphorylated STAT3 expression at 1 uM incubated for 1 hr followed by LPS challenge and measured after 4 to 6 hrs by Western blot analysis2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID637898Immunosuppressant activity in Balb/C mouse assessed as decrease in circulating CD3+ T cells at 1 mg/kg, po measured after 30 mins by flow cytometric analysis2012Bioorganic & medicinal chemistry letters, Jan-01, Volume: 22, Issue:1
Novel immunomodulators based on an oxazolin-2-one-4-carboxamide scaffold.
AID1660279Anti-colitis activity against DSS-induced C57BL/6 mouse model of chronic relapsing colitis assessed as reduction in stool bleeding at 0.15 mg/kg, po administered once daily for 30 days starting from day 13 post DSS-challenge cotreated with 20 mg/kg berber2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660306Toxicity in DSS-induced C57BL/6 mouse model of chronic relapsing colitis assessed as induction of abnormalities in liver at 0.3 mg/kg, po administered once daily for 30 days starting from day 13 post DSS-challenge by hematoxylin and eosin staining based l2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660296Anti-colitis activity against DSS-induced C57BL/6 mouse model of chronic relapsing colitis assessed as reduction in DSS-induced loss of colon crypt and columnar epithelium at 0.15 mg/kg, po administered once daily for 30 days starting from day 13 post DSS2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660307Toxicity in DSS-induced C57BL/6 mouse model of chronic relapsing colitis assessed as induction of abnormalities in kidney at 0.3 mg/kg, po administered once daily for 30 days starting from day 13 post DSS-challenge by hematoxylin and eosin staining based 2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1677871Prodrug conversion in human blood assessed as SPHK1/SPHK2-mediated compound phosphorylation by measuring Vmax by LC-MS/MS analysis2020Bioorganic & medicinal chemistry, 11-01, Volume: 28, Issue:21
Design and synthesis of analogues of the sphingosine-1-phosphate receptor 1 agonist IMMH001 with improved phosphorylation rate in human blood.
AID1660324Antiinflammatory activity against DSS-induced C57BL/6 mouse model of subchronic relapsing colitis assessed as reduction in TNFalpha mRNA level at 0.15 mg/kg, po administered once daily for 14 days starting from day 5 post DSS-challenge cotreated with 20 m2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
AID1660258Anti-colitis activity against DSS-induced C57BL/6 mouse model of subchronic relapsing colitis assessed as reduction in colon swelling at 0.3 mg/kg, po administered once daily for 14 days starting from day 5 post DSS-challenge2020Journal of natural products, 06-26, Volume: 83, Issue:6
Anti-inflammatory Efficacy of Combined Natural Alkaloid Berberine and S1PR Modulator Fingolimod at Low Doses in Ulcerative Colitis Preclinical Models.
[information is prepared from bioassay data collected from National Library of Medicine (NLM), extracted Dec-2023]

Research

Studies (2,693)

TimeframeStudies, This Drug (%)All Drugs %
pre-19900 (0.00)18.7374
1990's56 (2.08)18.2507
2000's478 (17.75)29.6817
2010's1535 (57.00)24.3611
2020's624 (23.17)2.80
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Study Types

Publication TypeThis drug (%)All Drugs (%)
Trials157 (5.67%)5.53%
Reviews380 (13.71%)6.00%
Case Studies227 (8.19%)4.05%
Observational98 (3.54%)0.25%
Other1,909 (68.89%)84.16%
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Clinical Trials (96)

Trial Overview

TrialPhaseEnrollmentStudy TypeStart DateStatus
A 4-month, Open-label, Multi-center Study to Explore Tolerability and Safety and Health Outcomes of FTY720 in Patients With Relapsing Forms of Multiple Sclerosis[NCT01127750]Phase 32,417 participants (Actual)Interventional2010-05-31Completed
Long-term, Prospective,Multinational, Parallel-cohort Study Monitoring Safety in Patients With MS Newly Started With Fingolimod Once Daily or Treated With Another Approved Disease-modifying Therapy[NCT01442194]3,076 participants (Actual)Observational2011-08-01Completed
A 6-month, Randomized, Active Comparator, Open-label, Multi-Center Study to Evaluate Patient Outcomes, Safety and Tolerability of Fingolimod (FTY720) 0.5 mg/Day in Patients With Relapsing Remitting Multiple Sclerosis Who Are Candidates for MS Therapy Chan[NCT01317004]Phase 461 participants (Actual)Interventional2011-05-31Completed
A Randomized, Single Dose, Crossover, Bioequivalence Study of Fingolimod 0.5 mg Capsule (Asofarma S.A.I. y C., Argentina) Compared With Gilenya 0.5 mg Capsule (Novartis Pharmaceuticals, Australia Pty Ltd) in Fasting Healthy Subjects[NCT03757338]Phase 133 participants (Actual)Interventional2015-10-23Completed
A 6-month Multicenter, Single-arm, Open-label Study to Investigate Changes in Biomarkers After Initiation of Treatment With 0.5 mg Fingolimod (FTY720) in Patients With Relapsing-remitting Multiple Sclerosis[NCT01310166]Phase 4447 participants (Actual)Interventional2011-02-28Completed
A Long Term, Multicenter, Non-interventional, Observational Study Monitoring Long-term Safety and Effectiveness of Fingolimod 0.5 mg in Patients With Multiple Sclerosis Who Have Participated in the Fingolimod Clinical Development Program[NCT01281657]64 participants (Actual)Observational2011-02-28Completed
Czech Pharmaco-epidemiological Real World Data Study Focused on Effectiveness of Different Disease Modifying Drugs[NCT05762003]17,478 participants (Actual)Observational2019-01-01Completed
A 32-week, Monocentric, Exploratory, Single Arm Study to Assess Immune Function and MRI Disease Activity in Patients With RRMS Transferred From Previous Treatment With Natalizumab to Gilenya® (Fingolimod)[NCT02325440]Phase 415 participants (Anticipated)Interventional2014-03-31Recruiting
Prevention of Paclitaxel-Associated Neuropathy With Fingolimod: a Pilot Trial[NCT03941743]Phase 12 participants (Actual)Interventional2019-12-12Completed
A Randomised Controlled Trial of Combinating an Immune Modulator Fingolimod With Alteplase Bridging With Mechanical Thrombectomy in Acute Ischemic Stroke[NCT04675762]Phase 2118 participants (Anticipated)Interventional2021-01-15Recruiting
An Investigator-initiated, Multicenter, Phase IV, Open-label Study to Evaluate the Biological Basis for Disease Progression in Relapsing-remitting Multiple Sclerosis Patients Treated in Routine Practice With Gilenya for 2 Years[NCT02137707]135 participants (Actual)Observational2012-11-30Completed
A Phase 2, Randomized, Placebo-Controlled Study to Evaluate Fingolimod for the Abrogation of Interstitial Fibrosis and Tubular Atrophy Following Kidney Transplantation[NCT05285878]Phase 220 participants (Anticipated)Interventional2022-07-28Enrolling by invitation
Evaluating the Effect of Fingolimod With Fish Oil Compared to Fingolimod With Placebo on Tumor Necrosis Factor-α , Interleukin1b , Interleukin6, and Interferon-gamma in Patients With Relapsing-Remitting Multiple Sclerosis[NCT02939079]Phase 2/Phase 350 participants (Actual)Interventional2015-04-30Completed
A 12-Month, Prospective, Multicenter, Two-cohort, Nonrandomized, Open-label Study in Adult Patients With Relapsing Multiple Sclerosis (RMS), to Investigate Changes in Immune Phenotype Biomarkers After Treatment With 0.5mg Fingolimod [FLUENT][NCT03257358]Phase 4382 participants (Actual)Interventional2017-09-19Completed
A Multicentric Randomized PRAGmatic Trial to Compare the Effectiveness of Fingolimod Versus Dimethyl-Fumarate on Patient Overall Disease Experience in Relapsing Remitting Multiple Sclerosis: Novel Data to Inform Decision-makers[NCT03345940]Phase 455 participants (Actual)Interventional2017-04-30Terminated(stopped due to termination of the study due to the slowness of the recruitment activity, according to the contract signed with the Sponsor)
A 24-month, Open-label, Prospective, Multicenter Interventional, Single-arm Study Assessing the Efficacy and Safety of Fingolimod (Gilenya) 0.5 mg in Relapsing Multiple Sclerosis (RMS) Patients in China[NCT04667949]Phase 498 participants (Actual)Interventional2021-02-20Active, not recruiting
Efficacy of Fingolimod in the Treatment of New Coronavirus Pneumonia (COVID-19)[NCT04280588]Phase 20 participants (Actual)Interventional2020-02-22Withdrawn(stopped due to No participants enrolled)
A Double-blind, Randomized, Multicenter, Placebo-controlled, Parallel-group Study Comparing the Efficacy and Safety of 0.5mg Fingolimod Administered Orally Once Daily Versus Placebo in Patients With Primary Progressive Multiple Sclerosis and An Open-label[NCT00731692]Phase 3970 participants (Actual)Interventional2008-07-28Terminated(stopped due to The extension study was terminated early after the results of the core study showed the study did not meet primary endpoint; confirmed disability progression)
A Double-blind, Randomized, Placebo-controlled, Parallel, Time-lagged, Ascending, Multi-centre, Multiple-dose Study to Measure the Magnitude and Time Course of the Effect of FTY720 on FEV1 and Other Pulmonary Function Tests (FVC, FEF25-75%, and FEV1/FVC) [NCT00785083]Phase 236 participants (Actual)Interventional2008-09-30Completed
Assessing Induction of Type II (M2) Monocytes/Macrophages in Patients Receiving Gilenya.[NCT02225977]125 participants (Actual)Observational2013-07-31Completed
A 18-month, Open-label, Rater-blinded, Randomized, Multi-center, Active-controlled, Parallel-group Pilot Study to Assess Efficacy and Safety of Fingolimod in Comparison to Interferon Beta 1b in Treating the Cognitive Symptoms Associated to Relapsing-remit[NCT01333501]Phase 4151 participants (Actual)Interventional2011-05-31Completed
A Phase 1 Clinical Study to Assess Safety and Efficacy of Oral Fingolimod (FTY720) in Children With Rett Syndrome.[NCT02061137]Phase 1/Phase 26 participants (Actual)Interventional2013-08-31Completed
A Multicenter, Randomized, Open-Label Study to Assess the Impact of Natalizumab Versus Fingolimod on Central Nervous System Tissue Damage and Recovery in Active Relapsing-Remitting Multiple Sclerosis Subjects[NCT02342704]Phase 4111 participants (Actual)Interventional2014-11-30Terminated(stopped due to Business Decision)
[NCT02193217]Phase 181 participants (Actual)InterventionalCompleted
Efficacy and Safety of Fingolimod in Minimal Invasive Treatment of Intracerebral Hemorrhage[NCT06087965]Phase 1/Phase 240 participants (Anticipated)Interventional2023-10-11Not yet recruiting
Combinating Fingolimod With Endovascular Treatment in Acute Ischemic Stroke[NCT04629872]Phase 230 participants (Anticipated)Interventional2020-11-12Recruiting
Fingolimod for Type 2 Diabetes Mellitus: a Pilot, Prospective, Randomized, and Open Label Single-center Study[NCT05307731]Phase 440 participants (Anticipated)Interventional2022-03-15Recruiting
A Phase III Multicenter, Randomized, Double-Blind, Double-Dummy Study To Evaluate Safety And Efficacy Of Ocrelizumab In Comparison With Fingolimod In Children And Adolescents With Relapsing-Remitting Multiple Sclerosis[NCT05123703]Phase 3233 participants (Anticipated)Interventional2022-02-04Recruiting
Pregnancy Outcomes in Women Exposed to Diroximel Fumarate[NCT05688436]1,178 participants (Anticipated)Observational [Patient Registry]2021-09-24Recruiting
A 12-month Double-blind, Randomized, Multicenter, Active-controlled, Parallel-group Study Comparing the Efficacy and Safety of 0.5 mg and 1.25 mg Fingolimod (FTY720) Administered Orally Once Daily Versus Interferon ß-1a (Avonex) Administered im Once Weekl[NCT00340834]Phase 31,292 participants (Actual)Interventional2006-05-31Completed
A 24-month, Double-blind, Randomized, Multicenter, Placebo-controlled, Parallel-group Study Comparing the Efficacy and Safety of Fingolimod 1.25 mg and 0.5 mg Administered Orally Once Daily Versus Placebo in Patients With Relapsing-remitting Multiple Scle[NCT00289978]Phase 31,272 participants (Actual)Interventional2006-01-31Completed
An Open-label, Single-dose, Parallel-group Study to Compare the Pharmacokinetics of FTY720 and Metabolites in Subjects With Severe Renal Impairment With That in Matched Healthy Control Subjects[NCT00731523]Phase 118 participants (Actual)Interventional2008-07-31Completed
An Extension of the 6-month, Double-blind, Randomized, Placebo-controlled, Parallel-group, Multicenter Study Comparing Efficacy and Safety of FTY720 0.5 mg and 1.25 mg Administered Orally Once Daily in Patients With Relapsing Multiple Sclerosis[NCT00670449]Phase 2143 participants (Actual)Interventional2008-04-30Completed
An Extension of the 24-month, Double-blind, Randomized, Multicenter, Placebo-controlled, Parallel-group Study Comparing Efficacy and Safety of Fingolimod (FTY720) 1.25 mg and 0.5 mg Administered Orally Once Daily Versus Placebo in Patients With Relapsing-[NCT00662649]Phase 3920 participants (Actual)Interventional2008-02-29Completed
A 4-month, Prospective, Open-label, Multi-center Phase IV Study to Assess Response to Fingolimod Initiation According to Coping Profile in Adult Patients With Highly Active Relapsing Remitting Multiple Sclerosis in France[NCT01420055]Phase 4189 participants (Actual)Interventional2011-08-31Completed
A 6-month, Double-blind, Randomized, Placebo-controlled, Parallel-group, Multicenter Study Comparing Efficacy and Safety of FTY720 0.5 mg and 1.25 mg Administered Orally Once Daily in Patients With Relapsing Multiple Sclerosis[NCT00537082]Phase 2171 participants (Actual)Interventional2007-09-30Completed
24-month Double-blind, Randomized, Multicenter, Placebo-controlled, Parallel-group Study Comparing the Efficacy and Safety of 0.5 mg and 1.25 mg Fingolimod (FTY720) Administered Orally Once Daily Versus Placebo in Patients With Relapsing-remitting Multipl[NCT00355134]Phase 31,083 participants (Actual)Interventional2006-06-30Completed
Double-blind, Randomized, Placebo-controlled, Parallel-group, Multicenter Study Evaluating the Safety, Tolerability and Effect on MRI Lesion Parameters of FTY720 vs Placebo in Patients With Relapsing Multiple Sclerosis Including 18 Month Extension Phase[NCT00333138]Phase 2281 participants (Actual)Interventional2003-05-31Completed
A 12 Month Study, With a 6-month, Double-blind, Randomized, Placebo-controlled, Multi-center Parallel- Groups, Treatment Phase Evaluating Efficacy and Safety of Fingolimod 0.5 mg and a 6-month, Open-label, Treatment Phase, in Chinese Patients With Relapsi[NCT01941004]Phase 30 participants (Actual)Interventional2014-06-30Withdrawn
A Two-year Extension to a One-year, Multicenter, Partially Blinded, Double Dummy, Randomized Study to Evaluate the Efficacy and Safety of FTY720 Combined With Reduced-dose or Full-dose Cyclosporine, USP [Modified] (Novartis Brand) and Corticosteroids Vers[NCT00239811]Phase 3684 participants Interventional2004-04-30Completed
Efficacy and Safety of FTY720 for the Treatment of Acute Stroke[NCT02002390]Phase 222 participants (Actual)Interventional2012-10-31Completed
Treatment of Established Chemotherapy-Induced Neuropathy With Fingolimod: A Pilot Trial[NCT03943498]Early Phase 12 participants (Actual)Interventional2019-05-24Completed
A 3-month Blinded, Randomized, Multicenter, Placebo-controlled Study to Evaluate the Effect of Treatment With Fingolimod on the Immune Response Following Seasonal Influenza Vaccination and Tetanus Toxoid Booster Injection in Patients With Relapsing Forms [NCT01199861]Phase 3138 participants (Actual)Interventional2010-08-31Completed
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
An Open Label Randomized, Single Dose, Two-way Crossover Bioequivalence Study to Determine the Bioequivalence of Fingolimod From Sphingomod 0.5 mg Hard Gelatin Capsules (Hikma Pharma, Egypt) Versus Gilenya 0.5 mg Hard Capsules (Novartis Pharma AG, Basle, [NCT04657744]Phase 134 participants (Actual)Interventional2020-08-10Completed
A 6-month, Randomized, Active Comparator, Open-label, Multi-Center Study to Evaluate Patient OutComes, Safety and Tolerability of Fingolimod (FTY720) 0.5 mg/Day in Patients With Relapsing Forms of Multiple Sclerosis Who Are Candidates for MS Therapy Chang[NCT01216072]Phase 41,053 participants (Actual)Interventional2010-08-31Completed
A 2-year Randomized, 3-arm, Double-blind, Non-inferiority Study Comparing the Efficacy and Safety of Ofatumumab and Siponimod Versus Fingolimod in Pediatric Patients With Multiple Sclerosis Followed by an Open-label Extension[NCT04926818]Phase 3180 participants (Anticipated)Interventional2021-10-05Recruiting
A Randomised Controlled Trial of Combinating an Immune Modulator Fingolimod With Alteplase Bridging With Mechanical Thrombectomy in Acute Ischemic Stroke[NCT02956200]Phase 20 participants (Actual)Interventional2016-11-30Withdrawn
Impact of Fingolimod Adherence on Outcomes[NCT05141669]694 participants (Actual)Observational2020-05-18Completed
A 2 Year, Double-blind, Randomized, Multicenter, Active-controlled Core Phase to Evaluate Safety & Efficacy of Daily Fingolimod vs Weekly Interferon β-1a im in Pediatric Patients With Multiple Sclerosis and 5 Year Fingolimod Extension Phase[NCT01892722]Phase 3220 participants (Anticipated)Interventional2013-07-26Recruiting
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
FINGORHYMS - Effects of Fingolimod on Heart Rhythm and Heart Rate Variability in Patients With Multiple Sclerosis[NCT03216915]100 participants (Anticipated)Observational2013-06-01Recruiting
A One-year, Multicenter, Partially Blinded, Double-dummy, Randomized Study to Evaluate the Efficacy and Safety of FTY720 Combined With Reduced-dose or Full-dose Cyclosporine, USP [Modified] (Novartis Brand) and Corticosteroids Versus Mycophenolate Mofetil[NCT00239785]Phase 3684 participants Interventional2003-04-30Completed
A Two-year Extension to a One-year, Multicenter, Partially Blinded, Double-dummy, Randomized Study to Evaluate the Efficacy and Safety of FTY720 Combined With Reduced-dose or Full-dose Cyclosporine, USP [Modified] (Novartis Brand) and Corticosteroids Vers[NCT00239863]Phase 3684 participants Interventional2004-05-31Completed
Two-year Extension of a One-year, Multicenter, Randomized, Partially-blinded Study of the Safety and Efficacy of FTY720 Combined With Corticosteroids and Full or Reduced-dose Cyclosporine, USP [Modified] (Novartis Brand) in de Novo Adult Renal Transplant [NCT00239902]Phase 2396 participants Interventional2002-05-31Completed
Safety and Efficacy of FTY720 in Adult Patients Who Receive a Kidney Transplant[NCT00099801]Phase 30 participants Interventional2005-01-31Completed
A Randomized, Parallel Group, Double-blind, Placebo Controlled, 14 Days Multiple-dose Treatment to Assess the Pulmonary and Cardiac Pharmacodynamics of FTY720 (0.5 and 1.25 mg) in Healthy Volunteers[NCT00416845]Phase 139 participants (Actual)Interventional2006-10-31Completed
Efficacy and Safety of FTY720 in Patients Who Receive a Kidney Transplantation[NCT00098735]Phase 3140 participants Interventional2004-04-30Completed
Safety and Efficacy of FTY720 in Adult Patients Who Receive a Kidney Transplant[NCT00099749]Phase 2/Phase 3255 participants Interventional2003-11-30Completed
The Multi-National Gilenya Pregnancy Exposure Registry in Multiple Sclerosis[NCT01285479]500 participants (Anticipated)Observational [Patient Registry]2011-10-15Recruiting
A Single Arm, Open-label, Multicenter Study Evaluating the Long-term Safety and Tolerability of 0.5 mg Fingolimod (FTY720) Administered Orally Once Daily in Patients With Relapsing Forms of Multiple Sclerosis[NCT01201356]Phase 34,125 participants (Actual)Interventional2010-09-13Completed
An Open-Label, Balanced, Randomized, 2-Treatment, 2-Sequence, 2-Period, Single Dose, Crossover Oral Bioequivalence Study of Two Formulations of Fingolimod Capsules (3 x 0.5 mg) in Healthy Adult Human Subjects Under Fasting Conditions[NCT05145621]Phase 126 participants (Actual)Interventional2015-10-29Completed
Pharmacogenetic Investigation of Susceptibility to Liver Toxicity in Patients With Multiple Sclerosis Treated With Fingolimod[NCT05516303]65 participants (Anticipated)Observational2022-06-07Recruiting
Efficacy and Safety of FTY720 in Patients Who Receive a Kidney Transplant[NCT00099736]Phase 3696 participants (Actual)Interventional2003-05-07Completed
Long-term Follow up of Patients With Relapsing-remitting Multiple Sclerosis Enrolled in the Multicenter, Single-arm, Open-label Biobank Study (CFTY720DDE01), to Investigate Changes in Biomarkers After 48 Months of Treatment With 0.5 mg Fingolimod (FTY720)[NCT02720107]Phase 4133 participants (Actual)Interventional2016-05-12Completed
Safety and Effectiveness of Generic Fingolimod (Sphingomod®, Hikma) in Patients With Relapsing-Remitting Multiple Sclerosis in Egypt[NCT05423769]30 participants (Anticipated)Observational2022-01-19Active, not recruiting
A 24-month Extension of a One-year, Multicenter, Double Blinded Double Dummy, Randomized Study to Evaluate the Safety and Efficacy of Two Doses of FTY720 Combined With Full-dose Cyclosporine, USP [Modified] (Novartis Brand) and Steroids Versus Mycophenola[NCT00239798]Phase 2255 participants Interventional2004-11-30Completed
A Two-year Extension to a One-year, Multicenter, Open-label, Randomised Study to Evaluate the Safety and Efficacy of FTY720 Combined With Tacrolimus and Steroids Versus Mycophenolate Mofetyl Combined With Tacrolimus and Steroids, in de Novo Adult Renal Tr[NCT00239876]Phase 3140 participants Interventional2005-05-31Completed
Revascularization Pretreated With Fingolimod in Acute Stroke[NCT04718064]20 participants (Anticipated)Interventional2021-02-01Not yet recruiting
A 12-month, Open-label, Multicenter Study to Evaluate the Efficacy and Safety of Fingolimod in the Treatment of Relapsing-remitting Multiple Sclerosis Patients With Neutralizing Antibodies to Interferon Beta[NCT01621269]Phase 40 participants (Actual)Interventional2013-06-30Withdrawn
Phase II/III Study to Investigate the Effects of Fingolimod Versus Interferon Beta-1b on Visual Recovery After Optic Neuritis[NCT01647880]Phase 2/Phase 315 participants (Actual)Interventional2013-07-31Terminated(stopped due to delayed recruitment)
A 48-week, Double-blind, Randomized, Multi-center, Parallel-group Study Comparing Structural Changes in the Retina and Evolution of Visual Function After Immediate Versus Delayed Treatment With Fingolimod in Patients With Acute Demyelinating Optic Neuriti[NCT01757691]Phase 22 participants (Actual)Interventional2013-08-31Terminated(stopped due to Discontinuation of this study was based on Novartis decision to discontinue development of fingolimod for the treatment of ADON)
The Long-term Effect of Fingolimod on Circulating Immunocompetent Mononuclear Cells in Patients With Multiple Sclerosis[NCT01755871]Phase 48 participants (Actual)Interventional2013-01-31Terminated(stopped due to lack of recruitment)
A 6-month, Randomized, Active Comparator, Open-label, Multi-Center Study to Evaluate Patient Outcomes, Safety and Tolerability of (Fingolimod) 0.5 mg/Day in Patients With Relapsing Remitting Multiple Sclerosis Who Are Candidates for Multiple Sclerosis (MS[NCT01534182]Phase 4298 participants (Actual)Interventional2012-01-31Completed
A 12-month, Prospective, Multi-center Post-authorization Commitment (PAC) Study Monitoring Safety in Adult Patients With Relapsing-remitting Multiple Sclerosis Newly Initiated on Gilenya (Fingolimod) in Taiwan (SPRING)[NCT04480853]Phase 430 participants (Anticipated)Interventional2020-10-12Recruiting
COMparison Between All immunoTherapies for Multiple Sclerosis. An Observational Long-term Prospective Cohort Study of Safety, Efficacy and Patient's Satisfaction of MS Disease Modulatory Treatments in Relapsing-remitting Multiple Sclerosis[NCT03193866]3,526 participants (Actual)Observational [Patient Registry]2017-06-02Active, not recruiting
A Multicenter, Randomized, Active-controlled Study to Assess the Safety, Tolerability, and Efficacy of FTY720 in Patients With Acute, Noninfectious Intermediate, Posterior and Pan Uveitis[NCT01791192]Phase 20 participants (Actual)Interventional2013-11-30Withdrawn
Effects of Fingolimod (Gilenya®) on Cytokine and Chemokine Levels in Relapsing Remitting Multiple Sclerosis Patients[NCT02373098]Phase 4126 participants (Actual)Interventional2015-03-31Completed
Phase IIa Double-Blind, Placebo-Controlled Study to Evaluate the Safety of Oral Fingolimod in Patients With Amyotrophic Lateral Sclerosis (ALS)[NCT01786174]Phase 230 participants (Actual)Interventional2013-08-31Completed
Safety and Efficacy of Fingolimod in Schizophrenia Patients Who Have Suboptimal Responses to Antipsychotic Drug Treatment[NCT01779700]Phase 240 participants (Actual)Interventional2013-01-31Completed
A 1-week, Open-label, Multi-center Study to Explore Conduction Abnormalities During First Dose Administration of Fingolimod in Patients With Relapsing-remitting Multiple Sclerosis[NCT01585298]Phase 46,998 participants (Actual)Interventional2012-04-29Completed
Effect of Fingolimod on Neurodegeneration, Brain Atrophy and Cognitive Impairment in Relapsing Remitting Multiple Sclerosis Patients[NCT02575365]Phase 44 participants (Actual)Interventional2016-02-16Terminated(stopped due to The trial was terminated due to low enrollment.)
A Double-blind, Randomized, Multicenter, Placebo-controlled, Parallel-group Study to Evaluate the Efficacy and Safety of Fingolimod 0.5 mg Administered Orally Once Daily Versus Placebo in Patients With Chronic Inflammatory Demyelinating Polyradiculoneurop[NCT01625182]Phase 3106 participants (Actual)Interventional2012-12-22Completed
A 32-week, Patient- and Rater-blinded, Randomized, Multi-center, Parallel-group Study to Evaluate Disease Control and Safety in Patients With Relapsing Remitting Multiple Sclerosis Transferred From Previous Treatment With Natalizumab to Fingolimod (FTY720[NCT01499667]Phase 3142 participants (Actual)Interventional2011-09-30Terminated(stopped due to Based on recent publications, determination of natalizumub washout period was no longer relevant.)
A Multi-centre, Open-label, Non-randomised, Parallel Group Clinical Trial to Assess the Efficacy of Fingolimod in Naive Patients Versus Fingolimod in Patients Previously Treated With Interferons or Glatiramer Acetate, Based on the Presence of Relapses in [NCT01498887]Phase 4347 participants (Actual)Interventional2011-12-24Completed
A 21-week, Multicenter, Open Label Study to Evaluate the Safety and Tolerability Profile of the Combination of a SSRI or SNRI Antidepressive Therapy With Oral Fingolimod in the Treatment of RRMS Patients With Mild to Moderate Depression[NCT01436643]Phase 454 participants (Actual)Interventional2011-11-30Terminated(stopped due to Due to slow enrollment the study was terminated early)
A Safety Study of Fingolimod With Radiation and Temozolomide in Newly Diagnosed High Grade Glioma[NCT02490930]Early Phase 15 participants (Actual)Interventional2015-07-31Completed
A 3-year Multi-center Study to Describe the Long Term Changes of Optical Coherence Tomography (OCT) Parameters in Patients Under Treatment With Gilenya®[NCT01705236]Phase 487 participants (Actual)Interventional2012-08-20Completed
A 12 -Month, Open-label, Multi-center Pilot Study to Explore the Health Outcomes of FTY720 in RRMS Patients Who Have Previously Been Treated With Other Disease Modifying Therapies (DMT)- Fine[NCT01578330]Phase 442 participants (Actual)Interventional2012-10-31Completed
A 4-month, Open-label, Multi-center Study to Explore the Safety and Tolerability of Fingolimod 0.5 mg in Patients With Relapsing-remitting Multiple Sclerosis[NCT01497262]Phase 3162 participants (Actual)Interventional2012-02-29Completed
A 12-month, Prospective, Randomized, Active-controlled, Open-label Study to Evaluate the Patient Retention of Fingolimod vs. Approved First-line Disease Modifying Therapies in Adults With Relapsing Remitting Multiple Sclerosis (PREFERMS)[NCT01623596]Phase 4881 participants (Actual)Interventional2012-06-08Completed
Funktionelle Evaluation Des Autonomen Nervensystems im Zusammenhang Mit Der Erstmaligen Einnahme Von 0,5mg Fingolimod (Gilenya) Bei Patienten Mit schubförmig Verlaufender Multipler Sklerose[NCT02048072]Phase 433 participants (Actual)Interventional2013-07-31Completed
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
Fingolimod as a Treatment of Cerebral Edema After Intracerebral Hemorrhage[NCT04088630]Early Phase 128 participants (Actual)Interventional2020-08-07Active, not recruiting
Long-term, Open-label, Multicenter Study Assessing Long-term Cardiovascular Risks in Patients Treated With Fingolimod[NCT02232061]Phase 46 participants (Actual)Interventional2014-09-29Completed
A 12-month, Randomized, Rater- and Dose-blinded Study to Compare the Efficacy and Safety of Fingolimod 0.25 mg and 0.5 mg Administered Orally Once Daily With Glatiramer Acetate 20 mg Administered Subcutaneously Once Daily in Patients With Relapsing-remitt[NCT01633112]Phase 31,064 participants (Actual)Interventional2012-08-09Terminated(stopped due to slow recruitment)
[information is prepared from clinicaltrials.gov, extracted Sep-2024]

Trial Outcomes

Change From Baseline in Patient-reported Activities of Daily Living (ADL)

The PRIMUS activity measure is a 15-item assessment used to evaluate patient-reported activities of daily living. The PRIMUS activities score was calculated summing the 15 items, after recoding the responses from 1 - 3 to 0 - 2. Therefore, the total score ranged from 0 - 3-, where high scores were indicative of greater function limitation. A negative change from baseline indicates improvement. (NCT01317004)
Timeframe: baseline, 6 months

Interventionscore on a scale (Mean)
Fingolimod0.19
Multiple Sclerosis Disease Modifying Treatment (MS DMT)0.15

Change From Baseline in Patient-reported Depression

The Beck Depression Inventory Fast Screen (BDI-FS) is a brief, multiple choice, self reported inventory designed to evaluate depression in patients with medical illness. The BDI-FS score was calculated summing the 7 items of the questionnaire. Each item ranged from 0 (not present) to 3 (severe). The total score ranges from 0-3 (minimal depression), 4-8 (mild depression), 9-12 (moderate depression) and 13-21 (severe depression). A negative change from baseline indicates improvement. (NCT01317004)
Timeframe: 6 months

Interventionscore on a scale (Mean)
Fingolimod-1.15
Multiple Sclerosis Disease Modifying Treatment (MS DMT)-0.12

Change From Baseline in Patient-reported Fatigue

The fatigue Severity Scale (FSS) is a 9-item scale used to assess fatigue. The FSS score was calculated summing the 9 items of the questionnaire and dividing by the number of non-missing items (each item is based on a 7-point Likert scale ranging from 1 (strongly disagree) to 7 (strongly agree)). A negative change from baseline indicates improvement. (NCT01317004)
Timeframe: 6 months

Interventionscore on a scale (Mean)
Fingolimod-0.18
Multiple Sclerosis Disease Modifying Treatment (MS DMT)-0.32

Change From Baseline in Patient-reported Treatment Satisfaction

The Treatment Satisfaction Questionnaire for Medication (TSQM-9) is a psychometric measure of a patient's satisfaction with medication. It consists of 3 subscales: effectiveness, convenience and global satisfaction. The scores were computed by adding items for each domain, i.e. 1 to 3 for effectiveness, 4 - 6 for convenience and 7 to 9 for global satisfaction. The lowest possible score (1 for each item and 3 for all 3 subscales) was subtracted from the composite score and divided by the greatest possible score range. The greatest range was (7-1) X 3 items = 18 for the effectiveness and convenience, and (5-1) x 3 items = 12 for global satisfaction. This provided a transformed score between 0 and 1 that was then multiplied by 100. A positive change from baseline indicates improvement. (NCT01317004)
Timeframe: baseline, 6 months

Interventionscore on a scale (Mean)
Fingolimod19.57
Multiple Sclerosis Disease Modifying Treatment (MS DMT)5.83

Change From Baseline in Patient-Reported Effectiveness and Convenience

The Treatment Satisfaction Questionnaire for Medication (TSQM-9) is a psychometric measure of a patient's satisfaction with medication. It consists of 3 subscales: effectiveness, convenience and global satisfaction. The scores were computed by adding items for each domain, i.e. 1 to 3 for effectiveness, 4 - 6 for convenience and 7 to 9 for global satisfaction. The lowest possible score (1 for each item and 3 for all 3 subscales) was subtracted from the composite score and divided by the greatest possible score range. The greatest range was (7-1) X 3 items = 18 for the effectiveness and convenience, and (5-1) x 3 items = 12 for global satisfaction. This provided a transformed score between 0 and 1 that was then multiplied by 100. A positive change from baseline indicates improvement. (NCT01317004)
Timeframe: 6 months

,
Interventionscore on a scale (Mean)
EffectivenessConvenience
Fingolimod13.5324.64
Multiple Sclerosis Disease Modifying Treatment (MS DMT)-1.6712.78

Change From Baseline in Patient-reported Health Related Quality of Life (QOL)

The SF-36v2 is a validated health-related quality of life instrument used in numerous disease states, including MS. It is a self-administered survey that measures 8 domains of health including: physical functioning, role limitations due to physical health, pain, general health, energy/fatigue, social functioning, role limitations due to emotional problems and emotional well-being. Additionally, two summary scale scores can be calculated: the Physical Component Summary (PCS) and the Mental Component Summary (MCS). If half or more questions within a domain were answered, then a score was calculated for that domain. Otherwise, the patient score for that domain was set to missing. If the patient was missing any 1 of the 8 scale scores, then the physical and mental component scores were set to missing. An algorithm was used to create a score from 0 to 100 for each domain score and component score. A positive change from baseline indicates improvement. (NCT01317004)
Timeframe: 6 months

,
Interventionscore on a scale (Mean)
Physical functioning (n=41,9)Role limitations due to physical health (n=42,9)Pain (n=45,9)General health (n=44,8)Energy/fatigue (n=43,9)Social functioning (n=45,9)Role limitations d/t emotional problems (n=45,9)Emotional well-being (n=43,9)PCS (n=40,8)MCS (n=40,8)
Fingolimod1.717.146.564.522.337.787.042.514.525.88
Multiple Sclerosis Disease Modifying Treatment (MS DMT)-1.115.5614.446.256.486.943.704.897.837.28

Physician-reported Clinical Global Impression of Improvement (CGI-I)

The CGI-I is a rating scale allowing a physician-reported global evaluation of the subject's improvement over time. The Investigator assessed the subject's clinical change relative to the symptoms at baseline on the CGI-I, a seven-point scale, with rating as follows: 1=Very much improved, 2=Much improved, 3=Minimally improved, 4=No change, 5=Minimally worse, 6=Much worse, 7=Very much worse. A lower score and a negative change from baseline indicate improvement. (NCT01317004)
Timeframe: 6 months

,
InterventionPercentage of participants (Number)
Much improvedMinimally improvedNo changeMinimally worse
Fingolimod13.6436.3647.732.27
Multiple Sclerosis Disease Modifying Treatment (MS DMT)11.1111.1166.6711.11

Serum Level of IFN-gamma

5 cc of venous blood is taken from patients and are kept in test tubes without EDTA. Test tubes are kept one hour immobilized so that clotted blood and serum are separated. Then, the serum is divided into four samples of 0.5 cc for immunologic testing of all primary outcomes. Immunologic testing is performed by sandwich ELISA method using Diaclone ® kits (made in France) and according to manufacturer's instructions. (NCT02939079)
Timeframe: 1 year after intervention

InterventionPg/ml (Mean)
Fingolimod and Fish Oil5.92
Fingolimod and Placebo5.99

Serum Level of IFN-gamma

5 cc of venous blood is taken from patients and are kept in test tubes without EDTA. Test tubes are kept one hour immobilized so that clotted blood and serum are separated. Then, the serum is divided into four samples of 0.5 cc for immunologic testing of all primary outcomes. Immunologic testing is performed by sandwich ELISA method using Diaclone ® kits (made in France) and according to manufacturer's instructions. (NCT02939079)
Timeframe: 6 months after intervention

InterventionPg/ml (Mean)
Fingolimod and Fish Oil5.71
Fingolimod and Placebo6.37

Serum Level of IFN-gamma

5 cc of venous blood is taken from patients and are kept in test tubes without EDTA. Test tubes are kept one hour immobilized so that clotted blood and serum are separated. Then, the serum is divided into four samples of 0.5 cc for immunologic testing of all primary outcomes. Immunologic testing is performed by sandwich ELISA method using Diaclone ® kits (made in France) and according to manufacturer's instructions. (NCT02939079)
Timeframe: Baseline

InterventionPg/ml (Mean)
Fingolimod and Fish Oil6.38
Fingolimod and Placebo6.98

Serum Level of IL1b

5 cc of venous blood is taken from patients and are kept in test tubes without EDTA. Test tubes are kept one hour immobilized so that clotted blood and serum are separated. Then, the serum is divided into four samples of 0.5 cc for immunologic testing of all primary outcomes. Immunologic testing is performed by sandwich ELISA method using Diaclone ® kits (made in France) and according to manufacturer's instructions. (NCT02939079)
Timeframe: 1 year after intervention

InterventionPg/ml (Mean)
Fingolimod and Fish Oil13.17
Fingolimod and Placebo13.15

Serum Level of IL1b

5 cc of venous blood is taken from patients and are kept in test tubes without EDTA. Test tubes are kept one hour immobilized so that clotted blood and serum are separated. Then, the serum is divided into four samples of 0.5 cc for immunologic testing of all primary outcomes. Immunologic testing is performed by sandwich ELISA method using Diaclone ® kits (made in France) and according to manufacturer's instructions. (NCT02939079)
Timeframe: 6 months after intervention

InterventionPg/ml (Mean)
Fingolimod and Fish Oil13.61
Fingolimod and Placebo13.47

Serum Level of IL1b

5 cc of venous blood is taken from patients and are kept in test tubes without EDTA. Test tubes are kept one hour immobilized so that clotted blood and serum are separated. Then, the serum is divided into four samples of 0.5 cc for immunologic testing of all primary outcomes. Immunologic testing is performed by sandwich ELISA method using Diaclone ® kits (made in France) and according to manufacturer's instructions. (NCT02939079)
Timeframe: Baseline

InterventionPg/ml (Mean)
Fingolimod and Fish Oil13.76
Fingolimod and Placebo14

Serum Level of IL6

5 cc of venous blood is taken from patients and are kept in test tubes without EDTA. Test tubes are kept one hour immobilized so that clotted blood and serum are separated. Then, the serum is divided into four samples of 0.5 cc for immunologic testing of all primary outcomes. Immunologic testing is performed by sandwich ELISA method using Diaclone ® kits (made in France) and according to manufacturer's instructions. (NCT02939079)
Timeframe: 1 year after intervention

InterventionPg/ml (Mean)
Fingolimod and Fish Oil6.17
Fingolimod and Placebo6.05

Serum Level of IL6

5 cc of venous blood is taken from patients and are kept in test tubes without EDTA. Test tubes are kept one hour immobilized so that clotted blood and serum are separated. Then, the serum is divided into four samples of 0.5 cc for immunologic testing of all primary outcomes. Immunologic testing is performed by sandwich ELISA method using Diaclone ® kits (made in France) and according to manufacturer's instructions. (NCT02939079)
Timeframe: 6 months after intervention

InterventionPg/ml (Mean)
Fingolimod and Fish Oil6.65
Fingolimod and Placebo6.17

Serum Level of IL6

5 cc of venous blood is taken from patients and are kept in test tubes without EDTA. Test tubes are kept one hour immobilized so that clotted blood and serum are separated. Then, the serum is divided into four samples of 0.5 cc for immunologic testing of all primary outcomes. Immunologic testing is performed by sandwich ELISA method using Diaclone ® kits (made in France) and according to manufacturer's instructions. (NCT02939079)
Timeframe: Baseline

InterventionPg/ml (Mean)
Fingolimod and Fish Oil6.19
Fingolimod and Placebo6.33

Serum Level of TNF-α

5 cc of venous blood is taken from patients and are kept in test tubes without EDTA. Test tubes are kept one hour immobilized so that clotted blood and serum are separated. Then, the serum is divided into four samples of 0.5 cc for immunologic testing of all primary outcomes. Immunologic testing is performed by sandwich ELISA method using Diaclone ® kits (made in France) and according to manufacturer's instructions. (NCT02939079)
Timeframe: 1 year after intervention

InterventionPg/ml (Mean)
Fingolimod and Fish Oil21.20
Fingolimod and Placebo16.47

Serum Level of TNF-α

5 cc of venous blood is taken from patients and are kept in test tubes without EDTA. Test tubes are kept one hour immobilized so that clotted blood and serum are separated. Then, the serum is divided into four samples of 0.5 cc for immunologic testing of all primary outcomes. Immunologic testing is performed by sandwich ELISA method using Diaclone ® kits (made in France) and according to manufacturer's instructions. (NCT02939079)
Timeframe: 6 months after intervention

InterventionPg/ml (Mean)
Fingolimod and Fish Oil28.03
Fingolimod and Placebo17.18

Serum Level of TNF-α

5 cc of venous blood is taken from patients and are kept in test tubes without ethylenediaminetetraacetic acid (EDTA). Test tubes are kept one hour immobilized so that clotted blood and serum are separated. Then, the serum is divided into four samples of 0.5 cc for immunologic testing of all primary outcomes. Immunologic testing is performed by sandwich ELISA method using Diaclone ® kits (made in France) and according to manufacturer's instructions. (NCT02939079)
Timeframe: Baseline

InterventionPg/ml (Mean)
Fingolimod and Fish Oil37.16
Fingolimod and Placebo28.79

Change From Baseline for New Gd-Enhancing T1 Lesion Count

(NCT03257358)
Timeframe: Baseline to Month 12

,
Interventionnumber of lesions (Mean)
Baseline (BL) n=125,147Month 12 n=33,28Change from BL to Month 12 n=33,28
Cohort 10.40.1-0.2
Cohort 20.20.20.2

Change From Baseline in Patient Determined Disease Steps (PDDS)

PDDS scoring ranges 0 to 8. 0 = Normal; 1 = Mild disability; 2 = Moderate disability; 3 = Gait disability; 4 = Early cane; 5 = Late cane; 6 = Bilateral support; 7 = Wheelchair/scooter; 8 = Bedridden. (NCT03257358)
Timeframe: Baseline to Month 12

,
Interventionscores (Mean)
Baseline (BL) n=163,217Month 12 n=103,188Change from BL to Month 12 n=103,188
Cohort 11.71.8-0.1
Cohort 21.81.8-0.0

Change From Baseline in T2 Lesion Burden

(NCT03257358)
Timeframe: Baseline to Month 12

,
Interventionnumber of lesions (Mean)
Baseline (BL) n=91,84Month 12 n=21,17Change from BL to Month 12 n=21,17
Cohort 18.16.5-0.8
Cohort 29.713.13.2

Change From Baseline to Month 12 in CD4+ Central Memory T Cells (CCR7+CD45RA-CD45RO+)

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 12

,
Interventioncells/uL (Mean)
Baseline (BL) n=147,188Change from BL to Month 12 n=83,150
Cohort 1374.6-356.8
Cohort 216.33.9

Change From Baseline to Month 12 in CD4+ Effector Memory T Cells (CCR7-CD45RA-CD45RO+)

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 12

,
Interventioncells/uL (Mean)
Baseline (BL) n=147,188Change from BL to Month 12 n=83,150
Cohort 174.3-50.4
Cohort 222.8-3.3

Change From Baseline to Month 12 in CD4+ Naive T Cells (CCR7+CD45RA+)

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 12

,
Interventioncells/uL (Mean)
Baseline (BL) n=147,188Change from BL to Month 12 n=82,150
Cohort 1404.4-376.3
Cohort 23.4-0.8

Change From Baseline to Month 12 in CD4+ Th1 Cells (CXCR3+)

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 12

,
Interventioncells/uL (Mean)
Baseline (BL) n=148,211Change from BL to Month 12 n=88,175
Cohort 153.8-42.3
Cohort 211.1-2.1

Change From Baseline to Month 12 in CD4+ Th17 Cells (CCR6+)

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 12

,
Interventioncells/uL (Mean)
Baseline (BL) n=148,212Change from BL to Month 12 n=88,175
Cohort 155.5-52.1
Cohort 22.40.5

Change From Baseline to Month 12 in CD4+ Th2 Cells (CCR4+)

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 12

,
Interventioncells/uL (Mean)
Baseline (BL) n=148,212Change from BL to Month 12
Cohort 135.9-36.3
Cohort 21.10.4

Change From Baseline to Month 12 in CD8+ Central Memory T Cells (CCR7+CD45RA-CD45RO+)

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 12

,
Interventioncells/uL (Mean)
Baseline (BL) n=147,188Change from BL to Month 12 n=83,150
Cohort 193.1-86.5
Cohort 25.60.0

Change From Baseline to Month 12 in CD8+ Effector Memory T Cells (CCR7-CD45RA-CD45RO+)

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 12

,
Interventioncells/uL (Mean)
Baseline (BL) n=147,188Change from BL to Month 12 n=83,150
Cohort 1108.2-55.9
Cohort 263.8-15.9

Change From Baseline to Month 12 in CD8+ Naive T Cells (CCR7+CD45RA+)

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 12

,
Interventioncells/uL (Mean)
Baseline (BL) n=147,188Change from BL to Month 12 n=83,150
Cohort 1150.9-126.3
Cohort 21.80.6

Change From Baseline to Month 12 in Memory B Lymphocytes (CD19+CD27+)

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 12

,
Interventioncells/uL (Mean)
Baseline (BL) n=144,212Change from BL to Month 12 n=82,174
Cohort 161.8-46.4
Cohort 23.31.4

Change From Baseline to Month 12 in Monocytes (CD14+)

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 12

,
Interventioncells/uL (Mean)
Baseline (BL) n=150,197Change from BL to Month 12 n=86,164
Cohort 1329.657.1
Cohort 2251.7112.4

Change From Baseline to Month 12 in Naive B Lymphocytes (CD19+CD27-)

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 12

,
Interventioncells/uL (Mean)
Baseline (BL) n=144,212Change from BL to Month 12 n=82,174
Cohort 1201.11-177.0
Cohort 218.1-0.5

Change From Baseline to Month 12 in Neutrophils (CD16+)

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 12

,
Interventioncells/uL (Mean)
Baseline (BL) n=149,197Change from BL to Month 12 n=87, 164
Cohort 14041.4-815.9
Cohort 23717.9-345.0

Change From Baseline to Month 12 in NK Cells (CD56+)

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 12

,
Interventioncells/uL (Mean)
Baseline (BL) n=149,197Change from BL to Month 12 n=87,164
Cohort 1166.4-32.6
Cohort 2181.0-28.9

Change From Baseline to Month 12 in Regulatory B Lymphocytes (CD19+CD24+CD38+)

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 12

,
Interventioncells/uL (Mean)
Baseline (BL) n=144,212Change from BL to Month 12 n=82,174
Cohort 112.3-7.7
Cohort 25.30.8

Change From Baseline to Month 12 in Total CD19+ Absolute Cell Count

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 12

,
Interventioncells/uL (Mean)
Baseline (BL) n=151,213Change from BL to Month 12 n=88,176
Cohort 1259.7-218.9
Cohort 221.41.0

Change From Baseline to Month 12 in Total CD19+ Differential Cell Count (%)

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 12

,
Interventioncells/uL (Mean)
Baseline (BL) n=152,213Change from BL to Month 12 n=89,176
Cohort 113.97-8.86
Cohort 24.810.20

Change From Baseline to Month 12 in Total CD4+ Absolute Cell Count

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 12

,
Interventioncells/uL (Mean)
Baseline (BL) n=156,213Change from BL to Month 12 n=94,176
Cohort 1936.3-844.9
Cohort 264.40.7

Change From Baseline to Month 12 in Total CD4+ Differential Cell Count (%)

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 12

,
Interventioncells/uL (Mean)
Baseline (BL) n=157,213Change from BL to Month 12 n=95,176
Cohort 149.40-37.90
Cohort 211.950.70

Change From Baseline to Month 12 in Total CD8+ Absolute Cell Count

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 12

,
Interventioncells/uL (Mean)
Baseline (BL) n=156,213Change from BL to Month 12 n=94,176
Cohort 1419.9-265.1
Cohort 2124.6-11.6

Change From Baseline to Month 12 in Total CD8+ Differential Cell Counts (%)

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 12

,
Interventioncells/uL (Mean)
Baseline (BL) n=157,213Change from BL to Month 12 n=95,176
Cohort 121.864.33
Cohort 225.250.34

Change From Baseline to Month 6 in CD4+ Central Memory T Cells (CCR7+CD45RA-CD45RO+)

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 6

,
Interventioncells/uL (Mean)
Baseline (BL) n=147,188Month 6 n=97,156Change from BL to Month 6, n=97,156
Cohort 1374.619.6-368.9
Cohort 216.318.31.1

Change From Baseline to Month 6 in CD4+ Effector Memory T Cells (CCR7-CD45RA-CD45RO+)

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 6

,
Interventioncells/uL (Mean)
Baseline (BL) n=147,188Month 6, n=97,156Change from BL to Month 6 n=97,156
Cohort 174.318.2-51.5
Cohort 222.821.7-2.0

Change From Baseline to Month 6 in CD4+ Naive T Cells (CCR7+ CD45RA+)

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 6

,
Interventioncells/uL (Mean)
Baseline (BL) n=147,188Month 6 n=97,156Change from BL to Month 6, n=97,156
Cohort 1404.47.6-411.4
Cohort 23.44.80.8

Change From Baseline to Month 6 in CD4+ Th1 Cells (CXCR3+)

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 6

,
Interventioncells/uL (Mean)
Baseline (BL) n=148,211Month 6 n=104,180Change from BL to Month 6 n=104,180
Cohort 153.87.6-43.6
Cohort 211.111.7-0.7

Change From Baseline to Month 6 in CD4+ Th17 Cells (CCR6+)

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 6

,
Interventioncells/uL (Mean)
Baseline (BL) n=148,212Month 6 n=104,181Change from BL to Month 6 n=104,181
Cohort 155.53.1-53.2
Cohort 22.42.80.5

Change From Baseline to Month 6 in CD4+ Th2 Cells (CCR4+)

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 6

,
Interventioncells/uL (Mean)
Baseline (BL) n=148,212Month 6 n=104,181Change from BL to Month 6 n=104,181
Cohort 135.91.7-36.1
Cohort 21.11.60.5

Change From Baseline to Month 6 in CD8+ Central Memory T Cells (CCR7+CD45RA-CD45RO+)

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 6

,
Interventioncells/uL (Mean)
Baseline (BL) n=147,188Month 6 n=97,156Change from BL to Month 6 n=97,156
Cohort 193.15.9-85.2
Cohort 25.66.10.0

Change From Baseline to Month 6 in CD8+ Effector Memory T Cells (CCR7-CD45RA-CD45RO+)

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 6

,
Interventioncells/uL (Mean)
Baseline (BL) n=147,188Month 6 n=97,156Change from BL to Month 6 n=97,156
Cohort 1108.255.4-40.6
Cohort 263.852.6-12.5

Change From Baseline to Month 6 in CD8+ Naive T Cells (CCR7+CD45RA+)

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 6

,
Interventioncells/uL (Mean)
Baseline (BL) n=147,188Month 6 n=97,156Change from BL to Month 6 n=97,156
Cohort 1150.94.2-139.3
Cohort 21.83.31.2

Change From Baseline to Month 6 in Memory B Lymphocytes (CD19+CD27+)

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 6

,
Interventioncells/uL (Mean)
Baseline (BL) n=144,212Month 6 n=101,176Change from BL to Month 6 n=101,176
Cohort 161.83.8-55.1
Cohort 23.34.00.3

Change From Baseline to Month 6 in Monocytes (CD14+)

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 6

,
Interventioncells/uL (Mean)
Baseline (BL) n=150,197Month 6 n=105,166Change from BL to Month 6 n=105,166
Cohort 1329.6384.766.1
Cohort 2251.7379.2123.1

Change From Baseline to Month 6 in Naive B Lymphocytes (CD19+CD27-)

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 6

,
Interventioncells/uL (Mean)
Baseline (BL) n=144,212Month 6 n=101,176Change from BL to Month 6 n=101,176
Cohort 1201.115.0-181.4
Cohort 218.117.8-0.2

Change From Baseline to Month 6 in Neutrophils (CD16+)

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 6

,
Interventioncells/uL (Mean)
Baseline (BL) n=149,197Month 6 n=105,166Change from BL to Month 6 n=105,166
Cohort 14041.43505.4-586.0
Cohort 23717.93312.6-439.6

Change From Baseline to Month 6 in NK Cells (CD56+)

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 6

,
Interventioncells/uL (Mean)
Baseline (BL) n=149,197Month 6 n=105,166Change from BL to Month 6 n=105,166
Cohort 1166.4133.6-29.4
Cohort 2181.0154.5-28.0

Change From Baseline to Month 6 in Regulatory B Lymphocytes (CD19+CD24+CD38+)

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 6

,
Interventioncells/uL (Mean)
Baseline (BL) n=144,212Month 6 n=101,176Change from BL to Month 6 n=101,176
Cohort 112.34.8-7.4
Cohort 25.36.10.9

Change From Baseline to Month 6 in Total CD19+ Absolute Cell Count

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 6

,
Interventioncells/uL (Mean)
Baseline (BL) n=151,213Month 6 n=106,179Change from BL to Month 6 n=106,179
Cohort 1259.719.5-231.3
Cohort 221.421.80.1

Change From Baseline to Month 6 in Total CD19+ Differential Cell Count (%)

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 6

,
Interventioncells/uL (Mean)
Baseline (BL) n=152,213Month 6 n=107,180Change from BL to Month 6 n=107,180
Cohort 113.975.38-8.53
Cohort 24.814.830.23

Change From Baseline to Month 6 in Total CD4+ Absolute Cell Count

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 6

,
Interventioncells/uL (Mean)
Baseline (BL) n=156,213Month 6 n=110,182Change from BL to Month 6 n=110,182
Cohort 1936.353.4-884.1
Cohort 264.471.31.4

Change From Baseline to Month 6 in Total CD4+ Differential Cell Count

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 6

,
Interventioncells/uL (Mean)
Baseline (BL) n=157,213Month 6 n=111,183Change from BL to Month 6 n=111,183
Cohort 149.4011.08-39.09
Cohort 211.9512.820.32

Change From Baseline to Month 6 in Total CD8+ Absolute Cell Count

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 6

,
Interventioncells/uL (Mean)
Baseline (BL) n=156,213Month 6 n=110,182Change from BL to Month 6 n=110,182
Cohort 1419.9120.1-266.2
Cohort 2124.6116.8-13.5

Change From Baseline to Month 6 in Total CD8+ Differential Cell Counts (%)

Blood samples (approximately 60-80 ml) were collected at specifiied visits for biomarker and hematology assessments. In Cohort 1 patients it was critical that the blood sample was collected prior to administration of fingolimod, first dose observation (FDO). A central laboratory was used for analysis of all specimens collected. (NCT03257358)
Timeframe: Baseline to Month 6

,
Interventioncells/uL (Mean)
Baseline (BL) n=157,213Month 6 n=111,183Change from BL to Month 6
Cohort 121.8625.334.63
Cohort 225.2524.99-0.39

Change From Baseline to Months 6 and 12 in the Anti-JCV Antibody Index (Index/Value)

(NCT03257358)
Timeframe: Baseline to Month 6 and 12

,
Interventioncells/uL (Mean)
Baseline (BL) n=159,215Change from BL to Month 6 n=116,195Change from BL to Month 12 n=100,180
Cohort 11.2730.0380.040
Cohort 21.3910.0450.145

Multiple Sclerosis (MS) Relapses During Treatment

A relapse is defined as the appearance of a new neurological abnormality or worsening of previously stable or improving pre-existing neurological abnormality, separated by at least 30 days from onset of a preceding clinical demyelinating event. The abnormality must be present for at least 24 hours and occur in the absence of fever (<37.5°C) or infection. (NCT03257358)
Timeframe: Baseline to Month 12

,
Interventionrelapses (Number)
Number of patients with relapsesTotal number of relapsesRelapses not requiring steroid useRelapses requiring steroid useMild relapseModerate relapseSevere relapse
Cohort 1111257561
Cohort 2131358850

Number of Participants Who Received Steroid Treatment for MS Relapses During Treatment

A relapse is defined as the appearance of a new neurological abnormality or worsening of previously stable or improving pre-existing neurological abnormality, separated by at least 30 days from onset of a preceding clinical demyelinating event. The abnormality must be present for at least 24 hours and occur in the absence of fever (<37.5°C) or infection. (NCT03257358)
Timeframe: Baseline to Month 12

,
InterventionParticipants (Count of Participants)
Patients with ≥ 1 relapsesPatients with relapse who received ≥ 1 steroidCorticosteroids for systemic useMethylprednisolone sodium succinateMethylprednisolonePrednisone
Cohort 11177430
Cohort 21388423

Change From Baseline in European Quality of Life - 5 Dimensions (EQ-5D Score)

EQ-5D: participant rated questionnaire to assess health-related quality of life in terms of a single utility score. Health State Profile component assesses level of current health for 5 domains: mobility, self care, usual activities, pain and discomfort, and anxiety and depression; 1 indicates better health state (no problems); 3 indicates worst health state (confined to bed). Scoring formula developed by EuroQoL Group assigns a utility value for each domain in the profile. Score is transformed and results in a total score range -0.594 to 1.000; higher score indicates a better health state. (NCT00731692)
Timeframe: Baseline, 36 months

InterventionScore on a scale (Mean)
FTY720 1.25 mg to 0.5 mg-0.0332
FTY720 0.5 mg to 0.5 mg-0.0475
Placebo-0.0539

Change From Baseline in Multiple Sclerosis Walking Scale (MSWS-12 Score)

The Multiple Sclerosis Walking Scaleis a patient reported measure of walking quality (Hobart et al 2003), consisting of 12 items asking patients to rate the impact of MS upon their walking ability. Responses were captured on a 3-point scale ranging from 1 (Not at all) to 3 (A lot) for items 1 to 3 and on a 5-point scale ranging from 1 (not limited) to 5 (extremely) for items 4 to 12. All 12 item scores were summed to obtain a total score ranging from 12 (good) to 54 (poor) which is the MSWS-12 scale score. The total score was transformed to a 0 to 100 scale score. The MSWS-12 scale score will be transformed to a 0-100 scale score before any summaries or statistical analyses are performed. The transformed score is obtained by subtracting 12 and divided by 42 and multiplying by 100 (i.e., transformed scale score = (raw scale score- 12)/42*100). (NCT00731692)
Timeframe: Baseline, 36 months

InterventionScore on a scale (Mean)
FTY720 1.25 mg to 0.5 mg6.4444
FTY720 0.5 mg to 0.5 mg5.5616
Placebo9.5899

Change From Baseline in PRIMUS-Activities

The activities subscale of PRIMUS contains 15 items and each item is given a score of 0 (able to do on own without difficulties), 1 (able to do on own with difficulties), or 2 (unable to do on own). All 15 items were summed to obtain a total score ranging from 0 (good) to 30 (poor). (NCT00731692)
Timeframe: Baseline, 36 months

InterventionScore on a scale (Mean)
FTY720 1.25 mg to 0.5 mg3.5504
FTY720 0.5 mg to 0.5 mg2.6324
Placebo2.8830

Change From Baseline in the Patient Reported Indices in Multiple Sclerosis (PRIMUS-QoL Score)

The quality of life scale contains 22 items. Each item will be given a score of 1 or 0. A score of 1 (or 0) indicates the presence (or absence) of the symptom or adverse quality of life. All 22 item scores will be summed to obtain a total score ranging from 0 (good) to 22 (poor), which is the PRIMUS QoL scale score (NCT00731692)
Timeframe: Baseline, 36 months

InterventionScore on a scale (Mean)
FTY720 1.25 mg to 0.5 mg0.2424
FTY720 0.5 mg to 0.5 mg0.5921
Placebo0.9597

Change From Baseline in Unidimensional Fatigue Impact (U-FIS) Score

Unidimensional Fatigue Impact Scale (U-FIS), contains 22 patient-reported items that assess the impact of fatigue on cognitive, physical, and psychosocial functioning. Responses formed a single unidimensional scale measuring fatigue impact. The U-FIS was calculated and analyzed according to the U-FIS scoring manual. The U-FIS scale contains 22 items with 5 possible outcomes for each item. Two response categories (about half the time and a lot of the time) were combined into 1 category to obtain 4 possible outcomes: 0 (never), 1 (a little of the time), 2 (about half the time/a lot of the time), and 3 (all the time). The 22 condensed item scores were summed to obtain a total score ranging from 0 (no fatigue) to 66 (severe fatigue impact). (NCT00731692)
Timeframe: Baseline, 36 months

InterventionScore on a scale (Mean)
FTY720 1.25 mg to 0.5 mg1.3197
FTY720 0.5 mg to 0.5 mg2.8451
Placebo3.1394

Change in MSFC Z-score and Subscale Scores From Baseline to Month 36

The Multiple Sclerosis Functional Composite (MSFC) is a multidimensional clinical outcome measure that includes quantitative tests of leg function/ambulation (Timed 25-Foot Walk), arm function (9-Hole Peg Test), and cognitive function (Paced Auditory Serial Addition Test). The overall MSFC z-score as an average of the three standardized scores derived using baseline data pooled over each treatment arm as reference population. Higher scores reflect better neurological function and a positive change from Baseline indicates improvement. (NCT00731692)
Timeframe: Baseline to Month 36

InterventionZ-scores (Mean)
FTY720 0.5 mg-0.189
Placebo-0.212

Kaplan Meier Estimate -Percentage of Participants With 3- Month Confirmed Disability Progression Based on 25' TWT.

The 25' TWT is a quantitative measure of lower extremity function designed and validated for evaluation of MS patients. N= Total number of patients included in the analysis (NCT00731692)
Timeframe: up to 36 months after the last patient was randomized

InterventionPercentage of Participants (Number)
FTY720 0.5 mg to 0.5 mg54.8
Placebo56.7

Kaplan Meier Estimate -Percentage of Participants With 3- Month Confirmed Disability Progression Based on 9-HPT.

The 9-HPT is a quantitative measure of upper extremity (arm and hand) function designed and validated for evaluation of MS patients. N= Total number of patients included in the analysis (NCT00731692)
Timeframe: up to 36 months after the last patient was randomized

InterventionPercentge of Participants (Number)
FTY720 0.5 mg to 0.5 mg25.0
Placebo24.9

Kaplan-Meier Estimate of the Risk of 3- Month Confirmed Disability Progression Based on Expanded Disability Status Scale (EDSS)

The Expanded Disability Status Scale (EDSS) is a scale for assessing neurologic impairment in MS (Kurtzke 1983) and includes a series of scores in each of 8 functional systems and the EDSS steps (ranging from 0 (normal) to 10 (death due to MS)). The functional systems are Visual, Brain Stem, Pyramidal, Cerebellar, Sensory, Bowel and Bladder, Cerebral and Other functions. Fatigue is not included in the Cerebral score of the EDSS. The score ranges from 0 (normal) to 10 (death due to MS) (NCT00731692)
Timeframe: up to 36 months after the last patient was randomized

InterventionPercentage of Participants (Number)
FTY720 0.5 mg to 0.5 mg54.3
Placebo58.7

Kaplan-Meier Estimate of the Risk of 3-month Confirmed Disability Progression Based on Composite Endpoint

3-month sustained increase from Baseline in EDSS (at least 1 point increase from Baseline for patients with a Baseline value of 5 or less or at least 0.5 point increase from Baseline for patients with a Baseline value of 5.5 or more) or 3-month sustained increase of at least 20% from BL in the time taken to complete the timed 25-foot walk test (25' TWT); or 3-month sustained increase of at least 20% from BL in the time taken to complete the 9-HPT. The 25' TWT is a quantitative measure of lower extremity function. The EDSS is a scale assessing neurologic impairment, including a series of scores in each of 8 functional systems: Visual, Brain Stem, Pyramidal, Cerebellar, Sensory, Bowel and Bladder, Cerebral and Other functions. The score ranges from 0 (normal) to 10 (death due to MS)). The 9-hole peg test (9-HPT) is a quantitative measure of upper extremity (arm and hand) function. (NCT00731692)
Timeframe: up to 36 months after the last patient was randomized

InterventionPercentage of Participants (Number)
FTY720 0.5 mg to 0.5 mg77.2
Placebo80.3

Number of Gd-enhancing Lesions at Month 36

Inflammatory disease, as measured by number of T1 Gd-enhancing lesions, was assessed by MRI scanning of the brain and full spinal cord. N= Total number of patients included in the analysis (NCT00731692)
Timeframe: Baseline to 36 months

InterventionGd-enhanced lesions per patient per scan (Least Squares Mean)
FTY720 0.5 mg to 0.5 mg0.05
Placebo0.21

Number of New/Enlarging T2 Lesions Per Year Measured From Baseline to Month 36

Inflammatory disease, as measured by number of new or newly-enlarging T2 lesions, was assessed by Magnetic resonance Imaging (MRI) scanning of the brain and full spinal cord. N= Total number of patients included in the analysis (NCT00731692)
Timeframe: Baseline to 36 months

InterventionT2 Lesions per year (Least Squares Mean)
FTY720 0.5 mg to 0.5 mg0.13
Placebo0.50

Percent Change From Baseline in Brain Volume at Month 36

The percent change from Baseline in brain volume was analyzed using a random coefficients model. The model included: 1) fixed effects: treatment and region and 2) continuous covariates: time, number of Gd enhancing lesions at Baseline, Baseline T2 volume, and normalized brain volume at Baseline. Time as a continuous covariate allowed for the estimation of different slopes and intercepts among treatment groups. (NCT00731692)
Timeframe: Baseline to month 36

InterventionPercent Change (Least Squares Mean)
FTY720 0.5 mg to 0.5 mg-1.49
Placebo-1.53

Percent Change in Total T2 Lesion Volume From Baseline to Month 36

Inflammatory disease as measured by percent change in total T2 lesion volume (mm3) was assessed by MRI. N= Total number of patients included in the analysis (NCT00731692)
Timeframe: Baseline to month 36

InterventionPercent Change (Mean)
FTY720 0.5 mg to 0.5 mg-9.2
Placebo8.9

Blood Concentrations of Fingolimod and Fingolimod-phosphate

"Concentrations of fingolimod and fingolimod-phosphate in whole blood were determined by validated liquid chromatography methods with tandem mass spectrometry. The lower limits of quantification were 0.08 ng/ml for fingolimod and 0.1 ng/ml for fingolimod-phosphate.~Venous blood samples were collected for the analysis." (NCT00731692)
Timeframe: Month 3 up to 36 months

,,
Interventionng/ml (Mean)
Month 3 Fingolimod (n=64, 179)Month 12 Fingolimod (n=23, 161)Month 18 Fingolimod (n=71,155)Month 24 Fingolimod (n=67, 160)Month 30 Fingolimod (n=62, 158)Month 36 Fingolimod (n=55, 118)End of treatment Fingolimod (n=32, 115)Month 3 Fingolimod-Phosphate (n=64, 179)Month 12 Fingolimod-Phosphate (n=23, 161)Month 18 Fingolimod-Phosphate (n=71,155)Month 24 Fingolimod-Phosphate (n=67, 160)Month 30 Fingolimod-Phosphate (n=62, 158)Month 36 Fingolimod-Phosphate (n=55, 118)End of treatment Fingolimod-Phosphate (n=32, 115
FTY720 0.5 mg to 0.5 mg2.582.552.592.642.602.632.571.401.431.411.441.481.511.50
FTY720 1.25 mg to 0.5 mg6.046.24NANANANANA3.203.21NANANANANA
FTY720 1.25mg to 0.5 mgNA2.872.442.412.522.442.02NA1.541.341.351.321.321.19

Change From Screening in Delis-Kaplan Executive Function System (DKEFS) Condition 1: Free Sorting, Confirmed Correct Sort- Card Set 1+2

The Delis-Kaplan Executive Function System - Sorting Test is one of the nine tests presented in the DKEFS manual and explores the patient's executive abilities. It has a standard form (version A, administered at screening and Month-18 visit) and an alternate form (version B, administered at Month-9 visit). The standard form consists of the practice card set, card set 1 and card set 2. The alternate form consists of the same practice card set, card set 3 and card set 4. The DKFES test consisted of two testing procedures: free sorting and sort recognition. In free sorting, six scores were obtained: Confirmed Correct sorts for card sets 1 and 2 (or 3 and 4 for version B), sum of confirmed Correct Sorts. In sort recognition, a description score for card set 1 and 2 (or 3 and 4 for version B) was obtained, as well as the sum of description scores of both sets. The total score ranged from 0 to 16. Higher values represent a better outcome (NCT01333501)
Timeframe: Screening (-1month), 18 month

InterventionRaw score (Least Squares Mean)
Fingolimod0.57
Interferon Beta 1b0.82

Change From Screening in DKEFS Condition 1: Free Sorting, Free Sorting, Description Score, Card Set 1+2

The Delis-Kaplan Executive Function System - Sorting Test is one of the nine tests presented in the DKEFS manual and explores the patient's executive abilities. It has a standard form (version A, administered at screening and Month-18 visit) and an alternate form (version B, administered at Month-9 visit). The standard form consists of the practice card set, card set 1 and card set 2. The alternate form consists of the same practice card set, card set 3 and card set 4. In free sorting, six scores were obtained: Confirmed Correct sorts for card sets 1 and 2 (or 3 and 4 for version B), sum of confirmed Correct Sorts, Free Sorting Description score for card set 1 and 2 (or 3 and 4 for version B) and sum of Free Sorting Description scores. In sort recognition, a description score for card set 1 and 2 (or 3 and 4 for version B) was obtained, as well as the sum of description scores of both sets. The total score ranged from 0 to 64. Higher values represent a better outcome (NCT01333501)
Timeframe: Screening (-1month), 18 month

InterventionRaw score (Least Squares Mean)
Fingolimod1.52
Interferon Beta 1b3.69

Change From Screening in DKEFS Condition 2: Sort Recognition, Sort Recognition Description Score- Card Set 1+2

The Delis-Kaplan Executive Function System - Sorting Test is one of the nine tests presented in the DKEFS manual and explores the patient's executive abilities. It has a standard form (version A, administered at screening and Month-18 visit) and an alternate form (version B, administered at Month-9 visit). The standard form consists of the practice card set, card set 1 and card set 2. The alternate form consists of the same practice card set, card set 3 and card set 4. Free sorting and sort recognition. In free sorting, six scores were obtained: Confirmed Correct sorts for card sets 1 and 2 (or 3 and 4 for version B), sum of confirmed Correct Sorts, Free Sorting Description score for card set 1 and 2 (or 3 and 4 for version B) and sum of Free Sorting Description scores. The total score ranged from 0 to 64. Higher values represent a better outcome. (NCT01333501)
Timeframe: Screening (-1month), 18 month

InterventionRaw score (Least Squares Mean)
Fingolimod2.15
Interferon Beta 1b7.38

Change From Screening in Montgomery-Asberg Depression Rating Scale (MADRS)

MADRS measures the overall severity of depressive symptoms. The MADRS had a 10-item checklist. Items are rated on a scale of 0-6, for a total numeric range of scores from 0 (depressive symptoms absent) to 60 (numerically highest level of depressive symptoms). (NCT01333501)
Timeframe: Screening (-1month), 18 month

InterventionRaw score (Least Squares Mean)
Fingolimod-0.77
Interferon Beta 1b0.13

Change From Screening in Paced Auditory Serial Addition Test - 2 (PASAT 2) Raw Score

Paced Auditory Serial Addition Test (PASAT) for working memory (and sustained attention and information processing speed). The patient hears a series of numbers from recordings that are presented at the rate of one every 2 seconds in the second part of the test (PASAT-2). The patient was asked to add each consecutive digit to the one immediately preceding it. Sixty-one digits are presented for each part, and each part has a maximum of 60 correct answers. The total score ranged from 0 to 60. Higher values represent a better outcome. (NCT01333501)
Timeframe: Screening (-1month), 18 month

InterventionRaw score (Least Squares Mean)
Fingolimod4.79
Interferon Beta 1b4.42

Change From Screening in Paced Auditory Serial Addition Test - 3 Seconds (PASAT 3) Raw Score

Paced Auditory Serial Addition Test (PASAT) for working memory (and sustained attention and information processing speed). The patient hears a series of numbers from recordings that are presented at the rate of one every 3 seconds in the first part of the test (PASAT-3). The patient is asked to add each consecutive digit to the one immediately preceding it. Sixty-one digits are presented for each part, and each part has a maximum of 60 correct answers. The total score ranged from 0 to 60. Higher values represent a better outcome. (NCT01333501)
Timeframe: Screening (-1month), 18 month

InterventionRaw score (Least Squares Mean)
Fingolimod7.14
Interferon Beta 1b6.68

Change From Screening in Selective Reminding Test - Consistent Long Term Retrieval (SRT-CLTR) Raw Score

Brief Repeatable Battery (BRB)- widely used as a clinical and research tool, with 68% sensitivity and 85% specificity. It consists of the serial administration of 5 tests. One of the tests is SRT (Selective Reminding Test) for episodic memory (verbal learning and delayed recall). A word recalled on two consecutive trials is considered to have entered long-term storage (LTS) on the first of these trials and scored as LTS on all following trials. The total of the words in LTS of all six trials is then summed. If a word in LTS is consistently recalled on all subsequent trials, it is then scored as Consistent Long Term Retrieval (CLTR). The total of the words in CLTR of all six trials is summed. The total score ranged from 0 to 72. Higher values represent a better outcome. (NCT01333501)
Timeframe: Screening (-1month), 18 month

InterventionRaw score (Least Squares Mean)
Fingolimod5.93
Interferon Beta 1b3.96

Change From Screening in Selective Reminding Test - Delayed Recall (SRT-D) Raw Score

The tests SRT (Selective Reminding Test) for episodic memory (verbal learning and delayed recall). The Delayed SRT test is the total number of words recalled after a delayed period. The total score ranged from 0 to 12. Higher values represent a better outcome. (NCT01333501)
Timeframe: Screening (-1month), 18 month

InterventionRaw score (Least Squares Mean)
Fingolimod0.57
Interferon Beta 1b0.39

Change From Screening in Selective Reminding Test - Long-Term Storage (SRT-LTS) Raw Score

Brief Repeatable Battery (BRB)- widely used as a clinical and research tool, with 68% sensitivity and 85% specificity. It consists of the serial administration of 5 tests. One of the tests is SRT (Selective Reminding Test) for episodic memory (verbal learning and delayed recall). A word recalled on two consecutive trials is considered to have entered long-term storage (LTS) on the first of these trials and scored as LTS on all following trials. The total of the words in LTS of all six trials is then summed. The total score ranged from 0 to 72. Higher values represent a better outcome. (NCT01333501)
Timeframe: Screening (-1month), 18 month

Interventionraw score (Least Squares Mean)
Fingolimod6.32
Interferon Beta 1b4.46

Change From Screening in Spatial Recall Test - Delayed Recall (SPART-D)

Spatial Recall Test (SPART) for visuospatial learning and delayed recall.Spatial Recall Test (10/36): The spatial recall test assesses visuospatial learning and delayed recall (10/36-D). A checkerboard with ten checkers arranged in a pattern was shown to the subject for ten seconds. The subject was then asked to reproduce the same pattern with ten checkers on an empty checkerboard. The test includes three consecutive trials. The score was the total number of correct responses for the tree trials. The total score ranged from 0 to 10. Higher values represent a better outcome. (NCT01333501)
Timeframe: Screening (-1month), 18 month

InterventionRaw score (Least Squares Mean)
Fingolimod0.41
Interferon Beta 1b0.44

Change From Screening in Spatial Recall Test (SPART) Raw Score

Spatial Recall Test (SPART) for visuospatial learning and delayed recall.Spatial Recall Test (10/36): The spatial recall test assesses visuospatial learning and delayed recall (10/36-D). A checkerboard with ten checkers arranged in a pattern is shown to the subject for ten seconds. The subject is then asked to reproduce the same pattern with ten checkers on an empty checkerboard. The test includes three consecutive trials. The score is the total number of correct responses for the tree trials. The total score ranged from 0 to 30. Higher values represent a better outcome. (NCT01333501)
Timeframe: Screening (-1month), 18 month

InterventionRaw score (Least Squares Mean)
Fingolimod1.46
Interferon Beta 1b3.06

Change From Screening in Symbol Digit Modalities Test (SDMT) Raw Score

Symbol Digit Modality Test (SDMT) for sustained attention and information processing speed. It presents a series of nine symbols, each of which is paired with a single digit labeled 1-9 in a key at the top of the sheet. The reminder of the page has a pseudo-randomized sequence of symbols, and the patient must respond with the digit associated with each of these as quickly as possible. The score is the number of correct answers in 90 seconds. The total score ranged from 0 to 110. Higher values represent a better outcome. (NCT01333501)
Timeframe: Screening (-1month), 18 month

InterventionRaw score (Least Squares Mean)
Fingolimod2.19
Interferon Beta 1b3.93

Change From Screening in the Number of New T2 Lesions

New T2 lesions at a specific visit were assessed relative to the previous visit scan. The total number of lesions (visit 8 to 18 month) is calculated as the sum of the number of lesions. (NCT01333501)
Timeframe: Screening (-1month), 18 month

InterventionNumber of new lesions (Mean)
Fingolimod1.25
Interferon Beta 1b3.33

Change From Screening in the Number of T1 Gd+ Enhancing Lesions

Total number of post-baseline Gd-enhanced lesions is calculated as a sum of all Gd-enhanced lesions seen on post-baseline scans per visit. Real (not per slice) lesions are counted in this analysis (NCT01333501)
Timeframe: Screening (-1month), 18 month

InterventionNumber of new lesions (Mean)
Fingolimod-0.64
Interferon Beta 1b0.59

Change From Screening in the Percentage of Brain Volume Change

Calculations of brain volume change were performed using the structural image evaluation of normalized atrophy (SIENA), software included in the Functional Magnetic Resonance Imaging of the Brain (FMRIB) software library. SIENA is a fully automated method for estimating temporal brain volume change. (NCT01333501)
Timeframe: Screening (-1month), 18 month

Interventionpercentage of brain volume (Mean)
Fingolimod-0.60
Interferon Beta 1b-0.96

Change From Screening in the Volume of Total T1 Hypointense Lesions

Volume of hypointense post-gadolinium T1 lesion component was measured by MRI scan. Means were estimated using a Mixed-effect model with repeated measures (MMRM) by-visit interaction. (NCT01333501)
Timeframe: Screening (-1month), 18 month

Interventionmm^3 (Mean)
Fingolimod391.96
Interferon Beta 1b213.93

Change From Screening in the Volume of Total T2 Lesions

Change in volume of total T2-weighted lesions by visit were summarized. Negative values indicate improvement (reduction in lesion volume) and positive values worsening (increase in lesion volume (NCT01333501)
Timeframe: Screening (-1 month), 18 months

Interventionmm^3 (Mean)
Fingolimod176.25
Interferon Beta 1b711.81

Change From Screening in Word List Generation (WLG)

Word List Generation (COWAT/WLG): The COWAT assesses verbal fluency on semantic stimulus by asking the patient to produce as many words as possible belonging to a semantic category. The test assessed the verbal fluency, recorded all the possible correct word that a patients should give in 90 sec. No maximum range is available. Higher values represent a better outcome. The score was the number of correct words. The more words the patient pronounces, the better it is. We can imagine that the minimum value might be zero words, , but it is not a score scale. (NCT01333501)
Timeframe: Screening (-1month), 18 month

InterventionRaw score (Least Squares Mean)
Fingolimod0.39
Interferon Beta 1b0.24

Changes From Baseline in Fatigue Impact Scale (mFIS, Total Score and Scores of the 3 Individual Domains).

Modified Fatigue Impact Scale (mFIS) questionnaire is described at each time point to evaluate fatigue by means of usual descriptive statistics. Three domains were also defined: Physical Subscale (sum of items 4, 6, 7, 10, 13, 14, 17, 20, 21 and therefore ranging from 0 to 36), Cognitive Subscale (sum of items 1, 2, 3, 5, 11, 12, 15, 16, 18, 19 and therefore ranging from 0 to 40) and Psychosocial Subscale (sum of items 8, 9 and therefore ranging from 0 to 8). Finally, mFIS - overall score ranged from 0 to 80. The mFIS total score was computed as the sum of scores for each item. Lower values represent a better outcome. (NCT01333501)
Timeframe: Baseline, 18 months

InterventionScore (Least Squares Mean)
Fingolimod2.36
Interferon Beta 1b6.67

Changes in the Environmental Status Scale Score (ESS)

The Environmental Status Scale (ESS) is used to quickly evaluate a patient for handicap. It was derived from a measure of socio-economic status. It consists of seven parameters: (1) actual work status, (2) financial and economic status, (3) personal residence or home, (4) personal assistance required, (5) transportation, (6) community services, (7) social activity. Each parameter has a single score from minimum 0 to maximum 5. ESS score is the sum of the points for all 7 parameters: minimum score: 0; maximum score: 35. The higher the score the greater the handicap (NCT01333501)
Timeframe: Baseline, 18 month

InterventionScore (Mean)
Fingolimod1.08
Interferon Beta 1b0.91

Changes in Quality of Life, by Means of the Multiple Sclerosis Quality of Life (MSQoL-54)

A 54 question measure covers 12 domains; assesses mental and physical health. The physical health composite score is a weighted average of the physical health scales, such as physical function, health perceptions, and energy. The mental health composite score is a weighted average of the mental health scales, such as overall quality of life, cognitive function, and health distress. Each domain has a range from 0 to 100 where higher means better. (NCT01333501)
Timeframe: Baseline, 18 months

,
InterventionScore (Least Squares Mean)
Mental health composite scorePhysical health composite score
Fingolimod4.76-1.80
Interferon Beta 1b-2.31-4.75

Change From Baseline in SDMT at Week 52

The SDMT measures the time to pair abstract symbols with specific numbers. The test requires elements of attention, visuoperceptual processing, working memory, and psychomotor speed. The score is the number of correctly coded items from 0-110 in 90 seconds. The total score provides a measure of the speed and accuracy of symbol-digit substitution. (NCT02342704)
Timeframe: Baseline, Week 52

Interventionunits on a scale (Mean)
Fingolimod2.11

Change From Baseline in Symbol Digit Modalities Test (SDMT) at Week 24

The SDMT measures the time to pair abstract symbols with specific numbers. The test requires elements of attention, visuoperceptual processing, working memory, and psychomotor speed. The score is the number of correctly coded items from 0-110 in 90 seconds. The total score provides a measure of the speed and accuracy of symbol-digit substitution. (NCT02342704)
Timeframe: Baseline, Week 24

Interventionunits on a scale (Mean)
Natalizumab3.79
Fingolimod3.24

Cumulative Number of New or Enlarging T2 Lesions

(NCT02342704)
Timeframe: Baseline, Week 24

Interventionlesions (Mean)
Natalizumab1.33
Fingolimod1.94

Change From Baseline in Total T1-Hypointense and Total T2-Hyperintense Lesion Volumes at Week 24

As assessed by magnetic resonance imaging (MRI). (NCT02342704)
Timeframe: Baseline, Week 24

,
Interventionpercentage change (Mean)
T1 Lesion Volume ChangeT2 Lesion Volume Change
Fingolimod1.813.32
Natalizumab0.50.08

Change From Baseline in Total T1-Hypointense and Total T2-Hyperintense Lesion Volumes at Week 52

As assessed by MRI. (NCT02342704)
Timeframe: Baseline, Week 52

Interventionpercentage change (Mean)
T1 Lesion Volume ChangeT2 Lesion Volume Change
Fingolimod-15.315.6

Cumulative Number of New T1-Gd+ Lesions

(NCT02342704)
Timeframe: Baseline, Week 4, Week 12, Week 24

,
Interventionlesions (Mean)
From Baseline to Week 4From Baseline to Week 12From Baseline to Week 24
Fingolimod1.692.272.6
Natalizumab0.620.680.72

Estimated Annualized Aggregate Relapse Rate (ARR) in the Core Phase of the Study

The ARR is defined as the number of confirmed relapses in a year. A relapse is defined as the appearance of a new or worsening of a previously stable or improving pre existing neurological abnormality, separated by at least 30 days from onset of a preceding relapse. The abnormality must be present for at least 24 hours and occur in the absence of fever or infection. The annualized ARR for each treatment group was calculated using negative binomial regression adjusted by treatment, country, number of relapses in the previous 2 years, and the baseline Expanded Disability Status Scale score. (NCT00340834)
Timeframe: Baseline to Month 12

InterventionEstimate relapses per year (Number)
Fingolimod 1.25 mg0.203
Fingolimod 0.5 mg0.161
Interferon β-1a µg/Fingolimod 1.25 or 0.5 mg0.331

Number of New or Newly Enlarged T2 Lesions in Comparison With Baseline in the Core Phase of the Study

The number of new or newly enlarged T2 lesions in comparison to baseline was assessed with T2-weighted magnetic resonance image (MRI) scans. A T2-weighted MRI scan utilizes particular values of the echo time (TE) and the repetition time (TR) parameters of image acquisition. Inflammation and tissue damage are seen as bright areas in T2 images and are often referred to as T2 lesions. T2-weighted MRI scans are a sensitive way to evaluate the brain for demyelinating diseases, such as multiple sclerosis. (NCT00340834)
Timeframe: Baseline to Month 12

InterventionT2 lesions (Mean)
Fingolimod 1.25 mg1.6
Fingolimod 0.5 mg1.6
Interferon β-1a µg/Fingolimod 1.25 or 0.5 mg2.6

Percentage of Participants Free of 3-month Disability Progression Assessed With the Expanded Disability Status Scale (EDSS) at the End of the Core Phase of the Study

The EDSS is a scale for assessing disability in 8 functional systems (visual, brain stem, pyramidal, cerebellar, sensory, bowel & bladder, cerebral, other functions). An overall score ranging from 0 (normal) to 10 (death due to MS) is calculated. Disability progression was determined by the EDSS score based on the following criteria: One point increase from baseline in patients with baseline EDSS score from 0 to 5.0; or half a point increase in patients with baseline EDSS score of 5.5 or above. Percent of patients free of disability progression was calculated using the Kaplan-Meier method. (NCT00340834)
Timeframe: Baseline to Month 12

InterventionPercentage of participants (Number)
Fingolimod 1.25 mg93.3
Fingolimod 0.5 mg94.1
Interferon β-1a µg/Fingolimod 1.25 or 0.5 mg92.1

Estimated Annualized Aggregate Relapse Rate (ARR) in the Core and Extension Phases of the Study

The ARR is defined as the number of confirmed relapses in a year. A relapse is defined as the appearance of a new or worsening of a previously stable or improving pre existing neurological abnormality, separated by at least 30 days from onset of a preceding relapse. The abnormality must be present for at least 24 hours and occur in the absence of fever or infection. The annualized ARR for each treatment group was calculated using negative binomial regression adjusted by treatment, country, number of relapses in the previous 2 years, and the baseline Expanded Disability Status Scale score. (NCT00340834)
Timeframe: Month 0 to end of study (up to approximately 4.5 years)

,,
InterventionEstimated relapses per year (Number)
Month 12 to Month 24, n=330, 356, 341Month 24 to Month 36, n=287, 321, 293Month 36 to Month 48, n=267, 303, 271Month 48 to end of study, n=36, 38, 29Month 0 to end of study, n=420, 429, 431
Fingolimod 0.5 mg0.1820.110NANA0.166
Fingolimod 1.25 mg0.1560.116NANA0.192
Interferon β-1a µg/Fingolimod 1.25 or 0.5 mg0.2660.121NANA0.271

Number of New or Newly Enlarged T2 Lesions in the Extension Phase of the Study

The number of new or newly enlarged T2 lesions in comparison to baseline was assessed with T2-weighted magnetic resonance image (MRI) scans. A T2-weighted MRI scan utilizes particular values of the echo time (TE) and the repetition time (TR) parameters of image acquisition. Inflammation and tissue damage are seen as bright areas in T2 images and are often referred to as T2 lesions. T2-weighted MRI scans are a sensitive way to evaluate the brain for demyelinating diseases, such as multiple sclerosis. (NCT00340834)
Timeframe: Month 12 to end of study (up to approximately 3.5 years)

,,
InterventionT2 lesions (Mean)
Month 12 to Month 24Month 24 to Month 36, n=255, 289, 258Month 36 to Month 48, n=36, 34, 35Last MRI scan to end of study, n=275, 309, 290
Fingolimod 0.5 mg0.871.040.590.86
Fingolimod 1.25 mg1.081.400.971.75
Interferon β-1a µg/Fingolimod 1.25 or 0.5 mg0.970.720.491.03

Percentage of Participants Free of 3-month and 6-month Disability Progression Assessed With the Expanded Disability Status Scale (EDSS) at the End of the Extension Phase of the Study

The EDSS is a scale for assessing disability in 8 functional systems (visual, brain stem, pyramidal, cerebellar, sensory, bowel & bladder, cerebral, other functions). An overall score ranging from 0 (normal) to 10 (death due to MS) is calculated. Disability progression was determined by the EDSS score based on the following criteria: One point increase from baseline in patients with baseline EDSS score from 0 to 5.0; or half a point increase in patients with baseline EDSS score of 5.5 or above. Percent of patients free of disability progression was calculated using the Kaplan-Meier method. (NCT00340834)
Timeframe: Baseline to end of study (up to approximately 4.5 years)

,,
InterventionPercentage of participants (Number)
Free of 3-month progressionFree of 6-month progression
Fingolimod 0.5 mg71.2879.76
Fingolimod 1.25 mg67.0179.54
Interferon β-1a µg/Fingolimod 1.25 or 0.5 mg73.4081.61

Estimated Annualized Aggregate Relapse Rate (ARR)

The ARR is defined as the number of confirmed relapses in a year. A relapse is defined as the appearance of a new or worsening of a previously stable or improving pre-existing neurological abnormality, separated by at least 30 days from onset of a preceding relapse. The abnormality must be present for at least 24 hours and occur in the absence of fever or infection. The annualized ARR for each treatment group was the mean of the annualized ARRs for all patients in the group calculated as the total number of confirmed relapses divided by the total number of days on study, multiplied by 365.25. (NCT00289978)
Timeframe: Baseline to end of study (Month 24)

InterventionRelapses per year (Number)
Fingolimod 1.25 mg0.16
Fingolimod 0.5 mg0.18
Placebo0.40

Number of New or Newly Enlarged T2 Lesions at Month 24 in Comparison With Baseline

The number of new or newly enlarged T2 lesions at Month 24 in comparison to baseline was assessed with T2-weighted magnetic resonance image (MRI) scans. A T2-weighted MRI scan utilizes particular values of the echo time (TE) and the repetition time (TR) parameters of image acquisition. Inflammation and tissue damage are seen as bright areas in T2 images and are often referred to as T2 lesions. T2-weighted MRI scans are a sensitive way to evaluate the brain for demyelinating diseases, such as multiple sclerosis. (NCT00289978)
Timeframe: Baseline to end of study (Month 24)

InterventionT2 lesions (Mean)
Fingolimod 1.25 mg2.5
Fingolimod 0.5 mg2.5
Placebo9.8

Percentage of Patients Free of Disability Progression at Month 24 Assessed With the Expanded Disability Status Scale (EDSS)

EDSS assesses disability in 8 functional systems. An overall score ranging from 0 (normal) to 10 (death due to MS) is calculated. Disability progression was determined by the following: One point increase from baseline in patients with baseline EDSS score from 0 to 5.0; or half a point increase in patients with baseline EDSS score of 5.5 or above. A 3-month confirmed disability progression required onset EDSS, 3-month confirming EDSS, and all EDSS in between to meet the disability progression criteria. Percent of free of disability progression was calculated using the Kaplan Meier method. (NCT00289978)
Timeframe: Baseline to end of study (Month 24)

InterventionPercentage of participants (Number)
Fingolimod 1.25 mg83.4
Fingolimod 0.5 mg82.3
Placebo75.9

Percentage of Patients Relapse-free at the End of the Study

Patients who did not experience any relapses confirmed by a neurologist during the study were regarded as relapse-free patients. (NCT00670449)
Timeframe: Baseline to the end of the study (up to 4 years)

InterventionPercentage of patients (Number)
Fingolimod 0.5 mg45.2
Fingolimod 1.25 mg62.1
Placebo-fingolimod48.3

Aggregate Annualized Relapse Rate (ARR) Based on Confirmed Relapses

The ARR was defined as the total number of relapses for all patients in the treatment arm / total number of days in the study for all patients in the treatment arm for the specific period of time × 365.25. General definition of relapse: Appearance of a new neurological abnormality or worsening of previously stable or improving pre-existing neurological abnormality, separated by at least 30 days from the onset of a preceding clinical demyelinating event. The abnormality must be present for at least 24 hours and occur in the absence of fever (< 37.5°C) or infection. A relapse was to be confirmed by a neurologist trained on the Expanded Disability Status Scale (EDSS). A relapse must be accompanied by an increase of at least half a step (0.5) on the EDSS or an increase of 1 point on 2 different Functional Systems (FS) of the EDSS or 2 points on 1 of the FS (excluding Bowel/Bladder or Cerebral FS). (NCT00670449)
Timeframe: Months 0-6, 6-12, 12-24, 24-36, 36-48, and 48 to the end of the study (up to 4 years)

,,
InterventionRelapses per year (Number)
Month 0 to 6 (N=57, 54, 57)Month 6 to 12 (N=47, 46, 50)Month 12 to 24 (N=44, 43, 39)Month 24 to 36 (N=41, 40, 34)Month 36 to 48 (N=28, 23, 23)Month 48 to end of study (N=8, 6, 3)
Fingolimod 0.5 mg0.5390.2270.1660.1910.1640.000
Fingolimod 1.25 mg0.4040.2840.2230.0580.0750.000
Placebo-fingolimod1.1310.2320.2220.1620.3090.000

Change From Core Study Baseline in the Expanded Disability Status Scale (EDSS) Score

Disability progression was measured by the EDSS score. A trained neurologist grades the multiple sclerosis (MS) disability of the patient on a scale of 0-5 (no to severe disability) in 8 Functional Systems (FS): Pyramidal, cerebellar, brainstem, sensory, bowel and bladder, visual, cerebral (or mental), and other. The EDSS score ranges from 0-10 (normal to dead) with higher scores indicating greater disability. A 3-month confirmed disability progression was defined as a 3-month sustained increase from baseline in the EDSS score, that is, every EDSS score obtained (scheduled or unscheduled) within 3-months after the first progression met the following progression criteria: One point (1) increase from baseline in patients with baseline EDSS score from 0 to 5.0; or half a point (0.5) increase in patients with baseline EDSS score of 5.5 or above. A 6-month confirmed disability progression was defined exactly the same except that the sustained progression had to last 6 months. (NCT00670449)
Timeframe: Baseline to Months 12, 24, 36, 48, and end of study (up to 4 years)

,,
InterventionUnits on a scale (Mean)
Month 12 (N=45, 43, 40)Month 24 (N=42, 40, 35)Month 36 (N=25, 21, 23)Month 48 (N=3, 4, 1)End of study (N=37, 36, 32)
Fingolimod 0.5 mg-0.02-0.120.161.170.00
Fingolimod 1.25 mg-0.02-0.06-0.07-0.63-0.17
Placebo-fingolimod-0.23-0.21-0.370.00-0.31

Percentage of Patients Free From 3-month and 6-month Confirmed Disability Progression at Their Last Expanded Disability Status Scale (EDSS) Assessment

Disability progression was measured by the EDSS score. A trained neurologist grades the multiple sclerosis (MS) disability of the patient on a scale of 0-5 (no to severe disability) in 8 Functional Systems (FS): Pyramidal, cerebellar, brainstem, sensory, bowel and bladder, visual, cerebral (or mental), and other. The EDSS score ranges from 0-10 (normal to dead) with higher scores indicating greater disability. A 3-month confirmed disability progression was defined as a 3-month sustained increase from baseline in the EDSS score, that is, every EDSS score obtained (scheduled or unscheduled) within 3-months after the first progression met the following progression criteria: One point (1) increase from baseline in patients with baseline EDSS score from 0 to 5.0; or half a point (0.5) increase in patients with baseline EDSS score of 5.5 or above. A 6-month confirmed disability progression was defined exactly the same except that the sustained progression had to last 6 months. (NCT00670449)
Timeframe: Baseline to the end of the study (up to 4 years)

,,
InterventionPercentage of patients (Number)
Free from 3-month confirmed disability progressionFree from 6-month confirmed disability progression
Fingolimod 0.5 mg74.387.1
Fingolimod 1.25 mg82.490.7
Placebo-fingolimod90.692.3

Percentage of Patients Free of Gd-enhanced T1 Weighted Magnetic Resonance Imaging (MRI) Lesions

To ensure consistency, MRI scans were evaluated centrally at the Institute of Neurotherapeutics in Kyoto, Japan. After checking the scans for completeness and quality, all scans were analyzed by blinded readers (experienced neurologists). The number of Gd-enhanced T1 weighted MRI lesions were counted and recorded. Lesions expanding through several slices were counted as only 1 lesion. (NCT00670449)
Timeframe: Months 6, 9, 12, 18, 24, 36, and 48

,,
InterventionPercentage of patients (Number)
Month 6 (N=44, 48, 50)Month 9 (N=44, 39, 41)Month 12 (N=44, 42, 36)Month 18 (N=42, 40, 37)Month 24 (N=38, 39, 33)Month 36 (N=25, 21, 22)Month 48 (N=3, 4, 1)
Fingolimod 0.5 mg88.690.997.792.994.792.0100.0
Fingolimod 1.25 mg97.994.997.692.594.985.775.0
Placebo-fingolimod58.092.788.994.690.990.9100.0

Percentage of Patients Free of New or Newly Enlarged T2 Weighted MRI Lesions

To ensure consistency, MRI scans were evaluated centrally at the Institute of Neurotherapeutics in Kyoto, Japan. After checking the scans for completeness and quality, all scans were analyzed by blinded readers (experienced neurologists). The number of new or newly enlarged T2 weighted MRI lesions were counted and recorded. New lesions were identified by comparing each lesion with previous scans. Lesions expanding through several slices were counted as only 1 lesion. (NCT00670449)
Timeframe: Months 0-3, 3-6, 6-9, 9-12, 12-18, 18-24, 24-36,36-48, and 48 to the end of the study (up to 4 years)

,,
InterventionPercentage of patients (Number)
Month 0 to 3 (N=49, 50, 51)Month 3 to 6 (N=43, 47, 48)Month 6 to 9 (N=44, 39, 40)Month 9 to 12 (N=44, 42, 35)Month 12 to 18 (N=43, 40, 37)Month 18 to 24 (N=39, 39, 33)Month 24 to 36 (N=25, 21, 22)Month 36 to 48 (N=3, 4, 1)Month 48 to end of study (N=3, 3, 1)
Fingolimod 0.5 mg69.486.088.693.288.494.992.0100.0100.0
Fingolimod 1.25 mg60.091.592.395.290.092.390.5100.0100.0
Placebo-fingolimod45.143.870.085.791.987.990.9100.0100.0

Annualized Aggregate Relapse Rate (ARR) During Months 0 to End of Study(Core [CFTY720D2301/NCT00289978] and Extension Study)

ARR is defined as the number of confirmed relapses in a year. A relapse is defined as the appearance of a new or worsening of a previously stable or improving pre-existing neurological abnormality, separated by at least 30 days from onset of a preceding relapse. The abnormality must be present for at least 24 hours and occur in the absence of fever or infection. The annualized ARR for each treatment group was the mean of the annualized ARRs for all patients in the group, calculated as the total number of confirmed relapses divided by the total number of days on study multiplied by 365.25. (NCT00662649)
Timeframe: Months 0 to end of study (maximum up to 60 months)

InterventionRelapses per year (Number)
Fingolimod 1.25 mg0.164
Fingolimod 0.5 mg0.185
Placebo-fingolimod0.357

Change (Expressed as Ratio) in the Annualized Aggregate Relapse Rate (ARR) From Months 0-24 (Core Study) to Months 24-48 (Extension Study)

ARR is defined as the number of confirmed relapses in a year. A relapse is defined as the appearance of a new or worsening of a previously stable or improving pre-existing neurological abnormality, separated by at least 30 days from onset of a preceding relapse. The abnormality must be present for at least 24 hours and occur in the absence of fever or infection. The annualized ARR for each treatment group was the mean of the annualized ARRs for all patients in the group, calculated as the total number of confirmed relapses divided by the total number of days on study multiplied by 365.25. (NCT00662649)
Timeframe: Months 0-24 (core study) and Months 24-48 (extension study)

InterventionRatio of relapses per year (Number)
Fingolimod 1.25 mg1.164
Fingolimod 0.5 mg0.850
Placebo-fingolimod 1.25 mg0.547
Placebo-fingolimod 0.5 mg0.446

Percent Change in Brain Volume From Month 0 End of Study (Core and Extension Study)

Calculations of brain volume change were performed using the structural image evaluation of normalized atrophy (SIENA), software included in the Functional Magnetic Resonance Imaging of the Brain (FMRIB) software library. SIENA is a fully automated method for estimating temporal brain volume change. (NCT00662649)
Timeframe: Months 0 to end of study (maximum up to 60 months)

InterventionPercent change (Mean)
Fingolimod 1.25 mg-1.639
Fingolimod 0.5 mg-1.674
Placebo-fingolimod-2.241

Time to First 3-month Confirmed Disability Progression up to End of Study Based on Expanded Disability Status Scale (EDSS): Kaplan-Meier Estimate of Percentage of Patients Free of Disability Progression

Kurtzke's Expanded Disability Status Scale (EDSS) is a scale for assessing neurologic impairment in multiple sclerosis (MS) includes a series of scores in each of eight functional systems such as Visual, Brain Stem, Pyramidal, Cerebellar, Sensory, Bowel & Bladder, Cerebral, and Other. The EDSS steps range from 0 (normal) to 10 (death due to MS). The Kaplan-Meier estimates of the percentage of participants free of disability progression at end of study and their 95% CIs were provided for each treatment group. (NCT00662649)
Timeframe: Core baseline to end of study (maximum up to 60 months)

InterventionPercentage of patients (Number)
Fingolimod 1.25 mg74.15
Fingolimod 0.5 mg73.90
Placebo-fingolimod66.28

Time to First Confirmed Relapse up to End of Study: Kaplan-Meier Estimate of Percentage of Patients Relapse-free

A relapse was confirmed when it was accompanied by an increase of at least half a step (0.5) on the Expanded Disability Status Scale (EDSS) or an increase of 1 point on two different Functional Systems (FS) of the EDSS or 2 points on one of the FS (excluding Bowel/Bladder or Cerebral FS). Kaplan-Meier estimates of the percentage of relapse-free patients at end of study and and 95% confidence intervals (CIs) were presented for the treatment groups. (NCT00662649)
Timeframe: Core baseline to end of study (maximum up to 60 months)

InterventionPercentage of patients (Number)
Fingolimod 1.25 mg59.85
Fingolimod 0.5 mg59.29
Placebo-fingolimod37.18

Annualized Aggregate Relapse Rate (ARR) During Months 0-24 (Core Study) and Months 24-48 (Extension Study)

ARR is defined as the number of confirmed relapses in a year. A relapse is defined as the appearance of a new or worsening of a previously stable or improving pre-existing neurological abnormality, separated by at least 30 days from onset of a preceding relapse. The abnormality must be present for at least 24 hours and occur in the absence of fever or infection. The annualized ARR for each treatment group was the mean of the annualized ARRs for all patients in the group, calculated as the total number of confirmed relapses divided by the total number of days on study multiplied by 365.25. (NCT00662649)
Timeframe: Months 0-24 (core study) and Months 24-48 (extension study)

,,,
InterventionRelapses per year (Number)
Months 0-24Months 24-48
Fingolimod 0.5 mg0.1110.095
Fingolimod 1.25 mg0.0640.074
Placebo-fingolimod 0.5 mg0.2920.130
Placebo-fingolimod 1.25 mg0.3010.164

Change in Mean Number of New or Newly Enlarged T2 Magnetic Resonance Imaging (MRI) Lesions During Months 0-24 (Core Study) and Months 24-48 (Extension Study)

The number of new or newly enlarged T2 lesions was assessed with T2-weighted MRI scans. A T2-weighted MRI scan utilizes particular values of the echo time (TE) and the repetition time (TR) parameters of image acquisition. Inflammation and tissue damage are seen as bright areas in T2 images and are often referred to as T2 lesions. T2 weighted MRI scans are a sensitive way to evaluate the brain for demyelinating diseases, such as multiple sclerosis. (NCT00662649)
Timeframe: Months 0-24 (core study) and Months 24-48 (extension study)

,,,
InterventionLesions (Mean)
Months 0-24Months 24-48Change from Months 0-24 to Months 24-48
Fingolimod 0.5 mg2.662.58-0.09
Fingolimod 1.25 mg2.141.71-0.43
Placebo-fingolimod 0.5 mg8.121.43-6.68
Placebo-fingolimod 1.25 mg12.832.34-10.48

Percent Change in Brain Volume From Month 0 to Month 24 (Core Study) and From Month 24 to Month 48 (Extension Study)

Calculations of brain volume change were performed using the structural image evaluation of normalized atrophy (SIENA), software included in the Functional Magnetic Resonance Imaging of the Brain (FMRIB) software library. SIENA is a fully automated method for estimating temporal brain volume change. (NCT00662649)
Timeframe: Months 0-24 (core study) and Months 24-48 (extension study)

,,,
InterventionPercent change (Mean)
Month 0 to Month 24, n=75, 109, 41, 49Month 24 to Month 48, n=75, 109, 41, 49
Fingolimod 0.5 mg-0.983-0.780
Fingolimod 1.25 mg-1.011-0.871
Placebo-fingolimod 0.5 mg-1.419-0.903
Placebo-fingolimod 1.25 mg-1.511-1.103

Percentage of Patients Free of New or Newly Enlarged T2 Magnetic Resonance Imaging (MRI) Lesions During Months 0-24 (Core Study) and Months 24-48 (Extension Study)

The number of new or newly enlarged T2 lesions was assessed with T2-weighted MRI scans. A T2-weighted MRI scan utilizes particular values of the echo time (TE) and the repetition time (TR) parameters of image acquisition. Inflammation and tissue damage are seen as bright areas in T2 images and are often referred to as T2 lesions. T2 weighted MRI scans are a sensitive way to evaluate the brain for demyelinating diseases, such as multiple sclerosis. (NCT00662649)
Timeframe: Months 0-24 (core study) and Months 24-48 (extension study)

,,,
InterventionPercentage of patients (Number)
Months 0-24Months 24-48
Fingolimod 0.5 mg50.369.3
Fingolimod 1.25 mg53.257.1
Placebo-fingolimod 0.5 mg23.255.1
Placebo-fingolimod 1.25 mg18.552.3

Annualized Relapse Rate (ARR) at 6 Months

Annualized relapse rate (ARR) of the treatment group is calculated by taking the total number of confirmed relapses for all the patients in the treatment group divided by the total number of days on study for all patients in the group and multiplied by 365.25 to obtain the annual rate. (NCT00537082)
Timeframe: 6 Months

InterventionRelapses per year (Number)
FTY720 1.25 mg0.407
FTY720 0.5 mg0.512
Placebo1.131

Number of Patients Free of Gadolinium-enhanced T1-Weighted Magnetic Resonance Imaging (MRI) Lesions at Both Month 3 and Month 6

Brain Magnetic Resonance Imaging (MRI) was performed at Month 3 and Month 6. Gadolinium-enhancing lesions (active lesions) represent acute inflammatory activity only and dissipate within 2-8 weeks of appearance. MRI scans were analyzed by blinded readers at the central MRI Evaluation Center to ensure consistency. Any Gd-enhanced T1 weighted MRI data obtained less than 14 days after the steroid used to treat MS relapses is invalid and excluded. (NCT00537082)
Timeframe: Month 3 and Month 6

InterventionParticipants (Number)
FTY720 1.25 mg43
FTY720 0.5 mg35
Placebo21

Number of Patients Free of MS Relapse up to Month 6 (Confirmed Relapse Only)

A relapse must have been confirmed by a neurologist and was confirmed when it was accompanied by an increase of at least half a step (0.5) on the Expanded Disability Status Scale (EDSS) or an increase of 1 point on two different Functional Systems (FS) of the EDSS or 2 points on one of the FS (excluding Bowel/Bladder or Cerebral FS). (NCT00537082)
Timeframe: up to Month 6

InterventionParticipants (Number)
FTY720 1.25 mg45
FTY720 0.5 mg45
Placebo37

Number of Patients Free of New or Newly Enlarged T2 Lesions

The number of T2 lesions were obtained from MRI scans at Screening visit. The numbers of new/newly enlarging T2 lesions were obtained from MRI scans at Month 3 or more. New lesions were identified by comparing each lesion already seen in previous examinations. Lesions expanding throughout several slices were counted as only one lesion. (NCT00537082)
Timeframe: up to Month 3 and up to Month 6

,,
InterventionParticipants (Number)
Month 3 (n = 50, 49, 51)Month 6 (n = 48, 48, 50)
FTY720 0.5 mg3431
FTY720 1.25 mg3028
Placebo2318

Aggregate Annualized Relapse Rate (ARR) Estimate up to End of Study

"ARR is the average number of relapses in a year calculated by negative binomial regression as the sum of confirmed relapses of all patients in the group divided by the sum of the number of days on study of all patients in the group and multiplied by 365.25.~A relapse was defined as the appearance of a new neurological abnormality or worsening of previously stable or improving pre-existing neurological abnormality, separated by at least 30 days from onset of a preceding clinical demyelinating event. The abnormality must be present for at least 24 hours and occur in the absence of fever (<37.5C) or known infection. A relapse must be confirmed by the Independent Evaluating Physician (examining neurologist).~ARR estimates were calculated from a negative binomial regression model adjusted for treatment, pooled center, number of relapses in the previous 2 years prior to enrollment, and Baseline expanded disability status scale (EDSS)." (NCT00355134)
Timeframe: From Baseline until end of study (up to approximately 54 months).

Interventionrelapses per year (Number)
Fingolimod 1.25 mg0.180
Fingolimod 0.5 mg0.192
Placebo0.363

Aggregate Annualized Relapse Rate (ARR) Estimate up to Month 24

"ARR is the average number of relapses in a year calculated by negative binomial regression as the sum of confirmed relapses of all patients in the group divided by the sum of the number of days on study of all patients in the group and multiplied by 365.25.~A relapse was defined as the appearance of a new neurological abnormality or worsening of previously stable or improving pre-existing neurological abnormality, separated by at least 30 days from onset of a preceding clinical demyelinating event. The abnormality must be present for at least 24 hours and occur in the absence of fever (<37.5C) or known infection. A relapse must be confirmed by the Independent Evaluating Physician (examining neurologist).~ARR estimates were calculated from a negative binomial regression model adjusted for treatment, pooled center, number of relapses in the previous 2 years prior to enrollment, and Baseline expanded disability status scale (EDSS)." (NCT00355134)
Timeframe: 24 months

Interventionrelapses per year (Number)
Fingolimod 1.25 mg0.203
Fingolimod 0.5 mg0.208
Placebo0.403

Percentage of Participants Relapse-free up to End of Study

Estimates of the percentage of participants relapse-free at end of study were generated from Kaplan-Meier curves of the time to first relapse. A relapse was defined as the appearance of a new neurological abnormality or worsening of previously stable or improving pre-existing neurological abnormality, separated by at least 30 days from onset of a preceding clinical demyelinating event. The abnormality must be present for at least 24 hours and occur in the absence of fever (<37.5C) or infection. A relapse was confirmed by an Independent Evaluating Physician. (NCT00355134)
Timeframe: From Baseline until the end of study (up to approximately 54 months)

Interventionpercentage of participants (Number)
Fingolimod 1.25 mg63.88
Fingolimod 0.5 mg66.57
Placebo49.12

Percentage of Participants Relapse-free up to Month 24

Estimates of the percentage of participants relapse-free at 24 months were generated from Kaplan-Meier curves of the time to first relapse. A relapse was defined as the appearance of a new neurological abnormality or worsening of previously stable or improving pre-existing neurological abnormality, separated by at least 30 days from onset of a preceding clinical demyelinating event. The abnormality must be present for at least 24 hours and occur in the absence of fever (<37.5C) or infection. A relapse was confirmed by an Independent Evaluating Physician. (NCT00355134)
Timeframe: 24 months

Interventionpercentage of participants (Number)
Fingolimod 1.25 mg73.2
Fingolimod 0.5 mg71.5
Placebo52.7

Change From Baseline in Lesion Volume at Month 24 (Core Phase)

Change from Baseline in lesion volume was measured by MRI for T2 lesions and for T1 hypointense lesions. (NCT00355134)
Timeframe: Baseline and Month 24

,,
Interventionmm^3 (Mean)
T2 lesions [N=248, 266, 251]T1 hypointense lesions [N=247, 266, 248]
Fingolimod 0.5 mg-223.27-111.28
Fingolimod 1.25 mg-436.92-99.13
Placebo541.83-37.68

Change From Baseline in Multiple Sclerosis Functional Composite (MSFC) Z-score

The Multiple Sclerosis Functional Composite (MSFC) is a multidimensional clinical outcome measure that includes quantitative tests of leg function/ambulation (Timed 25-Foot Walk), arm function (9-Hole Peg Test), and cognitive function (Paced Auditory Serial Addition Test). The overall MSFC z-score as an average of the three standardized scores derived using baseline data pooled over each treatment arm as reference population. Higher scores reflect better neurological function and a positive change from Baseline indicates improvement. (NCT00355134)
Timeframe: Baseline, Month 24 and end of study (up to approximately 54 months)

,,
Interventionunits on a scale (Mean)
Month 24 [N=250; 271; 258]End of Study [N=174; 187; 184]
Fingolimod 0.5 mg0.00-0.091
Fingolimod 1.25 mg-0.080.011
Placebo-0.070.019

Number of Gadolinium-enhanced T1 Lesions

Inflammatory disease activity was assessed by magnetic resonance imaging (MRI) measurement of the number of gadolinium-enhanced T1 lesions. (NCT00355134)
Timeframe: Month 24 and end of study (up to approximately 54 months)

,,
Interventionlesions (Mean)
Core Phase (Month 24) [N=251; 269; 256]End of Extension study [N=184; 194; 184]
Fingolimod 0.5 mg0.370.09
Fingolimod 1.25 mg0.240.46
Placebo1.220.45

Number of New or Newly Enlarged T2 Lesions

Inflammatory disease activity was assessed by magnetic resonance imaging (MRI) measurement of the number of new or newly enlarged T2 lesions, by year. (NCT00355134)
Timeframe: From Baseline until Month 48

,,
Interventionlesions (Mean)
Core Phase (Month 0 to 24) [N=245; 264; 251]Month 24 to 36 [N=103; 111; 102]Month 36 to 48 [N=24; 15; 15]
Fingolimod 0.5 mg2.30.450.07
Fingolimod 1.25 mg1.60.630.13
Placebo8.90.632.53

Percent Change From Baseline in Brain Volume

Brain volume was measured using magnetic resonance imaging (MRI). Change from Baseline in brain volume is expressed as a percentage of the Baseline brain volume. (NCT00355134)
Timeframe: Baseline, Month 24 and end of study (up to approximately 54 months)

,,
Interventionpercent change (Mean)
Month 24 [N=247; 266; 249]End of study [N=178; 187; 182]
Fingolimod 0.5 mg-0.858-1.266
Fingolimod 1.25 mg-0.595-1.130
Placebo-1.279-1.694

Percentage of Participants Free of 3-month Confirmed Disability Progression at Month 24 and End of Study

Disability progression was defined using the following criteria: One point increase from baseline in patients with Baseline Expanded Disability Status Scale (EDSS) score from 0 to 5.0; or half a point increase from Baseline in patients with Baseline EDSS score of 5.5 or above. A 3-month confirmed disability progression was defined as a 3-month sustained increase from Baseline in EDSS score. The EDSS quantifies disability in multiple sclerosis in 8 functional systems; the score ranges from 0 (normal) to 10 (death due to MS). Progression curves were generated by the Kaplan-Meier method. (NCT00355134)
Timeframe: 24 months and end of study (up to approximately 54 months)

,,
Interventionpercentage of participants (Number)
At month 24At end of study
Fingolimod 0.5 mg74.758.89
Fingolimod 1.25 mg78.366.64
Placebo71.063.51

Percentage of Participants Free of 6-month Confirmed Disability Progression at Month 24 and End of Study

Disability progression was defined using the following criteria: One point increase from baseline in patients with Baseline Expanded Disability Status Scale (EDSS) score from 0 to 5.0; or half a point increase from Baseline in patients with Baseline EDSS score of 5.5 or above. A 6-month confirmed disability progression was defined as a 6-month sustained increase from Baseline in EDSS score. The EDSS quantifies disability in multiple sclerosis in 8 functional systems; the score ranges from 0 (normal) to 10 (death due to MS). Progression curves were generated by the Kaplan-Meier method. (NCT00355134)
Timeframe: 24 months and end of study (up to approximately 54 months)

,,
Interventionpercentage of participants (Number)
At Month 24At end of study
Fingolimod 0.5 mg86.274.89
Fingolimod 1.25 mg86.979.92
Placebo82.275.03

Mean Number of Gadolinium (Gd)-Enhanced T1-weighted Lesions at End of Study

Total number of post-baseline Gd-enhanced lesions is calculated as a sum of all Gd-enhanced lesions seen on post-baseline scans per visit. Real (not per slice) lesions are counted in this analysis. The last observation was the last observation available for each patient which ranged from 1 to 2801 days (NCT00333138)
Timeframe: Last observation (Up to 80 months in average)

InterventionGD- enhanced T1 lesions (Mean)
Placebo0.8
Fingolimod (FTY720) 1.25 mg/Day0.4
Fingolimod (FTY720) 5.0 mg/Day0.5

Mean Number of Gadolinium (Gd)-Enhanced T1-weighted Lesions at Month 12

Total number of post-baseline Gd-enhanced lesions is calculated as a sum of all Gd-enhanced lesions seen on post-baseline scans per visit. Real (not per slice) lesions are counted in this analysis. (NCT00333138)
Timeframe: Month 12 (extension)

InterventionGD- enhanced T1 lesions (Mean)
Placebo0.4
Fingolimod (FTY720) 1.25 mg/Day1.0
Fingolimod (FTY720) 5.0 mg/Day0.2

Mean Number of Gadolinium (Gd)-Enhanced T1-weighted Lesions at Month 6 (Core)

Total number of post-baseline Gd-enhanced lesions is calculated as a sum of all Gd-enhanced lesions seen on post-baseline scans per visit. Real (not per slice) lesions are counted in this analysis. (NCT00333138)
Timeframe: Month 6 (Core)

InterventionGD- enhanced T1 lesions (Mean)
Placebo2.3
Fingolimod (FTY720) 1.25 mg/Day1.4
Fingolimod (FTY720) 5.0 mg/Day0.4

Mean Number of Gadolinium (Gd)-Enhanced T1-weighted Lesions at Month 60

Total number of post-baseline Gd-enhanced lesions is calculated as a sum of all Gd-enhanced lesions seen on post-baseline scans per visit. Real (not per slice) lesions are counted in this analysis. (NCT00333138)
Timeframe: Month 60 (extension)

InterventionGD- enhanced T1 lesions (Mean)
Placebo0.2
Fingolimod (FTY720) 1.25 mg/Day0.2
Fingolimod (FTY720) 5.0 mg/Day0.1

Change From Baseline in Volume of Total T2-weighted Lesions

Change in volume of total T2-weighted lesions by visit were summarized. Negative values indicate improvement (reduction in lesion volume) and positive values worsening (increase in lesion volume). The last observation was the last observation available for each patient which ranged from 1 to 2801 days. (NCT00333138)
Timeframe: Baseline to month 6, 12, 60 and Last observation (up to 80 months in average)

,,
Interventionmm^3 (Mean)
Month 6 (core) n=85, 89, 86Month 12 (extension) n=73, 77, 71Month 60 (extension) n=45, 49, 43Last observation (extension) n= 87, 89, 87
Fingolimod (FTY720) 1.25 mg/Day-115.811.66-204.09-92.24
Fingolimod (FTY720) 5.0 mg/Day-607.97-513.35-1429.98-907.48
Placebo146.15144.96-621.82-178.86

Mean Number of New T2-weighted Lesions

New T2 lesions at a specific visit were assessed relative to the previous visit scan. The total number of lesions (Month 1 to end of study) is calculated as the sum of the number of lesions at Months 1 to 6, Month 12, Month 60 and last observation. The last observation was the last observation available for each patient which ranged from 1 to 2801 days (NCT00333138)
Timeframe: (Core) Month 6 and (Extension) 12, 60, last observation (up to 80 months in average)

,,
InterventionGD enhanced T2 lesions (Mean)
Month 6 (Core), n= 82,88, 84Month 12 (Extension) n=71, 79, 71Month 60 (Extension) n=45, 50, 43Last observation (Extension) n=92, 93, 91
Fingolimod (FTY720) 1.25 mg/Day0.51.40.50.6
Fingolimod (FTY720) 5.0 mg/Day0.10.60.30.5
Placebo1.00.80.40.7

Mean Trough Blood Concentrations of FTY720

For each patient, the arithmetic mean of the two FTY720 trough blood levels from month 3 and 6 was calculated. This was taken as the patient's steady-state trough levels. Venous blood samples (3 mL) were collected before the dose in ethylenediaminetetraacetic acid (EDTA)-containing tubes at protocol-scheduled visits at months 3 and 6 in all patients. (NCT00333138)
Timeframe: Month 3 and 6

,
Interventionng/mL (Mean)
Month 3 (core)Month 6 (core)
FTY720 1.25 mg/Day7.647.38
FTY720 5.0 mg/Day30.528.9

Percentage of Participants Free of T1-weighted Lesions

A patient was defined as free of lesions if s/he had zero lesions. The last observation was the last observation available for each patient which ranged from 1 to 2801 days (NCT00333138)
Timeframe: Baseline, Months 6 (core), 12, 60 and Last Observation (up to 80 months in average)

,,
Interventionpercentage of participants (Number)
Baseline (Core) n=93, 92, 93Month 6 (Core) n= 83, 90, 87Month 12 (Extension) n= 72, 77, 71Month 60 (Extension) n= 45, 49, 44Last observation (Extension) n= 92, 93, 92
Fingolimod (FTY720) 1.25 mg/Day52.276.783.191.883.9
Fingolimod (FTY720) 5.0 mg/Day48.478.288.793.283.7
Placebo49.547.079.291.181.5

Percentage of Patients Free of Gd-enhanced T1-weighted and New T2- Weighted Lesions by Visit

A patient was defined as free of lesions if s/he had zero lesions. The sum of all new T2-weighted lesions at Month 1 to last observation was zero (the sum is missing if one of the assessments was missing). New T2 lesions at a specific visit were assessed relative to the previous visit scan. Exception: new T2 lesions at Month 24 were assessed relative to Month 12. The last observation was the last observation available for each patient which ranged from 1 to 2801 days (NCT00333138)
Timeframe: Month 6 and 12, 60, last observation (up to 80 months in average)

,,
Interventionpercentage of paticipants (Number)
Month 6 (Core), n= 80, 87, 83Month 12 (Extension) n=70, 77, 70Month 60 (Extension) n=45, 49, 43Last observation (Extension) n=92, 93,90
Fingolimod (FTY720) 1.25 mg/Day77.067.587.876.3
Fingolimod (FTY720) 5.0 mg/Day80.772.983.778.9
Placebo47.557.188.972.8

Time to Event Analysis: Kaplan Meier Estimates of Percentage of Relapse-free Patients

The Expanded Disability Status Scale (EDSS) is a scale for assessing disability in 8 functional systems (visual, brain stem, pyramidal, cerebellar, sensory, bowel & bladder, cerebral, other functions). An overall score ranging from 0 (normal) to 10 (death due to MS) is calculated. Disability progression was determined by the EDSS score based on the following criteria: One point increase from baseline in patients with baseline EDSS score from 0 to 5.0; or half a point increase in patients with baseline EDSS score of 5.5 or above. Percent of patients free of disability progression was calculated using the Kaplan-Meier method. The last observation was the last observation available for each patient which ranged from 1 to 2801 days (NCT00333138)
Timeframe: Month 6,12,60 and Last observation (up to 80 months in average)

,,
Interventionpercentage of participants (Number)
0 days (n=93,94,94)200 days (n=57, 73, 70)400 days (n=48, 63, 60)600 days (n=41, 57, 52)800 days (n=38, 48, 48)1000 days (n=35, 42, 45)1200 days (n=30, 38 40)1400 days (n=29, 36, 35)1600 days (n=27, 34, 34)1800 days (n=25, 33, 33)2000 days (n=23, 31, 31)2200 days (n=22, 28, 30)2400 days (n=21, 26, 27)2600 days (n=9, 12, 12)2880 days (n=0, 0, 0)
Fingolimod (FTY720) 1.25 mg/Day1.000.860.780.760.750.700.660.630.610.610.590.550.550.550
Fingolimod (FTY720) 5.0 mg/Day1.000.860.790.780.750.750.700.700.680.680.660.660.660.660
Placebo1.000.690.620.580.550.530.510.510.510.510.510.490.460.440

Volume of T2-weighted Lesions

Volume of total T2-weighted lesions by visit were summarized. The last observation was the last observation available for each patient which ranged from 1 to 2801 days (NCT00333138)
Timeframe: (Core) Month 6 and (Extension) 12, 60, last observation (up to 80 months in average)

,,
Interventionmm^3 (Mean)
Month 6 (Core) n=85, 91, 87Month 12 (Extension) n= 73, 79, 72Month 60 (Extension) n=45, 50, 44Last observation (Extension) n=87, 91, 88
Fingolimod (FTY720) 1.25 mg/Day10084.610707.19498.710090.2
Fingolimod (FTY720) 5.0 mg/Day8331.18363.26626.98061.8
Placebo9016.08264.16698.38757.2

Immune Response 3 Weeks After Seasonal Influenza Vaccination

"Percentage of participants who responded to treatment with the seasonal influenza vaccine 3 weeks after vaccination. Response was defined as patients fulfilling one of the following criteria for at least one of the three strains contained in the seasonal influenza vaccine:~Seroconversion: The pre-vaccination antibody titer measurement was <1:10 and the post-vaccination measurement is ≥1:40.~Significant increase in antibody titer: The pre-vaccination antibody titer measurement was ≥1:10 and the increase in antibody titer from this to the post-vaccination measurement is ≥ 4-fold." (NCT01199861)
Timeframe: Week 6 (pre-vaccination) and 3 weeks after vaccination (Study week 9)

Interventionpercentage of participants (Number)
Fingolimod53.3
Placebo83.7

Immune Response 3 Weeks After Tetanus Toxoid Booster

"Percentage of participants with an immune response to a single dose of tetanus toxoid three weeks after vaccination. A patient was considered a responder to tetanus toxoid booster vaccination if one of the following criteria was met:~Seroconversion: The pre-vaccination antibody titer measurement was <0.1 IU/ml and the post-vaccination measurement was ≥0.4 IU/ml.~Significant increase: The pre-vaccination antibody titer measurement was ≥0.1 IU/ml and the increase in antibody titer from this to the post-vaccination measurement was ≥4- fold." (NCT01199861)
Timeframe: Week 6 (pre-vaccination) and 3 weeks after vaccination (Study Week 9)

Interventionpercentage of participants (Number)
Fingolimod40.0
Placebo60.5

Immune Response 6 Weeks After Seasonal Influenza Vaccination

"Percentage of participants who responded to treatment with the seasonal influenza vaccine 6 weeks after vaccination. Response was defined as patients fulfilling one of the following criteria for at least one of the three strains contained in the seasonal influenza vaccine:~Seroconversion: The pre-vaccination antibody titer measurement was <1:10 and the post-vaccination measurement is ≥1:40.~Significant increase in antibody titer: The pre-vaccination antibody titer measurement was ≥1:10 and the increase in antibody titer from this to the post-vaccination measurement is ≥ 4-fold." (NCT01199861)
Timeframe: Week 6 (pre-vaccination) and 6 weeks after vaccination (Study week 12).

Interventionpercentage of participants (Number)
Fingolimod43.2
Placebo74.4

Immune Response 6 Weeks After Tetanus Toxoid Booster

"Percentage of participants with an immune response to a single dose of tetanus toxoid six weeks after vaccination. A patient was considered a responder to tetanus toxoid booster vaccination if one of the following criteria was met:~Seroconversion: The pre-vaccination antibody titer measurement was <0.1 IU/ml and the post-vaccination measurement was ≥0.4 IU/ml.~Significant increase: The pre-vaccination antibody titer measurement was ≥0.1 IU/ml and the increase in antibody titer from this to the post-vaccination measurement was ≥4- fold." (NCT01199861)
Timeframe: Week 6 (pre-vaccination) and 6 weeks after vaccination (Study Week 12)

Interventionpercentage of participants (Number)
Fingolimod37.5
Placebo48.8

Change From Baseline in Seasonal Influenza Vaccine Antibody-titer 3 Weeks After Vaccination

Change from Baseline was expressed by the ratio of post-vaccination to pre-vaccination antibody titer for each of the three strains included in the seasonal influenza vaccine. Inhibition of an immune response to each strain included in the seasonal influenza vaccine was assessed by the relative difference of the geometric mean antibody titer ratio on fingolimod as compared to placebo three weeks after a single dose of seasonal influenza vaccine. (NCT01199861)
Timeframe: Pre-vaccination (Week 6) and 3 weeks after vaccination (Study Week 9).

,
Interventionratio (Number)
A/California/7/09(H1N1)A/Perth/16/2009(H3N2)B/Brisbane/60/2008
Fingolimod2.450.491.34
Placebo4.140.362.40

Change From Baseline in Seasonal Influenza Vaccine Antibody-titer 6 Weeks After Vaccination

Change from Baseline was expressed by the ratio of post-vaccination to pre-vaccination antibody titer for each of the three strains included in the seasonal influenza vaccine. Inhibition of an immune response to each strain included in the seasonal influenza vaccine was assessed by the relative difference of the geometric mean antibody titer ratio on fingolimod as compared to placebo six weeks after a single dose of seasonal influenza vaccine. (NCT01199861)
Timeframe: Pre-vaccination (Week 6) and 6 weeks after vaccination (Study Week 12).

,
Interventionratio (Number)
A/California/7/09(H1N1)A/Perth/16/2009(H3N2)B/Brisbane/60/2008
Fingolimod1.810.361.08
Placebo2.910.282.07

Number of Participants With Adverse Events (AEs)

"Relationship to study drug was determined by the investigator (suspected/not suspected).~A serious AE is defined as an event which fulfills one of the following criteria:~is fatal or life-threatening;~results in persistent or significant disability/incapacity;~constitutes a congenital anomaly/birth defect;~requires inpatient hospitalization or prolongation of existing hospitalization;~is medically significant, i.e., jeopardizes the patient or may require intervention to prevent one of the outcomes listed above." (NCT01199861)
Timeframe: From first dose of study drug until 45 days after the last dose of study drug (130 days).

,
Interventionparticipants (Number)
Any adverse eventAE related to study drugSerious adverse eventAdverse events leading to discontinuation
Fingolimod824211
Placebo341120

Change From Baseline in Patient-reported Activities of Daily Living (ADL) Using the Multiple Sclerosis Activities Scale (PRIMUS-Activities) at Month 6

The PRIMUS activity measure is a 15-item assessment of patient-reported ADL. The PRIMUS-Activities total score was calculated by summing the 15 item scores after recoding the responses from 1 - 3 to 0 - 2. Totals scores range from 0 to 30 with higher scores indicating greater activity limitation. If no more than 20% of the items were missing, the total score was the product of the mean response of the non-missing items and the total number of items. If more than 20% of all items were missing, the total score was set to missing. A negative change from baseline indicates improvement. (NCT01216072)
Timeframe: Baseline, Month 6

Interventionunits on a scale (Mean)
Fingolimod-0.6
Multiple Sclerosis Disease Modifying Treatments (MS DMTs)-0.2

Change From Baseline in the Global Satisfaction Subscale of the Treatment Satisfaction Questionnaire for Medication (TSQM) at Month 6

The TSQM was developed and validated as a general measure for treatment satisfaction. It contains 14 items assessing the following 4 domains: effectiveness (sum of scores for questions 1 - 3), side effects (sum of scores for questions 4 - 8), convenience (sum of scores for questions 9 - 11) and Global Satisfaction (sum of scores for questions 12 - 14). The primary analysis was on Global Satisfaction. Question 12 scored as 1(not at all confident) to 5 (extremely confident); question 13 scored as 1(not at all certain) to 5(extremely certain); and question 14 scored as 1(extremely dissatisfied) to 7(extremely satisfied). The scores of the domain were added together and an algorithm was used to create a score of 0 to 100. Higher scores indicated greater satisfaction. A positive change from baseline indicates improvement. (NCT01216072)
Timeframe: Baseline, Month 6

Interventionunits on a scale (Mean)
Fingolimod22.5
Multiple Sclerosis Disease Modifying Treatments (MS DMTs)3.5

Change From Baseline in the Patient-reported Convenience Subscale Using the TSQM v1.4

The convenience subscale was scored as follows: questions 9 and 10 scored as 1(extremely difficult) to 7 (extremely easy), and question 11 scored as 1(extremely inconvenient) to 7 (extremely convenient). The scores of the domain were added together and an algorithm was used to create a score of 0 to 100. Higher scores indicated greater satisfaction. A positive change from baseline indicates improvement. (NCT01216072)
Timeframe: Baseline, Month 6

Interventionunits on a scale (Mean)
Fingolimod41.8
Multiple Sclerosis Disease Modifying Treatments (MS DMTs)1.5

Change From Baseline in the Patient-reported Effectiveness Subscale Using the TSQM v1.4

The effectiveness scale was scored as follows: 1(extremely dissatisfied) to 7(extremely satisfied). The scores of the domain were added together and an algorithm was used to create a score of 0 to 100. Higher scores indicated greater satisfaction. A positive change from baseline indicates improvement. (NCT01216072)
Timeframe: Baseline, Month 6

Interventionunits on a scale (Mean)
Fingolimod15.5
Multiple Sclerosis Disease Modifying Treatments (MS DMTs)2.8

Change From Baseline in the Patient-reported Side Effects Subscale Using the TSQM v1.4

The Side Effects subscale was scored as follows: question 4 scored as 0(no) or 1(yes); question 5 scored as 1(extremely bothersome) to 5(not at all bothersome); and questions 6 - 8 scored as 1(a great deal) to 5(not at all). The scores of the domain were added together and an algorithm was used to create a score of 0 to 100. Higher scores indicated greater satisfaction. A positive change from baseline indicates improvement. (NCT01216072)
Timeframe: Baseline, Month 6

Interventionunits on a scale (Mean)
Fingolimod22.9
Multiple Sclerosis Disease Modifying Treatments (MS DMTs)3.5

Change From Baseline in Patient-reported Depression Using the Beck Depression Inventory (BDI-II)

The Beck Depression Inventory (BDI-II) is a 21-question multiple-choice self-report inventory. Each item is scored from 0 to 3. The questions in the BDI-II refer to how the patient has been feeling over the past two weeks specifically. The BDI-II total score was calculated by summing the 21 item scores. Final scores ranged from 0 to 63 where higher scores indicated more severe depression. If no more than 20% of the items were missing, the total score was the product of the mean response of the non-missing items and the total number of items. If more than 20% of all items were missing, the total score was set to missing. A negative change indicates improvement. (NCT01216072)
Timeframe: Baseline, Month 3, Month 6

,
Interventionunits on a scale (Mean)
Change from Baseline to Month 3 (n=748,236)Change from Baseline to Month 6 (n=709,223)
Fingolimod-3.2-3.4
Multiple Sclerosis Disease Modifying Treatments (MS DMTs)-0.8-0.6

Change From Baseline in Patient-reported Fatigue Using the Fatigue Severity Scale (FSS)

The Fatigue Severity Scale (FSS) is a 9-item assessment scale measuring fatigue and its effects, using a scale from 1 to 7, with higher scores indicating greater fatigue, or greater negative effects of fatigue on daily living. The FSS 9 item total score was calculated by summing the first 9 item scores and dividing by the number of non-missing items. If no more than 20% of the items were missing, the total score was the product of the mean response of the non missing items and the total number of items. If more than 20% of all items were missing, the total score was set to missing. A negative change from baseline indicates improvement. (NCT01216072)
Timeframe: Baseline, Month 3, Month 6

,
Interventionunits on a scale (Mean)
Change from Baseline to Month 3 (n=710,232)Change from Baseline to Month 6 (n=687,218)
Fingolimod-0.3-0.4
Multiple Sclerosis Disease Modifying Treatments (MS DMTs)0.00.0

Change From Baseline in Patient-reported Health-related Quality-of-life Using the Short Form Health Survey v2 Standard (SF-36 v2)

The SF-36v2 is a validated health-related quality of life instrument used in numerous disease states, including MS. It is a self-administered survey that measures 8 domains of health including: physical functioning, role limitations due to physical health, bodily pain, general health perceptions, vitality, social functioning, role limitations due to emotional problems and general mental health. Additionally, two summary scale scores can be calculated: the Physical Component Summary (PCS) and the Mental Component Summary (MCS). If half or more questions within a domain were answered, then a score was calculated for that domain. Otherwise, the patient score for that domain was set to missing. If the patient was missing any 1 of the 8 scale scores, then the physical and mental component scores were set to missing. An algorithm was used to create a score from 0 to 100 for each domain score and component score. A positive change from baseline indicates improvement. (NCT01216072)
Timeframe: Baseline, Month 6

,
Interventionunits on a scale (Mean)
Physical Function (n=745,226)Role Limitations d/t Physical Health (n=742,226)Bodily Pain (n=742,226)General Health Perceptions (n=737,224)Vitality (n=745,227)Social Functioning (n=747,227)Role Limitation d/t Emotional Problems (n=745,226)General Mental Health (n=745,227)Physical Component Summary Scale (n=724,222)Mental Component Summary Scale (n=724,222)
Fingolimod1.52.82.00.82.82.61.62.21.72.3
Multiple Sclerosis Disease Modifying Treatments (MS DMTs)0.41.2-0.3-0.30.60.8-0.60.30.30.2

Number of Patients Who Experienced Adverse Events, Serious Adverse Events and Death

In this analysis, patients with all (serious and non-serious) adverse events, serious adverse events and death were reported. (NCT01216072)
Timeframe: 9 months (6 month core + 3 month Extension)

,,
InterventionParticipants (Number)
Adverse Events (serious and non-serious)Serious Adverse EventsDeath
Extension Fingolimod Period12370
Fingolimod617310
Multiple Sclerosis Disease Modifying Treatments (MS DMTs)15250

Physician-reported Clinical Global Impression of Improvement (CGI-I)

The CGI-I is a rating scale allowing a physician-reported global evaluation of the subject's improvement over time. The Investigator assessed the subject's clinical change relative to the symptoms at baseline on the CGI-I, a seven-point scale, with rating as follows: 1=Very much improved, 2=Much improved, 3=Minimally improved, 4=No change, 5=Minimally worse, 6=Much worse, 7=Very much worse. The assessments were completed at Month 3 and Month 6. A lower score indicates improvement. (NCT01216072)
Timeframe: Month 3, Month 6

,
Interventionunits on a scale (Mean)
Month 3 (n=758,242)Month 6 (n=727,228)
Fingolimod3.43.2
Multiple Sclerosis Disease Modifying Treatments (MS DMTs)3.93.9

Frequency of Relapses in Patients Treated for up to 24 Months

Frequency of relapses assessed by the annualized relapse rate (ARR). The ARR is defined as the average number of confirmed relapses per year (total number of confirmed relapses divided by the total days in the study multiplied by 365.25). (NCT01892722)
Timeframe: 24 months

InterventionConfirmed relapse per year (Mean)
Fingolimod0.122
Interferon Beta-1a0.675

Part I: Aggregate Annualized Relapse Rates (ARR) From First Dose of Fingolimod

Annualized relapse rate (ARR) is defined as the number of all relapses (including both confirmed and unconfirmed relapses) experienced during a specific period of time adjusted to a one-year period. ARR is calculated as follows: (total number of all relapses) / (total number of days in the study for all patients for that specific period of time) x 365.25. Month 0 is the first dose of fingolimod study drug among all studies in which patient participated. Only descriptive analysis performed. (NCT01201356)
Timeframe: Month 0 (Core Baseline) to End of Follow-up Visit (an average of 162 months)

InterventionAnnual number of relapses per patient (Number)
Month 0 to Month 6Month 0 to Month 12Month 0 to Month 24Month 0 to Month 36Month 0 to Month 48Month 0 to Month 60Month 0 to Month 72Month 0 to Month 84Month 0 to Month 96Month 0 to Month 108Month 0 to Month 120Month 0 to Month 132Month 0 to Month 144Month 0 to Month 156Month 0 to end of StudyMonth 0 to end of Follow-up
Fingolimod 0.5 mg/Day0.3250.2730.2370.2170.2080.1970.1900.1820.1770.1720.1700.1700.1690.1690.1660.169

Part I: Annualized Rate of Brain Atrophy (ARBA) Relative to First Dose of Fingolimod

"The annualized rate of brain volume change is an averaged annual percentage change in brain volume. ARBA was calculated as: ARBA = [(SIENA/100+1) ^ (365.25/#days)-1]*100 where SIENA=(Vk/V0-1)*100 and Vk is the brain volume at time k, V0 is the brain volume at time 0 and k is the total number of days in the study for all patients for that specific period of time) × 365.25. Only descriptive analysis performed." (NCT01201356)
Timeframe: Month 3 to Month 156

InterventionRatio (Mean)
Month 3Month 6Month 12Month 24Month 36Month 48Month 60Month 72Month 84Month 96Month 108Month 120Month 132Month 144Month 156
Fingolimod 0.5 mg/Day-0.73-0.39-0.35-0.37-0.35-0.37-0.34-0.35-0.35-0.31-0.33-0.36-0.43-0.36-0.35

Part I: Annualized Rates of New or Newly Enlarging T2 Lesions (ARneT2) Compared With First Dose of Fingolimod

Annualized rate of new/newly enlarging T2 lesions (ARneT2) is defined as the number of new or newly enlarging T2 lesions experienced during a specific period of time adjusted to a one-year period. ARneT2 was calculated as follows: (total number of new/newly enlarging T2 lesions) / (total number of days in the study for all patients for that specific period of time) x 365.25.Month 0 is the first dose of fingolimod study drug among all studies in which patient participated. Only descriptive analysis performed. (NCT01201356)
Timeframe: Month 0 (Core Baseline) to End of Study (an average of Month 156)

InterventionAnnual number of T2 lesions per patient (Number)
Month 0 to Month 3Month 0 to Month 6Month 0 to Month 12Month 0 to Month 24Month 0 to Month 36Month 0 to Month 48Month 0 to Month 60Month 0 to Month 72Month 0 to Month 84Month 0 to Month 96Month 0 to Month 108Month 0 to Month 120Month 0 to Month 132Month 0 to Month 144Month 0 to Month 156Month 0 to end of study
Fingolimod 0.5 mg/Day12.3242.0731.3601.0421.0111.0080.9570.9630.9060.8130.7130.7020.6590.6810.6370.751

Part I: Change From First Dose of Fingolimod in Expanded Disability Status Scale (EDSS)

The EDSS is a scale for assessing neurological impairment in MS (Kurtzke 1983) including (1) a series of scores in each of eight functional systems, and (2) the EDSS steps (ranging from 0 (normal) to 10 (death due to MS). The functional systems are Visual, Brain Stem, Pyramidal, Cerebellar, Sensory, Bowel and Bladder, Cerebral and Other functions. Based on the assessment of each FS, the participant's overall score is determined between 0 to 10. A negative change from baseline indicates improvement. Only descriptive analysis performed. (NCT01201356)
Timeframe: Month 0 (Core Baseline) to End of Follow-up Visit (an average of 162 months)

InterventionEDDS Overall Score (Mean)
Baseline (BL)Change from BL at Month 3Change from BL at Month 6Change from BL at Month 9Change from BL at Month 12Change from BL at Month 15Change from BL at Month 18Change from BL at Month 21Change from BL at Month 24Change from BL at Month 27Change from BL at Month 30Change from BL at Month 33Change from BL at Month 36Change from BL at Month 39Change from BL at Month 42Change from BL at Month 45Change from BL at Month 48Change from BL at Month 51Change from BL at Month 54Change from BL at Month 57Change from BL at Month 60Change from BL at Month 63Change from BL at Month 66Change from BL at Month 69Change from BL at Month 72Change from BL at Month 75Change from BL at Month 78Change from BL at Month 81Change from BL at Month 84Change from BL at Month 87Change from BL at Month 90Change from BL at Month 93Change from BL at Month 96Change from BL at Month 99Change from BL at Month 102Change from BL at Month 105Change from BL at Month 108Change from BL at Month 111Change from BL at Month 114Change from BL at Month 117Change from BL at Month 120Change from BL at Month 123Change from BL at Month 126Change from BL at Month 129Change from BL at Month 132Change from BL at Month 135Change from BL at Month 138Change from BL at Month 141Change from BL at Month 144Change from BL at Month 147Change from BL at Month 150Change from BL at Month 153Change from BL at Month 156Change from BL at Month 159Change from BL at Month 162Change from BL at End of studyChange from BL at Month 3 follow-upChange from BL at Month 6 follow-up
Fingolimod 0.5 mg/Day2.39-0.06-0.07-0.09-0.07-0.08-0.05-0.08-0.01-0.03-0.00-0.020.010.020.040.060.060.060.040.090.170.080.150.140.220.130.280.180.250.240.290.180.310.180.300.210.280.240.300.350.400.600.380.400.400.510.420.540.600.670.440.230.261.170.750.140.290.56

Part I: Change From First Dose of Fingolimod in Total T1 Hypointense Lesions Volume

T1 hypointense lesion (black hole) volume was summarized by presenting descriptive statistics for change from first dose of fingolimod baseline values by visit. (NCT01201356)
Timeframe: Month 3 to End of Study (Study Completion Visit)

Interventionmm^3 (Mean)
T1 volume change at Month 3T1 volume change at Month 12T1 volume change at Month 24T1 volume change at Month 36T1 volume change at Month 48T1 volume change at Month 60T1 volume change at Month 72T1 volume change at Month 84T1 volume change at Month 96T1 volume change at Month 108T1 volume change at Month 120T1 volume change at Month 132T1 volume change at End of Study
Fingolimod 0.5 mg/Day-519.849.164.1151.08524.8853.6975.7930.1753.4628.7817.3726.7800.6

Part I: Change From First Dose of Fingolimod in Total T2 Lesions Volume

Total volume of T2 lesions was summarized by presenting descriptive statistics for change from first dose of fingolimod baseline values by visit. (NCT01201356)
Timeframe: Month 3 to End of Study (Study Completion Visit)

Interventionmm^3 (Mean)
T2 volume change at Month 3T2 volume change at Month 6T2 volume change at Month 12T2 volume change at Month 24T2 volume change at Month 36T2 volume change at Month 48T2 volume change at Month 60T2 volume change at Month 72T2 volume change at Month 84T2 volume change at Month 96T2 volume change at Month 108T2 volume change at Month 120T2 volume change at Month 132T2 volume change at Month 144T2 volume change at Month 156T2 volume change at End of Study
Fingolimod 0.5 mg/Day-749.5-207.0-55.016.2235.8875.11546.31719.21635.81303.91562.01393.1905.9702.7274.01588.5

Part I: Number of Participants With Confirmed 6-month Disability Progression After First Dose of Fingolimod

Disability progression was defined based on an increase in the EDSS score by 1.5 point for patients with a first dose of fingolimod (FDF) baseline EDSS score of 0, 1 point for patients with FDF baseline EDSS of >=1 and <=5.5, and by 0.5 points for patients with an FDF baseline EDSS>5.5, confirmed after 6 months and all intermediate EDSS assessments. A 6-month confirmed disability progression was defined as a 6-month sustained increase from the reference (potential onset of progression) value in the EDSS scores. i.e., every EDSS score (scheduled or unscheduled) within a 6-month duration after the first progression should meet the progression criteria as specified above. The confirmation could only happen at a scheduled visit and in the absence of a relapse. Only descriptive analysis performed. (NCT01201356)
Timeframe: Month 12 to Month 156

InterventionParticipants (Number)
Month 12Month 24Month 36Month 48Month 60Month 72Month 84Month 96Month 108Month 120Month 132Month 144Month 156
Fingolimod 0.5 mg/Day212336434519590659714753767772775776777

Part I: Number of Participants With Relapses (Confirmed and Unconfirmed) From First Dose of Fingolimod

"A relapse is defined as the appearance of a new neurological abnormality or worsening of previously stable or improving pre-existing neurological abnormality, separated by at least 30 days from onset of a preceding clinical demyelinating event. The abnormality must be present for at least 24 hours and occur in the absence of fever (<37.5°C) or infection.~In Study Part One, a relapse must be confirmed by an Expanded Disability Status Scale (EDSS) certified Physician within 7 days of the onset of symptoms. A relapse is confirmed when it is accompanied by an increase of at least half a step (0.5) on the EDSS or an increase of 1 point on two different Functional Systems (FS) of the EDSS or 2 points on one of the FS (excluding Bowel/Bladder or Cerebral FS). Month 0 is the first dose of fingolimod study drug among all studies in which patient participated. Only descriptive analysis performed." (NCT01201356)
Timeframe: Month 0 (Core Baseline) to End of Follow-up Visit (an average of 162 months)

InterventionParticipants (Number)
Month 0 to Month 6Month 0 to Month 12Month 0 to Month 24Month 0 to Month 36Month 0 to Month 48Month 0 to Month 60Month 0 to Month 72Month 0 to Month 84Month 0 to Month 96Month 0 to Month 108Month 0 to Month 120Month 0 to Month 132Month 0 to Month 144Month 0 to Month 156Month 0 to end of StudyMonth 0 to end of Follow-up
Fingolimod 0.5 mg/Day65599214611794212723832616279329443036306330723075307929703079

Part I: Percent Brain Volume Change (PBVC) Relative to First Dose of Fingolimod

Descriptive statistics on percent brain volume change from first dose of fingolimod baseline were presented by visit. A negative change from baseline indicates improvement. (NCT01201356)
Timeframe: Month 3 to Month 156

InterventionPercent change (Mean)
Percent volume change at Month 3Percent volume change at Month 6Percent volume change at Month 12Percent volume change at Month 24Percent volume change at Month 36Percent volume change at Month 48Percent volume change at Month 60Percent volume change at Month 72Percent volume change at Month 84Percent volume change at Month 96Percent volume change at Month 108Percent volume change at Month 120Percent volume change at Month 132Percent volume change at Month 144Percent volume change at Month 156
Fingolimod 0.5 mg/Day-0.19-0.20-0.36-0.74-1.03-1.44-1.65-2.02-2.38-2.41-2.91-3.42-4.61-4.21-4.33

Parts I and II: Number of Participants With Adverse Events, Serious Adverse Event, and Death

Analysis of absolute and relative frequencies for Adverse Event (AE), Serious Adverse Event (SAE) and Deaths by primary System Organ Class (SOC) to demonstrate that Fingolimod 0.5 mg/day is safe in patients with relapsing forms of Multiple Sclerosis (MS) through the monitoring of relevant clinical and laboratory safety parameters. Only descriptive analysis performed. (NCT01201356)
Timeframe: Baseline (Part I) to Month 6 Follow-up (Part II), up to 8 years

InterventionParticipants (Count of Participants)
Adverse Event (AEs)Serious Adverse Events (SAEs)Deaths
Fingolimod 0.5 mg/Day212551516

Part I: Number of Participants With Categorized Change From First Dose of Fingolimod in Expanded Disability Status Scale (EDSS) Overall Score

"The EDSS is a scale for assessing neurological impairment in MS (Kurtzke 1983) including (1) a series of scores in each of eight functional systems, and (2) the EDSS steps (ranging from 0 (normal) to 10 (death due to MS). The functional systems are Visual, Brain Stem, Pyramidal, Cerebellar, Sensory, Bowel and Bladder, Cerebral and Other functions. Based on the assessment of each FS, the participant's overall score is categorized as Improvement, Stable or Deterioration.~If baseline EDSS score is <=5, improvement is indicated by an EDSS score change of <= -1, stable is indicated by an EDSS score change of > -1 and <= 0.5, deterioration is indicated by an EDSS score change of > 0.5; if baseline EDSS score is > 5, improvement is indicated by an EDSS score change of <= -0.5, stable is indicated by an EDSS score change of > -0.5 and <= 0, deterioration is indicated by an EDSS score change of > 0. Only descriptive analysis performed." (NCT01201356)
Timeframe: Month 3 to Month 6 Follow-up

InterventionParticipants (Count of Participants)
Month 372327500Month 672327500Month 972327500Month 1272327500Month 1572327500Month 1872327500Month 2172327500Month 2472327500Month 2772327500Month 3072327500Month 3372327500Month 3672327500Month 3972327500Month 4272327500Month 4572327500Month 4872327500Month 5172327500Month 5472327500Month 5772327500Month 6072327500Month 6372327500Month 6672327500Month 6972327500Month 7272327500Month 7572327500Month 7872327500Month 8172327500Month 8472327500Month 8772327500Month 9072327500Month 9372327500Month 9672327500Month 9972327500Month 10272327500Month 10572327500Month 10872327500Month 11172327500Month 11472327500Month 11772327500Month 12072327500Month 12372327500Month 12672327500Month 12972327500Month 13272327500Month 13572327500Month 13872327500Month 14172327500Month 14472327500Month 14772327500Month 15072327500Month 15372327500Month 15672327500Month 15972327500Month 16272327500End of study72327500Month 3 follow-up72327500Month 6 follow-up72327500
ImprovementStableDeterioration
Fingolimod 0.5 mg/Day374
Fingolimod 0.5 mg/Day3126
Fingolimod 0.5 mg/Day269
Fingolimod 0.5 mg/Day508
Fingolimod 0.5 mg/Day2900
Fingolimod 0.5 mg/Day340
Fingolimod 0.5 mg/Day482
Fingolimod 0.5 mg/Day2425
Fingolimod 0.5 mg/Day304
Fingolimod 0.5 mg/Day422
Fingolimod 0.5 mg/Day1930
Fingolimod 0.5 mg/Day322
Fingolimod 0.5 mg/Day405
Fingolimod 0.5 mg/Day1764
Fingolimod 0.5 mg/Day285
Fingolimod 0.5 mg/Day345
Fingolimod 0.5 mg/Day1361
Fingolimod 0.5 mg/Day271
Fingolimod 0.5 mg/Day376
Fingolimod 0.5 mg/Day1524
Fingolimod 0.5 mg/Day320
Fingolimod 0.5 mg/Day1299
Fingolimod 0.5 mg/Day294
Fingolimod 0.5 mg/Day313
Fingolimod 0.5 mg/Day1271
Fingolimod 0.5 mg/Day270
Fingolimod 0.5 mg/Day305
Fingolimod 0.5 mg/Day1204
Fingolimod 0.5 mg/Day326
Fingolimod 0.5 mg/Day1172
Fingolimod 0.5 mg/Day292
Fingolimod 0.5 mg/Day309
Fingolimod 0.5 mg/Day1065
Fingolimod 0.5 mg/Day288
Fingolimod 0.5 mg/Day284
Fingolimod 0.5 mg/Day1015
Fingolimod 0.5 mg/Day276
Fingolimod 0.5 mg/Day282
Fingolimod 0.5 mg/Day948
Fingolimod 0.5 mg/Day287
Fingolimod 0.5 mg/Day875
Fingolimod 0.5 mg/Day263
Fingolimod 0.5 mg/Day250
Fingolimod 0.5 mg/Day838
Fingolimod 0.5 mg/Day264
Fingolimod 0.5 mg/Day244
Fingolimod 0.5 mg/Day784
Fingolimod 0.5 mg/Day247
Fingolimod 0.5 mg/Day256
Fingolimod 0.5 mg/Day788
Fingolimod 0.5 mg/Day251
Fingolimod 0.5 mg/Day694
Fingolimod 0.5 mg/Day245
Fingolimod 0.5 mg/Day175
Fingolimod 0.5 mg/Day577
Fingolimod 0.5 mg/Day234
Fingolimod 0.5 mg/Day182
Fingolimod 0.5 mg/Day540
Fingolimod 0.5 mg/Day191
Fingolimod 0.5 mg/Day165
Fingolimod 0.5 mg/Day499
Fingolimod 0.5 mg/Day216
Fingolimod 0.5 mg/Day503
Fingolimod 0.5 mg/Day193
Fingolimod 0.5 mg/Day440
Fingolimod 0.5 mg/Day206
Fingolimod 0.5 mg/Day156
Fingolimod 0.5 mg/Day437
Fingolimod 0.5 mg/Day201
Fingolimod 0.5 mg/Day136
Fingolimod 0.5 mg/Day398
Fingolimod 0.5 mg/Day226
Fingolimod 0.5 mg/Day150
Fingolimod 0.5 mg/Day456
Fingolimod 0.5 mg/Day217
Fingolimod 0.5 mg/Day132
Fingolimod 0.5 mg/Day407
Fingolimod 0.5 mg/Day207
Fingolimod 0.5 mg/Day147
Fingolimod 0.5 mg/Day450
Fingolimod 0.5 mg/Day227
Fingolimod 0.5 mg/Day120
Fingolimod 0.5 mg/Day413
Fingolimod 0.5 mg/Day204
Fingolimod 0.5 mg/Day143
Fingolimod 0.5 mg/Day386
Fingolimod 0.5 mg/Day187
Fingolimod 0.5 mg/Day117
Fingolimod 0.5 mg/Day375
Fingolimod 0.5 mg/Day186
Fingolimod 0.5 mg/Day110
Fingolimod 0.5 mg/Day325
Fingolimod 0.5 mg/Day159
Fingolimod 0.5 mg/Day283
Fingolimod 0.5 mg/Day154
Fingolimod 0.5 mg/Day103
Fingolimod 0.5 mg/Day261
Fingolimod 0.5 mg/Day131
Fingolimod 0.5 mg/Day87
Fingolimod 0.5 mg/Day138
Fingolimod 0.5 mg/Day74
Fingolimod 0.5 mg/Day198
Fingolimod 0.5 mg/Day101
Fingolimod 0.5 mg/Day181
Fingolimod 0.5 mg/Day79
Fingolimod 0.5 mg/Day31
Fingolimod 0.5 mg/Day94
Fingolimod 0.5 mg/Day52
Fingolimod 0.5 mg/Day88
Fingolimod 0.5 mg/Day42
Fingolimod 0.5 mg/Day14
Fingolimod 0.5 mg/Day45
Fingolimod 0.5 mg/Day26
Fingolimod 0.5 mg/Day11
Fingolimod 0.5 mg/Day36
Fingolimod 0.5 mg/Day10
Fingolimod 0.5 mg/Day39
Fingolimod 0.5 mg/Day18
Fingolimod 0.5 mg/Day12
Fingolimod 0.5 mg/Day30
Fingolimod 0.5 mg/Day19
Fingolimod 0.5 mg/Day21
Fingolimod 0.5 mg/Day7
Fingolimod 0.5 mg/Day22
Fingolimod 0.5 mg/Day15
Fingolimod 0.5 mg/Day16
Fingolimod 0.5 mg/Day8
Fingolimod 0.5 mg/Day3
Fingolimod 0.5 mg/Day9
Fingolimod 0.5 mg/Day5
Fingolimod 0.5 mg/Day2
Fingolimod 0.5 mg/Day6
Fingolimod 0.5 mg/Day0
Fingolimod 0.5 mg/Day1
Fingolimod 0.5 mg/Day610
Fingolimod 0.5 mg/Day2419
Fingolimod 0.5 mg/Day785
Fingolimod 0.5 mg/Day203
Fingolimod 0.5 mg/Day907
Fingolimod 0.5 mg/Day381
Fingolimod 0.5 mg/Day29
Fingolimod 0.5 mg/Day111

Percentage of Participants With Disability Progression as Measured by Expanded Disability Status Scale (EDSS) (FAS)

"EDSS is a scale for assessing neurologic impairment in MS. It is a two-part system including~(1) a series of scores in each of eight functional systems, and (2) the EDSS steps (ranging from 0 (normal) to 10 (death due to MS). The definition of disability progression was based on increases in EDSS from baseline and depended on the EDSS baseline value: Disability progression was defined as a 1.5 increase in EDSS from baseline in subjects with a baseline EDSS score between 0.0 and 0.5, as a 1.0 increase in EDSS from baseline in subjects with a baseline EDSS score between 1.0 and 5.0 inclusive and 0.5 increase from baseline in subjects with EDSS score > 5.0." (NCT02720107)
Timeframe: Baseline up to approximately 48 months

Interventionpercentage of participants (Number)
Fingolimod21.54

Change From Baseline in Disability Progression Assessed With the Expanded Disability Status Scale (EDSS) at Month 6 and Month 48 (FAS)

"EDSS is a scale for assessing neurologic impairment in MS. It is a two-part system including~(1) a series of scores in each of eight functional systems, and (2) the EDSS steps (ranging from 0 (normal) to 10 (death due to MS). The definition of disability progression was based on increases in EDSS from baseline and depended on the EDSS baseline value: Disability progression was defined as a 1.5 increase in EDSS from baseline in subjects with a baseline EDSS score between 0.0 and 0.5, as a 1.0 increase in EDSS from baseline in subjects with a baseline EDSS score between 1.0 and 5.0 inclusive and 0.5 increase from baseline in subjects with EDSS score > 5.0." (NCT02720107)
Timeframe: Baseline, month 6 up to approximately 48 months

Interventionscores on a scale (Mean)
BaselineMonth 6Month 48Change from baseline to month 6Change from month 6 to month 48Change from baseline to month 48
Fingolimod2.72.62.7-0.10.20.0

Change in Immune Status of B Cells, Monocytes and Natural Killer Cells (NK) Cells (FAS)

Changes in immune status of B cells (CD19+, CD20+, CD69+), monocytes (CD14+) and NK cells (CD56+) were analyzed as a percentage of parent cell population (CD4+, CD8+ or total lymphocytes) by flow cytometry (NCT02720107)
Timeframe: Baseline up to approximately 48 months

Interventionpercentage of parent population (Least Squares Mean)
B cellsMonocytesNatural Killer cells
Fingolimod-7.242.328.0

Change in T Cells Status (Decrease or Increase) at Month 48 (FAS)

Aim of trial was to was to show reduction of CD4+ and CD8+ naïve T cells (CCR7+CD45RA+), central memory T cells (CCR7+CD45RA-), central memory Th17 cells (CD4+ CCR4+ and CCR6+), and an elevation of 2 types of effector memory T cells TEM (CCR7- CD45RA-) and TEMRA (CCR7- CD45RA+) in peripheral venous blood. Changes from baseline to month 48 in biomarkers were analyzed for all patients in the FAS. (NCT02720107)
Timeframe: Baseline up to approximately 48 months

Interventionpercentage of parent population (Least Squares Mean)
CD4+ Naïve T cellsCD4+Central memory T cellsCD4+ Effector memory T cellsCD8+ Naïve T cellsCD8+ Central memory T cellsCD8+ Effector memory T cellsTH17 central memory cells
Fingolimod-23.7-1.222.2-37.2-1.6-12.9-0.6

Number of Particpants With Adverse Events as a Measure of Safety and Tolerability

Number of particpants with Adverse events as a measure of safety and tolerability (NCT01757691)
Timeframe: Weeks 0, 4, 8, 12, 18, 24, 36, 48, 60

InterventionParticipants (Number)
Adverse Events (AE)DeathNon -Fatal Seriuos Aderse Event (SAE)
Fingolimod 0.5mg/Daily100

Change in Patient-reported Depression

The Beck Depression Inventory (BDI-I) scale was used to measure this outcome. The scale consists of 21 items to assess the intensity of depression in clinical and normal patients. Each item is a list of four statements arranged in increasing severity about a particular symptom of depression. Each item was scored from 0 - 3. If more than one score was provided for an item, the maximum score was considered the item score. The total score was calculated as the sum of all individual items and then compared to a key to determine the depression's severity. The standard key ranges were: 0 - 9 indicated minimal depression; 10 - 18 indicated mild depression; 19 - 29 indicated moderate depression and 30 - 63 indicated severe depression. Higher total scores indicate more severe depressive symptoms. (NCT01534182)
Timeframe: Baseline, 6 months

Interventionscores on a scale (Mean)
Fingolimod-2.88
Standard Disease Modifying Therapy (DMT)-1.86

Change in Patient-reported Treatment Satisfaction

The Treatment Satisfaction Questionnaire for Medication (TSQM) contains 14 items assessing the following 4 domains: effectiveness (items 1 - 3), side effects (items 4 - 8), convenience (items 9 - 11) and global satisfaction (items 12 - 14). The primary outcome was measured on the global satisfaction domain. Item 12 scored as 1 (not at all confident) to 5 (extremely confident); item 13 scored as 1 (not at all certain) to 5 (extremely certain); and item 14 scored as 1 (extremely dissatisfied) to 7 (extremely satisfied). Responses to items were summed and transformed: specifically, TSQM v 1.4 domain scale scores were computed by adding the items loading on each domain. The lowest possible score was subtracted from the composite score and divided by the greatest possible score range. This provided a transformed score between 0 and 1 that was then multiplied by 100. The final transformed score ranges from 0 to 100, with higher scores indicating better treatment satisfaction. (NCT01534182)
Timeframe: Baseline, 6 months

Interventionscores on a scale (Mean)
Fingolimod22.69
Standard Disease Modifying Therapy (DMT)13.92

Change in Patient-reported Health-related Quality-of-life Using the Short Form Health Survey v2 Acute (SF-36 v2 Acute)

The SF-36 is a health-related quality of life instrument used in numerous disease states, including MS (Brazier et al 1992). It is a self-administered survey that measures 8 domains of health including: physical functioning, role limitations due to physical health, bodily pain, general health perceptions, vitality, social functioning, role limitations due to emotional problems and general mental health. Two summary scale scores can be calculated: the Physical Component Summary (PCS) and the Mental Component Summary (MCS). Each domain was scored by adding the individual items from the domain and transforming the resulting scores into a 0 to 100 scale with higher scores indicating better health status or functioning. (NCT01534182)
Timeframe: Baseline, 6 months

,
Interventionscores on scale (Mean)
Physical functioning (n=225,61)Role limitations due to physical health (n=226,61)Bodily pain (n=225,61)General health (n=226,61)Vitality (n=226,61)Social functioning (n=226,61)Role limit. due to emotional problems (n=224,61)Mental health (n=226,61)Physical component summary (n=222,61)Mental component summary (n=222,61)
Fingolimod3.626.505.244.287.725.597.185.601.513.16
Standard Disease Modifying Therapy (DMT)1.394.300.934.431.951.645.332.790.681.68

Changes in Patient-reported Effectiveness, Side Effects and Convenience

TSQM v 1.4 domains for effectiveness, side effects and convenience were used to evaluate this outcome. The effectiveness domain for items 1 - 3 was scored as: 1 (extremely dissatisfied) to 7 (extremely satisfied). For the side effects domain, item 4 scored as 0(no) or 1(yes); item 5 scored as 1 (extremely bothersome) to 5 (not at all bothersome); and items 6 - 8 scored as 1 (a great deal) to 5 (not at all). For the convenience domain, items 9 and 10 scored as 1(extremely difficult) to 7 (extremely easy), and item 11 scored as 1 (extremely inconvenient) to 7 (extremely convenient). For each domain, scale scores were computed by adding the items loading on each domain. The lowest possible score was subtracted from the composite score and divided by the greatest possible score range. This provided a transformed score between 0 and 1 that was then multiplied by 100. The final transformed score ranges from 0 to 100, with higher scores indicating better treatment satisfaction. (NCT01534182)
Timeframe: Baseline, 6 months

,
Interventionscores on a scale (Mean)
EffectivenessSide effectsConvenience
Fingolimod21.5726.7525.38
Standard Disease Modifying Therapy (DMT)11.5613.0710.57

Number of Patients Who Experienced Adverse Events, Serious Adverse Events and Death

Participants were monitored for adverse events, serious adverse events and death throughout the study. (NCT01534182)
Timeframe: 6 months

,
InterventionParticipants (Number)
Adverse events (serious and non-serious)Serious adverse eventsDeaths
Fingolimod7320
Standard Disease Modifying Therapy (DMT)2600

Absolute Peripheral Blood Cytokine and Chemokine Measurements in Healthy Controls and RRMS Patients

"Peripheral blood chemokine cytokine levels measured by flow cytometry at baseline and between visits during fingolimod treatment.~Absolute counts of the cells were calculated according to the absolute lymphocyte counts and the percentages of cells. This allowed for a clear determination of cell counts and thus increased the reliability of the results." (NCT02373098)
Timeframe: Baseline, Month 3, Month 6

,,,
Interventionabsolute cell count (Mean)
CD3CD4 (Lymphogate)CD3CD8 (Lymphogate)CD3CD4 (CD3 gate)CD3CD8 (CD3 gate)CD4+IFNg+ (in CD4+)CD4+IL17+ (in CD4+)CD8+IFNg+ (in CD8+)CD8+IL17+ (in CD8+)IFNg+ (in CD4+CD25+)IL17+ (in CD4+CD25+)CD4+IL10+ (in CD4+)CD4+IL4+ (in CD4+)CD4-CD8-IL4+ (in CD4-CD8-)CD8+IL10+ (in CD8+)IL10+ (in CD4+CD25+)IL4+ (in CD4+CD25+)CD4+TNFa+ (in CD4+)CD4+IL9+ (in CD4+)CD8+TNFa+ (in CD8+)CD8+IL9+ (in CD8+)TNFa+ (in CD4+CD25+)IL9+ (in CD4+CD25+)
Healthy Controls41.1923.2357.3835.117.421.2316.451.717.12.351.641.441.310.522.571.8846.860.22350.1452.825.06
Visit 144.9122.9162.9931.23.611.275.474.21.982.060.720.962.620.311.810.523.720.5411.780.923.290.59
Visit 213.6432.9323.2659.527.851.265.739.227.071.851.511.451.770.252.881.2229.450.420.930.1330.390.72
Visit 313.932.222459.6613.972.0214.72.239.312.72.142.322.640.674.031.4949.960.4741.530.1440.41.3

Baseline Flow Cytometry Analyses for Blood Cytokines and Chemokines in Healthy Controls and RRMS Patients

baseline peripheral blood flow cytometric analysis in study participants (NCT02373098)
Timeframe: Baseline

,
Interventionabsolute cell count (Mean)
CD3 absCD19 absNK absNKT absHi CD16CD56 absCD4CD25 (CD3 gate)Hi CD4CD25 (CD3 gate)
FTY7201610.94327.63623.57104.3620.4427.083.33
Healthy Controls2048.14311.63281.33135.8626.6426.082.83

Baseline Serum Cytokine and Chemokine Levels of Healthy Controls and RRMS Patients - ELISA

Blood samples were taken at baseline and measurements were performed before treatment of fingolimod. (NCT02373098)
Timeframe: Baseline

,
Interventionpg/ml (Mean)
CCL5 (RANTES)IL-17ACXCL13IL6IL8IL13IL23VLA4CXCL10=IP-10 (CXCR3 ligand)CCL2=MCP-1IL4TNF alphaIL22
FTY720404.311.06139.6866.533.570.33.040.5818.1862.892.361.0622.41
Healthy Controls212.261.15110.80.883.250.491.291.1716.0952.882.021.122.36

Percent Blood Cytokines and Chemokines Via Flow Cytometry Analyses of Healthy Controls and RRMS Patients at Baseline

Baseline peripheral blood flow cytometric analyses in study participants evaluated by flow cytometry analysis. (NCT02373098)
Timeframe: Baseline

,
InterventionPercent of cells (Mean)
CD3 %CD19 %NK %NKT %Hi CD16CD56 %CD3CD4 % (Lymphogate)CD3CD8 % (Lymphogate)CD3CD4 % (CD3 gate)CD3CD8 % (CD3 gate)CD4+IFNg+ (in CD4+)CD4+IL17+ (in CD4+)CD8+IFNg+ (in CD8+)CD8+IL17+ (in CD8+)IFNg+ (in CD4+CD25+)IL17+ (in CD4+CD25+)CD4+IL10+ (in CD4+)CD4+IL4+ (in CD4+)CD4-CD8-IL4+ (in CD4-CD8-)CD8+IL10+ (in CD8+)CD8+IL4+ % (in CD8+)IL10+ (in CD4+CD25+)IL4+ (in CD4+CD25+)CD4+TNFa+ (in CD4+)CD4+IL9+ (in CD4+)CD8+TNFa+ (in CD8+)CD8+IL9+ (in CD8+)TNFa+ (in CD4+CD25+)IL9+ (in CD4+CD25+)
FTY72071.9213.7210.175.190.9344.9122.9162.9931.203.611.275.474.201.982.060.720.962.620.311.871.810.5023.720.5411.780.9023.290.59
Healthy Controls76.5211.5711.045.651.0541.1923.2357.3835.117.421.2316.451.717.12.351.641.441.310.525.642.571.8846.860.22350.1452.825.06

Percent of Peripheral Blood Cytokine and Chemokine Measurements in Healthy Controls and RRMS Patients

Peripheral blood chemokine cytokine levels measured by flow cytometry in healthy controls at baseline and in RRMS patients between visits during fingolimod treatment (NCT02373098)
Timeframe: Baseline, Month 3, Month 6

,,,
Interventionpercent of cell count (Mean)
CD3 %CD19 %NK %NKT %Hi CD16CD56 %CD8+IL4+ % (in CD8+)
Healthy Controls76.5211.570.4275.651.055.64
Visit 171.9213.7210.175.190.931.87
Visit 257.616.1334.4815.555.293.13
Visit 359.295.1433.9117.666.201.91

Peripheral Blood Cytokine and Chemokine Measurements in Healthy Controls and RRMS Patients

Peripheral blood chemokine cytokine levels measured by flow cytometry during fingolimod treatment in healthy controls at baseline and in RRMS patients between visits (NCT02373098)
Timeframe: Baseline, Month 3, Month 6

,,,
Interventionpg/ml (Mean)
CD3 absCD19 absNK absNKT absHi CD16CD56 absCD4CD25 (CD3 gate)Hi CD4CD25 (CD3 gate)
Healthy Controls2048.14311.63281.33135.8626.6426.082.83
Visit 11610.94327.63623.57104.3620.4427.033.33
Visit 2339.7432.9203.6688.4529.359.242.03
Visit 3314.7526.56181.9788.9830.049.221.77

Serum Cytokine and Chemokine Levels inRRMS Patients Between Visits

Change of serum cytokine and chemokine levels measured by ELISA in RRMS patients treated with fingolimod between visits (NCT02373098)
Timeframe: Baseline, month 3, month 6

,,
Interventionpg/ml (Mean)
CXCL13=BLCIL6IL8IL13CCL5= RANTESIL23VLA4CXCL10=IP-10 (CXCR3 ligand)CCL2=MCP-1IL4IL17ATNFIL22
Visit 1139.6866.533.570.3404.313.040.5818.1862.892.361.061.0622.41
Visit 2105.4249.983.840.35384.122.010.7618.4674.381.941.091.2622.45
Visit 3105.86149.624.290.48480.342.150.8220.5492.732.121.041.1222.99

ALSFRS-R Total Score at Weeks 0, 2, 4 and 8

The ALSFRS-R is a quickly administered (5 minutes) ordinal rating scale (ratings 0-4) used to determine subjects' assessment of their capability and independence in 12 functional activities. All 12 activities are relevant in ALS. Initial validity was established by documenting that in ALS patients, change in ALSFRS-R scores correlated with change in strength over time, was closely associated with quality of life measures, and predicted survival. (NCT01786174)
Timeframe: Week 0, Week 2, Week 4 and Week 8

,
Interventionscores on a scale (Mean)
Week 0Week 2Week 4Week 8
Gilenya (Fingolimod)38.6038.1538.0336.74
Placebo38.6038.2937.8837.10

Change in Slow Vital Capacity Score (SVC)

The vital capacity (VC) (percent of predicted normal) was determined using the slow VC method. Vital Capacity is the maximum amount of air a person can expel from the lungs after a maximum inhalation. A subject's VC depends on their age, sex and height. The value is recorded as a percent of predicted normal. (NCT01786174)
Timeframe: Week 0, Week 2, Week 4 and Week 8

,
InterventionPercentage of predicted max value (Mean)
Week 0Week 2Week 4Week 8
Gilenya (Fingolimod)88.2888.5486.5186.02
Placebo88.2888.5486.7087.59

Forced Expiratory Volume in 1 Second (FEV1) / Slow Vital Capacity (SVC) Ratio

"Forced Expiratory Volume (FEV1): Forced Expiratory Volume (FEV1) is the amount of air which can be forcibly exhaled from the lungs in the first second of a forced exhalation.~Slow Vital Capacity (SVC): Vital Capacity is the maximum amount of air a person can expel from the lungs after a maximum inhalation. A subject's VC depends on their age, sex and height. The value is recorded as a percent of predicted normal." (NCT01786174)
Timeframe: Screening, Week 0, Week 2, and Week 4

,
InterventionPercentage of predicted max value (Mean)
ScreeningWeek 0Week 2Week 4
Gilenya (Fingolimod)77.3974.3675.4676.34
Placebo77.3974.3673.1471.49

Forced Expiratory Volume in 1 Second (FEV1)

Forced Expiratory Volume (FEV1): Forced Expiratory Volume (FEV1) is the amount of air which can be forcibly exhaled from the lungs in the first second of a forced exhalation. (NCT01786174)
Timeframe: Screening, Week 0, Week 2, and Week 4

,
InterventionPercentage of predicted max value (Mean)
ScreeningWeek 0Week 2Week 4
Gilenya (Fingolimod)84.7081.9381.1980.38
Placebo84.7081.9380.3078.16

Lymphocyte (T-Cell) Subset Trajectories

Gilenya (fingolimod) has been shown to successfully reduce circulating lymphocytes (a type of white blood cell) by blocking their egress (exit) from the lymph nodes. A secondary objective of the study is to quantify the effect of the treatment on circulating lymphocyte populations in patients with ALS. (NCT01786174)
Timeframe: Week 0, Week 2, and Week 4

,
Intervention10^3/uL (Mean)
Week 0Week 2Week 4
Gilenya (Fingolimod)1.7510.5800.499
Placebo1.7511.7321.822

Cognition Change - BACS

The Brief Assessment of Cognition in Schizophrenia (BACS) is a battery specifically designed to measure treatment-related changes in cognition by utilizing 6 tasks, and has alternate forms, thus minimizing practice effects. Each task generates a raw score (with a higher score indicating better performance): verbal memory 0-75; digit sequencing 0-28; token motor task 0-100; semantic&letter fluency 0-148; symbol coding 0-110; and tower of London 0-22. The raw scores are used to generate a composite score that is calculated by summing t-scores derived by comparisons with a normative sample of 404 healthy controls. The six brief assessments' t-scores, are summed, and averaged to provide a composite t-score. The composite score min and max are between -43 and 100. A higher score indicating better cognitive performance. (NCT01779700)
Timeframe: Baseline, 4 weeks, 8 weeks

,
Interventionscore on a scale (Mean)
BACS Composite Score - BaselineBACS Composite Score - 4 weeksBACS Composite Score - 8 weeks
Fingolimod25.1727.6731.13
Placebo30.9532.1033.50

Cognition Change - Trails B

The Trail Making Test-Part B (Trails B) is a measure of visual attention and task switching. The task requires a subject to 'connect-the-dots' of 25 consecutive targets on a sheet of paper. In Part B version the subject alternates between numbers and letters (1, A, 2, B, etc.) The goal of the test is for the subject is to finish part B as quickly as possible, the time taken to complete the test is used as the primary performance metric. The score is the number of seconds it took to complete the test. (NCT01779700)
Timeframe: Baseline, 4 weeks, 8 weeks

,
Interventionseconds (Mean)
Trails B - BaselineTrails B - 4 weeksTrails B - 8 weeks
Fingolimod108.0097.8884.53
Placebo123.64100.60102.33

Levels of Lymphocyte

To determine the safety of fingolimod, as measured by the absolute lymphocyte count (NCT01779700)
Timeframe: Baseline, 4 weeks, 8 weeks

,
Intervention10^3 lymphocytes/uL (Mean)
Absolute lymphocyte count - BaselineAbsolute lymphocyte count - 4 weeksAbsolute lymphocyte count - 8 weeks
Fingolimod1.990.440.49
Placebo1.971.942.00

Negative Symptom Change - PANSS

The Positive and Negative Syndrome Scale (PANSS) is a semi-structured interview, containing 30 items that assess symptoms of psychotic disorders including positive, negative, and general psychopathology symptoms. Positive symptoms are rated on 7 items, negative symptoms are rated on 7 items, and general psychopathology on 16 items. Scores for each item range from 1=absent to 7=extreme. Positive, negative, and general psychopathology symptoms can each respectively render total scores. Positive total scores ranging from 7-49, negative total scores ranging from 7-49, and general psychopathology scores ranging from 16-112. When all items are summed together a total score is generated. Total scores for all items range from 30-210, a lower score reflecting fewer symptoms. (NCT01779700)
Timeframe: Baseline, 4 weeks, 8 weeks

,
Interventionscore on a scale (Mean)
PANSS Negative Score - BaselinePANSS Negative Score - 4 weeksPANSS Negative Score - 8 weeks
Fingolimod15.0013.8915.44
Placebo16.5914.8015.06

Plasma Cytokines Levels - IFNgamma

To assess IFNgamma plasma cytokines levels changes in participants taking fingolimod versus placebo (NCT01779700)
Timeframe: Baseline, 4 weeks, 8 weeks

,
Interventionpg/ml (Mean)
IFNgamma - BaselineIFNgamma - 4 weeksIFNgamma - 8 weeks
Fingolimod5.324.614.32
Placebo8.324.955.62

Plasma Cytokines Levels - IL-10

To assess IL-10 plasma cytokines levels changes in participants taking fingolimod versus placebo (NCT01779700)
Timeframe: Baseline, 4 weeks, 8 weeks

,
Interventionpg/ml (Mean)
IL10 - BaselineIL10 - 4 weeksIL10 - 8 weeks
Fingolimod16.1115.5016.28
Placebo11.949.2310.42

Plasma Cytokines Levels - IL-17A

To assess IL-17A plasma cytokines levels changes in participants taking fingolimod versus placebo (NCT01779700)
Timeframe: Baseline, 4 weeks, 8 weeks

,
Interventionpg/ml (Mean)
IL17A - BaselineIL17A - 4 weeksIL17A - 8 weeks
Fingolimod3.092.582.58
Placebo3.722.432.69

Plasma Cytokines Levels - IL-1BETA

To assess IL-1BETA plasma cytokines levels changes in participants taking fingolimod versus placebo (NCT01779700)
Timeframe: Baseline, 4 weeks, 8 weeks

,
Interventionpg/ml (Mean)
IL1BETA - BaselineIL1BETA - 4 weeksIL1BETA - 8 weeks
Fingolimod0.870.810.82
Placebo0.690.720.74

Plasma Cytokines Levels - IL-2

To assess IL-2 plasma cytokines levels changes in participants taking fingolimod versus placebo (NCT01779700)
Timeframe: Baseline, 4 weeks, 8 weeks

,
Interventionpg/ml (Mean)
IL2 - BaselineIL2 - 4 weeksIL2 - 8 weeks
Fingolimod1.741.961.77
Placebo1.271.201.20

Plasma Cytokines Levels - IL-4

To assess IL-4 plasma cytokines levels changes in participants taking fingolimod versus placebo (NCT01779700)
Timeframe: Baseline, 4 weeks, 8 weeks

,
Interventionpg/ml (Mean)
IL4 - BaselineIL4 - 4 weeksIL4 - 8 weeks
Fingolimod30.9927.4024.20
Placebo30.7431.3432.16

Plasma Cytokines Levels - IL-6

To assess IL-6 plasma cytokines levels changes in participants taking fingolimod versus placebo (NCT01779700)
Timeframe: Baseline, 4 weeks, 8 weeks

,
Interventionpg/ml (Mean)
IL6 - BaselineIL6 - 4 weeksIL6 - 8 weeks
Fingolimod2.152.222.16
Placebo1.901.811.96

Plasma Cytokines Levels - IL-8

To assess IL-8 plasma cytokines levels changes in participants taking fingolimod versus placebo (NCT01779700)
Timeframe: Baseline, 4 weeks, 8 weeks

,
Interventionpg/ml (Mean)
IL8 - BaselineIL8 - 4 weeksIL8 - 8 weeks
Fingolimod4.293.814.18
Placebo3.833.693.69

Plasma Cytokines Levels - TNFa

To assess TNFa plasma cytokines levels changes in participants taking fingolimod versus placebo (NCT01779700)
Timeframe: Baseline, 4 weeks, 8 weeks

,
Interventionpg/ml (Mean)
TNFa - BaselineTNFa - 4 weeksTNFa - 8 weeks
Fingolimod2.112.022.00
Placebo1.761.861.61

Positive Symptom Change - PANSS

The Positive and Negative Syndrome Scale (PANSS) is a semi-structured interview, containing 30 items that assess symptoms of psychotic disorders including positive, negative, and general psychopathology symptoms. Positive symptoms are rated on 7 items, negative symptoms are rated on 7 items, and general psychopathology on 16 items. Scores for each item range from 1=absent to 7=extreme. Positive, negative, and general psychopathology symptoms can each respectively render total scores. Positive total scores ranging from 7-49, negative total scores ranging from 7-49, and general psychopathology scores ranging from 16-112. When all items are summed together a total score is generated. Total scores for all items range from 30-210, a lower score reflecting fewer symptoms. (NCT01779700)
Timeframe: Baseline, 4 weeks, 8 weeks

,
Interventionscore on a scale (Mean)
PANSS Positive Score - BaselinePANSS Positive Score - 4 weeksPANSS Positive Score - 8 weeks
Fingolimod10.8311.8910.06
Placebo13.6414.0013.06

QTcB Change

To determine the safety of fingolimod, as measured by the electrocardiogram (ECG) QT interval corrected by Bazett's (QTcB) value. (NCT01779700)
Timeframe: Screening, Day 0, Day 7, Day 14, Day 21, Day 28, Day 35, Day 42, Day 49, Day 56, Day 84, Day 112

,
Interventionms (Mean)
QTcB - ScreeningQTcB - Day 0QTcB - Day 7QTcB - Day 14QTcB - Day 21QTcB - Day 28QTcB - Day 35QTcB - Day 42QTcB - Day 49QTcB - Day 56QTcB - Day 84QTcB - Day 112
Fingolimod416.50418.39415.50418.28419.67424.06426.18425.53420.79420.67421.50419.67
Placebo416.41423.27417.00420.15412.00415.74419.00414.33413.53418.41420.68418.58

Symptom Changes - PANSS Total Score

The Positive and Negative Syndrome Scale (PANSS) is a semi-structured interview, containing 30 items that assess symptoms of psychotic disorders including positive, negative, and general psychopathology symptoms. Positive symptoms are rated on 7 items, negative symptoms are rated on 7 items, and general psychopathology on 16 items. Scores for each item range from 1=absent to 7=extreme. Positive, negative, and general psychopathology symptoms can each respectively render total scores. Positive total scores ranging from 7-49, negative total scores ranging from 7-49, and general psychopathology scores ranging from 16-112. When all items are summed together a total score is generated. Total scores for all items range from 30-210, a lower score reflecting fewer symptoms. (NCT01779700)
Timeframe: Baseline, 4 weeks, 8 weeks

,
Interventionscore on a scale (Mean)
PANSS Total Score - BaselinePANSS Total Score - 4 weeksPANSS Total Score - 8 weeks
Fingolimod48.8949.7849.06
Placebo56.5053.6054.94

Verbal Memory - BACS

The Brief Assessment of Cognition in Schizophrenia (BACS) is a battery specifically designed to measure treatment-related changes in cognition. The BACS utilizes 6 tasks, and has alternate forms, thus minimizing practice effects. Each task generates a raw score (with a higher score indicating better performance): verbal memory 0-75; digit sequencing 0-28; token motor task 0-100; semantic&letter fluency 0-148; symbol coding 0-110; and tower of London 0-22. The raw scores are used to generate a composite score that is calculated by summing t-scores derived by comparisons with a normative sample of 404 healthy controls. The six brief assessments' t-scores, are summed, and averaged to provide a composite t-score. The composite score min and max are between -43 and 100. A higher score indicating better cognitive performance. (NCT01779700)
Timeframe: Baseline, 4 weeks, 8 weeks

,
Interventionscore on a scale (Mean)
BACS Verbal Memory - BaselineBACS Verbal Memory - 4 weeksBACS Verbal Memory - 8 weeks
Fingolimod30.8933.5034.69
Placebo37.4535.4536.11

Number of Participants With Bradyarrhythmic Electrocardiogram (ECG) Events

The number of participants with bradyarrhythmic electrocardiogram (ECG) events was assessed. Bradyarrhythmic ECG events are defined as QTc Fridericia time > 450 ms for males and > 470 ms for females. (NCT01585298)
Timeframe: up to day 7

InterventionParticipants (Number)
Fingolimod26

Number of Patients With Heart Rate Below 45 Beats Per Minute (BPM)

Number of patients with heart rate below 45 beats bpm in ECG during first dose observation (NCT01585298)
Timeframe: baseline during 6 hour monitoring post dose

InterventionParticipants (Number)
Fingolimod63

Number of Participants With Prolonged QTc Interval (Friderica)

"Number of patients with conduction abnormalities such as QT prolongation, first degree AV block during treatment initiation.~The QT interval is a period between the activation and the regeneration of ventricular contraction. A prolonged QT interval can be a potential marker of cardiac arrhythmias.~Two patients of the safety analysis set discontinued the study without having received treatment, but safety information was collected on these two patients." (NCT01585298)
Timeframe: baseline post-dose

InterventionParticipants (Number)
Female, QTcFridericia Interval > 470 msMale, QTcF Interval > 450 ms
Fingolimod76

Number of Patients With Cardiac Adverse Events

The number of participants with the occurrence of subsequent cardiac adverse events (AEs) and serious cardiac AEs during study was assessed. Cardiac events were defined as the following Medical Dictionary for Regulatory Activities (MedDRA) preferred terms: angina pectoris, chest discomfort, dizziness, dyspnoea, dyspnoea exertional, fatigue, palpitations, syncope, vertigo, vertigo positional and vision blurred. (NCT01585298)
Timeframe: 7 days

InterventionParticipants (Number)
Cardiac eventsSerious cardiac AEs
Fingolimod4899

Participants With 2nd or 3rd Degree Atrioventricular (AV) Block

AV Blocks/Heart block is an abnormal heart rhythm where the heart beats too slowly; the electrical signals that tell the heart to contract are partially intermittent (Type 2:1) or slowed (1st and 2nd degree) or blocked (3rd degree) between the upper chambers (atria) and the lower chambers (ventricles). In 2nd degree AV Blocks, electrical impulses are intermittent (type 2:1) or delayed w/ each subsequent heartbeat (Mobitz type I) until a beat fails to reach the ventricles entirely. This type of block often is physiologic and observed in a highly relaxed state & during sleep. In 2nd degree AV Blocks type II, the atria electrical impulses are unable to reach the ventricles, a more serious condition. In 3rd degree AV Blocks (complete heart block), none of the electrical impulses reach either the atria or the ventricles. Patients can experience simultaneously both types of 2nd or 3rd degree AV Blocks without any symptoms. (NCT01585298)
Timeframe: baseline, during 6 hour monitoring post first dose observation

InterventionParticipants (Number)
Any AV block ll degree or higherAV block ll degree: Mobitz type I degreeAV block II degree: Mobitz type II degreeAV block II degree: 2:1AV block III degree
Fingolimod1201170431

Time to First Confirmed Worsening on the Adjusted Inflammatory Neuropathy Cause and Treatment (INCAT) Disability Scale

Confirmed worsening in CIDP was measured by the adjusted INCAT Disability Scale. The adjusted INCAT disability scale measures arm disability and leg disability. For arm disability the scale ranges from 0 (no upper limb problems) to 5 (inability to use either arm for any purposeful movement). The leg disability scale ranges from 0 (walking not affected) to 5 (restricted to wheelchair, unable to stand and walk a few steps with help). The total adjusted INCAT disability score is calculated by the sum of the arm and leg disability scores where the total score ranges from 0 to 10. A confirmed worsening was defined as an increase by 1 or more points on the adjusted INCAT disability scale from the value at baseline. (NCT01625182)
Timeframe: Month 12

InterventionDays (Median)
Fingolimod (FTY720)721.0
Placebo540.0

Change From Baseline for Grip Strength, Dominant Hand

Grip strength measurements were done using a vigorimeter. With this device, the pressure in the bulb exercised by the participant was registered on a manometer via a rubber junction tube. Both the dominant and non-dominant hands were tested. A negative change from baseline indicates deterioration. (NCT01625182)
Timeframe: baseline, Month 6, Month 12

,
InterventionkPa (Least Squares Mean)
Month 6Month 12
Fingolimod (FTY720)-2.6-0.8
Placebo-3.8-3.9

Change From Baseline for Grip Strength, Non-dominant Hand

Grip strength measurements were done using a vigorimeter. With this device, the pressure in the bulb exercised by the participant was registered on a manometer via a rubber junction tube. Both the dominant and non-dominant hands were tested. A negative change from baseline indicates deterioration. (NCT01625182)
Timeframe: baseline, Month 6, Month 12

,
InterventionkPa (Least Squares Mean)
Month 6Month 12
Fingolimod (FTY720)-2.7-1.2
Placebo-6.1-5.0

Change From Baseline for Rasch-Built Linearly Weighted Overall Disability Scale (R-ODS)

This questionnaire was constructed using the patients' perception of their ability to perform daily and social activities. The questionnaire comprises 24 items ranging from ability to read a book or newspaper (as the easiest item to accomplish) to ability to run (most difficult item to accomplish). The obtained raw summed score was translated subsequently to a convenient centile metric score ranging from 0 (most severe disability) to 100 (no disability at all). A higher score indicated a better health status. A negative change from baseline indicates deterioration. (NCT01625182)
Timeframe: baseline, Month 6, Month 12

,
Interventionscore on a scale (Least Squares Mean)
Month 6Month 12
Fingolimod (FTY720)-6.4-5.7
Placebo-5.5-5.1

Number of Active (New or Newly Enlarging) T2 Lesions During the 24 Weeks After the Last Natalizumab Infusion (Baseline)

Lesions will be measured by MRIs and the number of active (new or newly enlarging) T2 lesions will be calculated for 24 weeks from baseline. (NCT01499667)
Timeframe: Baseline up to 24 weeks

InterventionCount of Active T2 Lesions (Mean)
8-week Washout + Fingolimod (FTY720)3.2
12-week Washout + Fingolimod (FTY720)4.4
16-week Washout + Fingolimod (FTY720)7.7

Number of Active (New or Newly Enlarging) T2 Lesions During the First 8 Weeks of Fingolimod Treatment

Lesions were measured by MRIs and the number of active (new or newly enlarging) T2 lesions was calculated for first 8 weeks of fingolimod treatment. (NCT01499667)
Timeframe: Number of active T2 lesions during 8 wks of fingolimod treatment

InterventionCount of Active T2 Lesions (Mean)
8-week Washout + Fingolimod (FTY720)1.5
12-week Washout + Fingolimod (FTY720)2.1
16-week Washout + Fingolimod (FTY720)4.2

Number of Active (New or Newly Enlarging) T2 Lesions From the Last Natalizumab Infusion (Baseline) Through 8 Weeks of Fingolimod Treatment

Active lesions were measured on brain MRI scans, performed at week 8, compared to the prior scan. The primary variable was analyzed by fitting a negative binomial regression model adjusted for washout group. (NCT01499667)
Timeframe: Number of active T2 lesions from last natalizumab dose through 8 weeks of fingolimod treatment

InterventionCount of Active T2 Lesions (Mean)
8-week Washout + Fingolimod (FTY720)2.1
12-week Washout + Fingolimod (FTY720)1.7
16-week Washout + Fingolimod (FTY720)8.2

Number of Active (New or Newly Enlarging) T2 Lesions From the Last Natalizumab Infusion (Baseline) up to the Initiation of Fingolimod Treatment

Lesions were measured by MRIs and the number of active (new or newly enlarging) T2 lesions was calculated from baseline to beginning of treatment. (NCT01499667)
Timeframe: 8, 12 and 16 weeks (number of active T2 lesions during the washout period only)

InterventionCount of active T2 lesions (Mean)
8-week Washout + Fingolimod (FTY720)0.4
12-week Washout + Fingolimod (FTY720)2.1
16-week Washout + Fingolimod (FTY720)3.6

Change From Baseline in Expanded Disability Status Scale (EDSS) by Washout Group

Kurtzke's Expanded Disability Status Scale (EDSS) measures the changes in neurologic impairment, either chronic (progression over time), or acute (MS relapses). The EDSS steps range from 0 (normal) to 10 (death due to MS). Relapse severity is assessed based on severity of neurologic impairment as evaluated using the EDSS. (NCT01499667)
Timeframe: Baseline to week 16 and week 32

,,
InterventionUnits on a scale (Mean)
Week 16 (n=40, 33, 39)Week 32 (n= 40,30,39)
12-week Washout + Fingolimod (FTY720)-0.03-0.13
16-week Washout + Fingolimod (FTY720)0.230.08
8-week Washout + Fingolimod (FTY720)0.110.11

Cumulative Number of Gadolinium-enhancing T1 Lesions From the Last Natalizumab Infusion

Gadolinium-enhancing lesions will be measured on post-contrast T1-weighted brain MRI scans (NCT01499667)
Timeframe: 8 weeks and 24 weeks

,,
InterventionNumber of Gd enhanced T1 Lesions (Mean)
Week 8 (n=1,1,0)Week 24 (n=10,12,21)
12-week Washout + Fingolimod (FTY720)2.03.4
16-week Washout + Fingolimod (FTY720)NA3.6
8-week Washout + Fingolimod (FTY720)25.06.3

Number of Participants With Adverse Events (AE), Serious Adverse Events (SAE) and Death During Fingolimod Treatment

Adverse events were summarized by the number of patients having any adverse event overall. (NCT01499667)
Timeframe: Baseline to maximum of 16 weeks

,,
Interventionparticipants (Number)
Any Adverse EventsSerious Adverse EventsDeath
12-week Washout + Fingolimod (FTY720)2050
16-week Washout + Fingolimod (FTY720)2830
8-week Washout + Fingolimod (FTY720)3520

Number of Participants With Adverse Events (AE), Serious Adverse Events (SAE) and Death During Washout Period

Adverse events were summarized by the number of patients having any adverse event overall. (NCT01499667)
Timeframe: Baseline to maximum of 16 weeks

,,
Interventionparticipants (Number)
Any Adverse EventsSerious Adverse EventsDeath
12-week Washout + Fingolimod (FTY720)1200
16-week Washout + Fingolimod (FTY720)2510
8-week Washout + Fingolimod (FTY720)1300

Annual Relapse Rate (ARR)

ARR = 365 days * number of relapses / total days taking the study medication. (NCT01498887)
Timeframe: 12 months

InterventionRelapses per year (Mean)
Naive or de Novo Participants0.290
Previously Treated With First-line DMTs Participants0.354

Change From Baseline in Cerebral Volume

Cerebral volume was assessed by magnetic resonance imaging (MRI). A negative change from baseline indicates improvement. (NCT01498887)
Timeframe: baseline, 12 months

InterventionPercent change (Mean)
Naive or de Novo Participants-0.595
Previously Treated With First-line DMTs Participants-0.387

Change From Baseline in Expanded Disability Status Scale (EDSS) Score

The EDSS is an ordinal clinical rating scale ranging from a total score of 0 (normal neurologic examination) to 10 (death due to MS) in half-point increments. A negative change from baseline indicates improvement. (NCT01498887)
Timeframe: baseline, 12 months

Interventionscore on a scale (Mean)
Naive or de Novo Participants0.000
Previously Treated With First-line DMTs Participants-0.077

Mean Number of T2 Active Lesions

The mean number of new or enlarged T2 active lesions was assessed by MRI. (NCT01498887)
Timeframe: 12 months

InterventionT2 lesions (Mean)
Naive or de Novo Participants2.0
Previously Treated With First-line DMTs Participants1.6

Percentage of Relapse-free Participants

Relapse-free participants were defined as participants who experienced no new neurological symptom or worsening of an existing one (relapses) during the 12-month treatment period with 0.5 mg fingolimod. (NCT01498887)
Timeframe: 12 months

InterventionPercent (Number)
Naive or de Novo Participants71.89
Previously Treated With First-line DMTs Participants66.67

Time to First Relapse

Time to first relapse was defined as the time from the first day of treatment to the first day of a new neurological symptom or worsening of an existing one. (NCT01498887)
Timeframe: first day of treatment to the first day of a new neurological symptom or worsening of an existing one, up to 12 months

Interventionmonths (Median)
Naive or de Novo ParticipantsNA
Previously Treated With First-line DMTs ParticipantsNA

Percentage of Participants With Mild, Moderate or Severe Relapse

The investigator classified a relapse as moderate-severe if oral or intravenous (IV) treatment (according to the local clinical practice) with steroids and/or hospitalization was needed. If neither oral nor IV treatment with steroids nor hospitalization was needed, the relapse was considered as mild. (NCT01498887)
Timeframe: 12 months

,
InterventionPercentage of participants (Number)
MildModerateSevere
Naive or de Novo Participants42.5557.450.00
Previously Treated With First-line DMTs Participants38.4656.415.13

Number of Participants Who Experienced Adverse Events, Serious Adverse Events and Death

"In this analysis patients with all (serious and non-serious) adverse events, and death were reported.~See Safety Section." (NCT01436643)
Timeframe: 21 weeks

,,,
InterventionParticipants (Number)
Any Adverse EventDeathSerious Adverse Event
Citalopram and Fingolimod1201
Fingolimod1501
Fluoxetine and Fingolimod1100
Venlafaxine and Fingolimod1201

Change From Baseline to Month 36 in Average Retinal Nerve Fiber Layer Thickness (RNFLT)

The primary endpoint was the change, i.e. the absolute difference, in average RNFL thickness from baseline to month 36 (or last values in case of missing data) in the Full Analysis Set (FAS). Average RNFL thickness was the average of valid measurements of the right and left eye and assessed by optical coherence tomography (OCT). (NCT01705236)
Timeframe: Baseline, month 36

Interventionmicrometer (Mean)
Fingolimod - Longitudinal Assessment-1.5

Change From Baseline to Month 12 and 24 in Average Retinal Nerve Fiber Layer Thickness (RNFLT)

Change from baseline in average RNFL thickness to months 12 and 24 (or last values in case of missing data) in the Full Analysis Set (FAS). Average RNFL thickness was the average of valid measurements of the right and left eye and assessed by optical coherence tomography (OCT). (NCT01705236)
Timeframe: Baseline, month 12, month 24

Interventionmicrometer (Mean)
Change from baseline to month 12Change from baseline to month 24
Fingolimod - Longitudinal Assessment-0.8-1.1

Change From Baseline to Month 12, 24 and 36 in Average Quadrant Retinal Nerve Fiber Layer Thickness (RNFLT)

Change from baseline in average quadrant RNFL thickness to months 12, 24 and 36 (or last values in case of missing data) in the Full Analysis Set (FAS). Average quadrant RNFL thickness was the average of valid measurements of the right and left eye and assessed by optical coherence tomography (OCT). Quadrant RNFL thickness were: Nasal-inferior; nasal-superior; temporal-inferior; temporal-superior. (NCT01705236)
Timeframe: Baseline, month 12, month 24, month 36

Interventionmicrometer (Mean)
Nasal-superior RNFL thickness: month 12Nasal-superior RNFL thickness: month 24Nasal-superior RNFL thickness: month 36Nasal-inferior RNFL thickness: month 12Nasal-inferior RNFL thickness: month 24Nasal-inferior RNFL thickness: month 36Temporal-inferior RNFL thickness: month 12Temporal-inferior RNFL thickness: month 24Temporal-inferior RNFL thickness: month 36Temporal-superior RNFL thickness: month 12Temporal-superior RNFL thickness: month 24Temporal-superior RNFL thickness: month 36
Fingolimod - Longitudinal Assessment0.5-0.4-0.7-1.2-1.6-2.1-1.2-1.6-2.3-0.5-0.4-1.1

Change From Baseline to Month 12, 24 and 36 in Ganglion Cell Inner Plexiform (GCIP)

"Change from baseline in GCIP to months 12, 24 and 36 (or last values in case of missing data) in the Full Analysis Set (FAS).~The change in Ganglion cell layer thickness (GCLT) had been defined as secondary endpoint in the protocol, but OCT measured the GCIP instead. This was done because both layers were not clearly separable by OCT. GCIP was calculated as mean of the inner sectors (nasal, superior, temporal, and inferior) and declared as usual parameter instead. This change was introduced prior to data base lock, but the derivation of GCIP was corrected after data base lock." (NCT01705236)
Timeframe: Baseline, month 12, month 24, month 36

Interventionmicrometer (Mean)
Change from baseline to month 12Change from baseline to month 24Change from baseline to month 36
Fingolimod - Longitudinal Assessment-0.49-0.42-0.46

Change From Baseline to Month 12, 24 and 36 in Total Macular Volume (TMV)

Change from baseline in TMV to months 12, 24 and 36 (or last values in case of missing data) in the Full Analysis Set (FAS). (NCT01705236)
Timeframe: 12, 24 and 36 months

InterventionCubic millimeter (Mean)
Change from baseline to month 12Change from baseline to month 24Change from baseline to month 36
Fingolimod - Longitudinal Assessment-0.03-0.04-0.06

Number of Participants With Adverse Events

Number of participants with adverse events and specifically macular edema. (NCT01705236)
Timeframe: 36 months

InterventionParticipants (Count of Participants)
No. of subjects with any AENo. of subjects with macular edema
Fingolimod - Longitudinal Assessment800

Mean Patient-reported Health-related Quality-of-life With Fingolimod (Short Form Health Survey: SF-36).

The SF-36v2 is a validated health-related quality of life instrument used in numerous disease states, including MS. It is a self-administered survey that measures 8 domains of health including: physical functioning, role limitations due to physical health, bodily pain, general health perceptions, vitality, social functioning, role limitations due to emotional problems and general mental health. Additionally, two summary scale scores can be calculated: the Physical Component Summary (PCS) and the Mental Component Summary (MCS). If half or more questions within a domain were answered, then a score was calculated for that domain. Otherwise, the patient score for that domain was set to missing. If the patient was missing any 1 of the 8 scale scores, then the physical and mental component scores were set to missing. An algorithm was used to create a score from 0 to 100 for each domain score and component score. A positive change from baseline indicates improvement. (NCT01578330)
Timeframe: Month 1

Interventionscore on scale (Mean)
Physical FunctionPhysical RolePainGeneral HealthVitalitySocial FunctionEmotional RoleMental Health
Fingolimod, FTY72059.753.661.747.828.961.848.431.5

Mean Patient-reported Health-related Quality-of-life With Fingolimod (Short Form Health Survey: SF-36).

The SF-36v2 is a validated health-related quality of life instrument used in numerous disease states, including MS. It is a self-administered survey that measures 8 domains of health including: physical functioning, role limitations due to physical health, bodily pain, general health perceptions, vitality, social functioning, role limitations due to emotional problems and general mental health. Additionally, two summary scale scores can be calculated: the Physical Component Summary (PCS) and the Mental Component Summary (MCS). If half or more questions within a domain were answered, then a score was calculated for that domain. Otherwise, the patient score for that domain was set to missing. If the patient was missing any 1 of the 8 scale scores, then the physical and mental component scores were set to missing. An algorithm was used to create a score from 0 to 100 for each domain score and component score. A positive change from baseline indicates improvement. (NCT01578330)
Timeframe: Month 12

Interventionscore on scale (Mean)
Physical Function (n=31)Physical Role (n=32)Pain (n=32)General Health (n=31)Vitality (n=32)Social Function (n=32)Emotional Role (n=31)Mental Health (n=32)
Fingolimod, FTY72059.546.965.950.833.165.852.736.3

Mean Patient-reported Health-related Quality-of-life With Fingolimod (Short Form Health Survey: SF-36).

The SF-36v2 is a validated health-related quality of life instrument used in numerous disease states, including MS. It is a self-administered survey that measures 8 domains of health including: physical functioning, role limitations due to physical health, bodily pain, general health perceptions, vitality, social functioning, role limitations due to emotional problems and general mental health. Additionally, two summary scale scores can be calculated: the Physical Component Summary (PCS) and the Mental Component Summary (MCS). If half or more questions within a domain were answered, then a score was calculated for that domain. Otherwise, the patient score for that domain was set to missing. If the patient was missing any 1 of the 8 scale scores, then the physical and mental component scores were set to missing. An algorithm was used to create a score from 0 to 100 for each domain score and component score. A positive change from baseline indicates improvement. (NCT01578330)
Timeframe: Month 6

Interventionscore on scale (Mean)
Physical Function (n=33)Physical Role (n=34)Pain (n=34)General Health (n=33)Vitality (n=34)Social Function (n=34)Emotional Role (n=34)Mental Health (n=34)
Fingolimod, FTY72057.947.862.648.535.260.654.836.5

Mean Patient-Reported Treatment Satisfaction Questionnaire for Medication Scores (TSQM-9)

The Treatment Satisfaction Questionnaire for Medication (TSQM-9) is a psychometric measure of a patient's satisfaction with medication. It consists of 3 subscales: effectiveness, convenience and global satisfaction. The scores were computed by adding items for each domain, i.e. 1 to 3 for effectiveness, 4 - 6 for convenience and 7 to 9 for global satisfaction. The lowest possible score (1 for each item and 3 for all 3 subscales) was subtracted from the composite score and divided by the greatest possible score range. The greatest range was (7-1) X 3 items = 18 for the effectiveness and convenience, and (5-1) x 3 items = 12 for global satisfaction. This provided a transformed score between 0 and 1 that was then multiplied by 100. A positive change from baseline indicates improvement. (NCT01578330)
Timeframe: Baseline and month 12

InterventionScores on a scale (Mean)
Baseline (n=34)Month 12 (n=32)
Fingolimod, FTY72032.044.7

Number (%) of Patients With AE of Special Interest Including Bradyarrhythmia, BP Increase, Liver Transaminase Elevations, Infections , Macula Oedema.

The incidence of events in special areas of safety interest (including bradyarrhythmias, BP increase, liver function, infections and macular oedema) were assessed by the nature and frequency of AE reporting. These areas of special interest have been identified and potential risks of fingolimod based on knowledge from clinical trials and post-marketing reporting. (NCT01497262)
Timeframe: 4 months

InterventionPercent of Participants (Number)
BradyarrhythmiasBlood pressure increaseHypertensionLiver Transaminase evaluationsInfectionsMacula Oedema
Fingolimod 0.5 mg8.60.62.53.728.40

Number of Participants With Adverse Events as a Measure of Safety and Tolerability

Any Adverse Event was defined as occurrence of any symptom regardless of intensity grade, Serious Adverse Event (SAEs) assessed as medical occurrences that result in death, are life threatening, require hospitalization or prolongation of hospitalization or result in persistent or significant disability/incapacity (NCT01497262)
Timeframe: 28 weeks

InterventionParticipants (Number)
Adverse Events (AE)Serious Adverse Event (SAE)Deaths
Fingolimod 0.5 mg100120

Participant Retention Rate Over 12 Months

Comparison effectiveness of fingolimod versus approved first-line disease modifying therapies by measuring the rate of participant retention on randomized treatment over a 12-month period (Full analysis set) (NCT01623596)
Timeframe: at 12 months

Interventionparticipants (Number)
Fingolimod352
Disease Modifying Therapy (MS-DMT)125

Change From Baseline of Symbol Digit Modalities Test (SDMT) Scores (Oral Test) by Visit (Randomized Treatment / Randomized Phase)

"Summary statistics Compare cognitive impairment measured by Symbol Digit Modalities Test (SDMT) scores. The SDMT score and its change from baseline value were summarized by visit. For the change from baseline values at each visit, ANCOVA adjusted for treatment naivety, corresponding baseline values, and age was performed for treatment comparisons~The SDMT measures the time to pair abstract symbols with specific numbers. The test requires elements of attention, visuoperceptual processing, working memory, and psychomotor speed. The score is the number of correctly coded items from 0-110 in 90 seconds. The total score provides a measure of the speed and accuracy of symbol-digit substitution.~NOTE: Higher scores indicate better performance." (NCT01623596)
Timeframe: baseline, 6 months, 12 months, and Last assessment which is either at Month 12 or at early discontinuation

,
InterventionSDMT score (Mean)
Baseline (n=76,70)Change at 6 months (n=70,64)Change at 12 months (n=58, 18)Change at Last assessment (n=73, 65)
Disease Modifying Therapy (MS-DMT)51.600.201.400.40
Fingolimod52.002.203.203.30

Change From Baseline of Symbol Digit Modalities Test (SDMT) Scores (Written Test) by Visit (Randomized Treatment / Randomized Phase)

"Summary statistics Compare cognitive impairment measured by Symbol Digit Modalities Test (SDMT) scores. The SDMT score and its change from baseline value were summarized by visit. For the change from baseline values at each visit, ANCOVA adjusted for treatment naivety, corresponding baseline values, and age was performed for treatment comparisons~The SDMT measures the time to pair abstract symbols with specific numbers. The test requires elements of attention, visuoperceptual processing, working memory, and psychomotor speed. The score is the number of correctly coded items from 0-110 in 90 seconds. The total score provides a measure of the speed and accuracy of symbol-digit substitution.~NOTE: Higher scores indicate better performance." (NCT01623596)
Timeframe: baseline, 6 months, 12 months, Last assessment which is either at Month 12 or at early discontinuation

,
InterventionSDMT score (Mean)
Baseline (n=355,348)Change at 6 months (n=339,322)Change at 12 months (n=282,105)Change at Last assessment (n=342, 324)
Disease Modifying Therapy (MS-DMT)48.500.700.400.70
Fingolimod48.90-0.500.700.80

Number of Satisfied Participants Per Medication Satisfaction Questionnaire (MSQ) Score

"Summary statistics for Medication Satisfaction Questionnaire[Question: Overall, how satisfied are you with your current medication?] (Randomized treatment / randomized phase): Fingolimod vs MS-DMT" (NCT01623596)
Timeframe: Baseline, 1 month, 3 months, 6 months, 9 months, at 12 months & Last assessment during randomized phase which is either at Month 12 or at early discontinuation

,
Interventionparticipants (Number)
Baseline, Extremely dissatisfiedBaseline, Very dissatisfiedBaseline, Somewhat dissatisfiedBaseline, Neither dissatisfied nor satisfiedBaseline, Somewhat satisfiedBaseline, Very satisfiedBaseline, Extremely satisfiedBaseline, Missing1 month, Extremely dissatisfied1 month, Very dissatisfied1 month, Somewhat dissatisfied1 month, Neither dissatisfied nor satisfied1 month, Somewhat satisfied1 month, Very satisfied1 month, Extremely satisfied1 month, Missing3 months, Extremely dissatisfied3 months, Very dissatisfied3 months, Somewhat dissatisfied3 months, Neither dissatisfied nor satisfied3 months, Somewhat satisfied3 months, Very satisfied3 months, Extremely satisfied3 months, Missing6 months, Extremely dissatisfied6 months, Very dissatisfied6 months, Somewhat dissatisfied6 months, Neither dissatisfied nor satisfied6 months, Somewhat satisfied6 months, Very satisfied6 months, Extremely satisfied6 months, Missing9 months, Extremely dissatisfied9 months, Very dissatisfied9 months, Somewhat dissatisfied9 months, Neither dissatisfied nor satisfied9 months, Somewhat satisfied9 months, Very satisfied9 months, Extremely satisfied9 months, Missing12 months, Extremely dissatisfied12 months, Very dissatisfied12 months, Somewhat dissatisfied12 months, Neither dissatisfied nor satisfied12 months, Somewhat satisfied12 months, Very satisfied12 months, Extremely satisfied12 months, MissingLast assessment, Extremely dissatisfiedLast assessment, Very dissatisfiedLast assessment, Somewhat dissatisfiedLast assessment,Neither dissatisfied nor satisfiedLast assessment, Somewhat satisfiedLast assessment, Very satisfiedLast assessment, Extremely satisfiedLast assessment, Missing
Disease Modifying Therapy (DS-DMT)161339614631172052937527489974010193842537391308291113244667262324612152958292753448226026301466157586994376
Fingolimod1022296235421421981483252161154451111464315314420815112557130151367812214213514563771127341151508214152345431281623

Percent Change From Baseline in Brain Volume From Month 12 to Last Visit (Randomized)

Summary statistics for percent change from month 12 in brain volume by visit (Randomized treatment / randomized phase) in patients treated with fingolimod vs.DMTs as measured by MRI (NCT01623596)
Timeframe: 12 months, and Last assessment which is either at Month 12 or at early discontinuation

,
Interventionpercent change in brain volume (Mean)
Percent change at 12 months (n=323, 111)Percent change at Last assessment (n=370, 246)
Disease Modifying Therapy-0.555-0.420
Fingolimod-0.396-0.385

Primary and Secondary Reasons for Discontinuation From Randomized Treatment: Randomized Set

"Reasons for discontinuation in participants treated with fingolimod vs. DMT over 12 months of treatment~Total discontinued (Primary reason): Fingolimod arm: 27, MS-DMT arm: 27 = 54 participants~Total discontinued (Secondary reason): Fingolimod arm: 257, MS-DMT arm: 256 = 513 participants~Throughout the study, investigators evaluated each patient for occurrence of randomized treatment discontinuation and determined the primary and secondary reasons for such discontinuation. At every visit, the investigator evaluated the patients and determined if they should continue on randomized treatment or change to alternative treatment. Treatment discontinuation was a clinically meaningful measure related to safety, efficacy, and tolerability over time, reflecting the therapeutic effectiveness of study treatment." (NCT01623596)
Timeframe: at 12 months

,
Interventionparticipants (Number)
Occurrence of relapse (Primary reason)Disease activity present in MRI (Primary reason)Injection site reaction (Primary reason)Flu-like symptoms (Primary reason)Lipoatrophy (Primary reason)Depression (Primary reason)Hepatic side effects (Primary reason)Spasticity (Primary reason)Infection (Primary reason)Macular edema (Primary reason)Bradycardia (Primary reason)Needle phobia (Primary reason)Inconvenient administration (Primary reason)Frequency of injections (Primary reason)Neutralizing antibodies present (Primary reason)Other (Primary reason)Occurrence of relapse (Secondary reason)Disease activity present in MRI (Secondary reason)Injection site reaction (Secondary reason)Flu-like symptoms (Secondary reason)Lipoatrophy (Secondary reason)Depression (Secondary reason)Hepatic side effects (Secondary reason)Spasticity (Secondary reason)Infection (Secondary reason)Macular edema (Secondary reason)Bradycardia (Secondary reason)Needle phobia (Secondary reason)Inconvenient administration (Secondary reason)Frequency of injections (Secondary reason)Neutralizing antibodies present (Secondary reason)Other (Secondary reason)
Disease Modifying Therapy (MS-DMT)146613414310001333290583540161602000185545065
Fingolimod5000017001000001302010051010000017

RMSSD Normal Breathing

"Root Mean Square of the Successive Differences (RMSSD) is one of a few time-domain tools used to assess heart rate variability, the successive differences being neighboring RR or pulse intervals.~It is calculated as the square root of the mean of the squares of the successive differences between adjacent RR intervals or pulse intervals.~In this study pulse intervals were measured non-invasively during five minutes, while subjects were supine, breathing regularly.~Measurements were done at two timepoints t=0 and t=4,5hours. RMSSD was compared between these two timepoints." (NCT02048072)
Timeframe: t-4,5 hours

Interventionmilliseconds (Mean)
Gilenya14.10

Participants Who Experienced at Least One Qualifying Cardiovascular Adverse Event

Participants from study CFTY720D2406 who experienced a qualifying cardiovascular adverse event were transferred to this study. Qualifying cardiovascular events included, but were not limited to, sudden unexplained death, cardiovascular death, myocardial infarction (MI), Q-wave MI, stroke (ischemic or hemorrhagic), unstable angina requiring hospitalization, congestive heart failure requiring hospitalization, complete heart block, ventricular fibrillation, torsade de pointes, hypertensive emergency and any other suspected life threatening cardiovascular condition. (NCT02232061)
Timeframe: Within 6 months of qualifying event up to 64 months

Interventionparticipants (Number)
Fingolimod0

Change From Baseline in T2 Lesion Volume

Inflammatory activity based on MRI measurement of new/newly enlarged T2 lesion volume (NCT01633112)
Timeframe: Baseline, 12 months/end of study

Interventioncubic centimeters (cc) (Mean)
FTY720 0.5 mg-0.14
FTY720 0.25 mg-0.05
GA 20 mg0.42

Confirmed Annualized Relapse Rate

Annualized relapse rate (ARR) was defined as the average number of confirmed relapses per year (i.e., the total number of confirmed relapses divided by the total days in the study multiplied by 365.25). The number of relapses included all the confirmed relapses experienced during the study from first dose to end of study. (NCT01633112)
Timeframe: up to 12 months

Interventionrelapses/year (Number)
FTY720 0.5 mg0.153
FTY720 0.25 mg0.221
GA 20 mg0.258

Gd Enhancing T1 Lesion Count

Inflammatory activity based on MRI measurement of Gd enhancing T1 lesion count (NCT01633112)
Timeframe: At 12 months/end of study

Interventionlesions (Mean)
FTY720 0.5 mg0.4
FTY720 0.25 mg0.4
GA 20 mg0.9

New or Newly Enlarging T2 Lesions

Inflammatory activity based on MRI measurement of new/newly enlarged T2 lesion count. (NCT01633112)
Timeframe: At 12 months/end of study

InterventionLesions (Mean)
FTY720 0.5 mg2.6
FTY720 0.25 mg3.3
GA 20 mg5.7

Number of Participants Free of New/Newly Enlarged T2 Lesions

Inflammatory activity based on MRI measurement of new/newly enlarged T2 lesion count. (NCT01633112)
Timeframe: At 12 months/end of study

InterventionParticipants (Number)
FTY720 0.5 mg156
FTY720 0.25 mg155
GA 20 mg96

Percent Brain Volume Change From Baseline

Using a Central MRI vendor to ensure calibrated MRI scanning equipment across all sites, MRI scans were performed on subjects following the established parameters and transferred to the central vendor for review of quality and assessment/evaluation. (NCT01633112)
Timeframe: Baseline, 12 months, end of study

InterventionPercentages of volume change (Mean)
FTY720 0.5 mg-0.652
FTY720 0.25 mg-0.636
GA 20 mg-0.561

Percentage of Patients Free of New T1 Hypointense Lesions

Based on MRI measures of new T1 hypointense lesions (NCT01633112)
Timeframe: 12 months

InterventionPercentage (Number)
FTY720 0.5 mg55.3
FTY720 0.25 mg52.1
GA 20 mg44.3

Change From Baseline in TSQM Scales

Treatment Satisfaction Questionnaire for Medication (TSQM) was developed and validated as a general measure for treatment satisfaction. Each scale score was calculated by summing individual items and then transformed to a 0-100 scale. Higher summary scores indicate better satisfaction with study drug. (NCT01633112)
Timeframe: 6 months, 12 months/end of study

,,
Interventionscore on a scale (Mean)
Global Satisfaction (Month 6)Global Satisfaction (Month 12)Effectiveness (Month 6)Effectiveness (Month 12)Side Effects (Month 6)Side Effects (Month 12)Convenience (Month 6)Convenience (Month 12)
FTY720 0.25 mg23.420.518.217.918.817.226.526.5
FTY720 0.5 mg20.819.215.216.816.916.230.729.5
GA 20 mg14.49.412.98.09.37.64.40.8

Gd Enhancing T1 Lesion Volume

Inflammatory activity based on MRI measurement of Gd enhancing T1 lesion count (NCT01633112)
Timeframe: Baseline, 12 months/end of study

,,
Interventioncubic centimeter (Mean)
BaselineMonth 12/end of study
FTY720 0.25 mg0.320.05
FTY720 0.5 mg0.310.06
GA 20 mg0.220.12

Research Highlights

Safety/Toxicity (93)

ArticleYear
Adverse effects of immunotherapies for multiple sclerosis: a network meta-analysis.
The Cochrane database of systematic reviews, 11-30, Volume: 11
2023
A short washout period from fingolimod to anti-CD20 therapy is safe and decreases the risk of reactivation.
Revue neurologique, Volume: 179, Issue: 9
2023
Real-life outcomes for oral disease-modifying treatments of relapsing-remitting multiple sclerosis patients: Adherence and adverse event profiles from Marmara University.
Turkish journal of medical sciences, Volume: 53, Issue: 3
2023
A Prospective, Observational Study Assessing Effectiveness, Safety, and QoL of Greek Patients with Multiple Sclerosis Under Treatment with Fingolimod.
Advances in therapy, Volume: 40, Issue: 5
2023
Third COVID-19 vaccine dose for people with multiple sclerosis who did not seroconvert following two doses of BBIBP-CorV (Sinopharm) inactivated vaccine: A pilot study on safety and immunogenicity.
Frontiers in immunology, Volume: 14
2023
Effectiveness and safety of switching from fingolimod and natalizumab to rituximab in patients with relapsing remitting multiple sclerosis.
Multiple sclerosis and related disorders, Volume: 71
2023
Efficacy and safety of adjunctive therapy with fingolimod in patients with schizophrenia: A randomized, double-blind, placebo-controlled clinical trial.
Schizophrenia research, Volume: 254
2023
S1PR1 modulators in multiple sclerosis: Efficacy, safety, comparison, and chemical structure insights.
European journal of medicinal chemistry, Mar-15, Volume: 250
2023
Comparison of the safety and efficacy of fingolimod and tofacitinib in the zebrafish model of colitis.
Food and chemical toxicology : an international journal published for the British Industrial Biological Research Association, Volume: 170
2022
Post marketing new adverse effects of oral therapies in multiple sclerosis: A systematic review.
Multiple sclerosis and related disorders, Volume: 68
2022
The Patient and Clinician Assessment of Gastrointestinal (GI) Related Adverse Events Associated with Oral Disease-Modifying Therapies in Multiple Sclerosis: A Qualitative Study.
Advances in therapy, Volume: 39, Issue: 11
2022
Humoral response and safety of the third booster dose of BNT162b2 mRNA COVID-19 vaccine in patients with multiple sclerosis treated with ocrelizumab or fingolimod.
Journal of neurology, Volume: 269, Issue: 12
2022
Comparative safety of high-efficacy disease-modifying therapies in relapsing-remitting multiple sclerosis: a systematic review and network meta-analysis.
Neurological sciences : official journal of the Italian Neurological Society and of the Italian Society of Clinical Neurophysiology, Volume: 43, Issue: 9
2022
Novel multiple sclerosis agents-associated cardiotoxicity: A real-world pharmacovigilance study.
International journal of cardiology, 09-01, Volume: 362
2022
The efficacy and safety of fingolimod plus standardized treatment versus standardized treatment alone for acute ischemic stroke: A systematic review and meta-analysis.
Pharmacology research & perspectives, Volume: 10, Issue: 3
2022
Difference in safety and humoral response to mRNA SARS-CoV-2 vaccines in patients with autoimmune neurological disorders: the ANCOVAX study.
Journal of neurology, Volume: 269, Issue: 8
2022
The safety and efficacy of fingolimod: Real-world data from a long-term, non-interventional study on the treatment of RRMS patients spanning up to 5 years from Hungary.
PloS one, Volume: 17, Issue: 4
2022
Real-world effectiveness and safety of fingolimod in patients with multiple sclerosis in the Czech Republic: results from core and extension parts of the GOLEMS study up to 48 months.
BMC neurology, Apr-15, Volume: 22, Issue: 1
2022
The CLARION study design and status update: a long-term, registry-based study evaluating adverse events of special interest in patients with relapsing multiple sclerosis newly started on cladribine tablets.
Current medical research and opinion, Volume: 38, Issue: 7
2022
Is pregnancy in MS patients safe and what is its impact on MS course? Real World evidence of 1533 pregnancies in Czech Republic.
Multiple sclerosis and related disorders, Volume: 59
2022
Long-term real-world effectiveness and safety of fingolimod over 5 years in Germany.
Journal of neurology, Volume: 269, Issue: 6
2022
Risk for Cardiovascular Adverse Events Associated With Sphingosine-1-Phosphate Receptor Modulators in Patients With Multiple Sclerosis: Insights From a Pooled Analysis of 15 Randomised Controlled Trials.
Frontiers in immunology, Volume: 12
2021
Immunogenicity and safety of mRNA COVID-19 vaccines in people with multiple sclerosis treated with different disease-modifying therapies.
Neurotherapeutics : the journal of the American Society for Experimental NeuroTherapeutics, Volume: 19, Issue: 1
2022
A phase II, Multicentre, Randomised, Double-Blind, Placebo-controlled Study to Evaluate Safety, Tolerability, and Efficacy of Amiselimod in Patients with Moderate to Severe Active Crohn's Disease.
Journal of Crohn's & colitis, Jun-24, Volume: 16, Issue: 5
2022
Four-year safety and effectiveness data from patients with multiple sclerosis treated with fingolimod: The Spanish GILENYA registry.
PloS one, Volume: 16, Issue: 10
2021
Real-world retrospective study of effectiveness and safety of FINgOlimod in relapsing remitting multiple sclerosis in the Middle East and North Africa (FINOMENA).
Clinical neurology and neurosurgery, Volume: 203
2021
Safety of disease-modifying treatments in SARS-CoV-2 antibody-positive multiple sclerosis patients.
Multiple sclerosis and related disorders, Volume: 49
2021
Longitudinal analysis of safety and medication adherence of patients in the Fingolimod patient support program: a real-world observational study.
Scientific reports, 02-18, Volume: 11, Issue: 1
2021
Short-term laboratory and related safety outcomes for the multiple sclerosis oral disease-modifying therapies: an observational study.
Expert opinion on drug safety, Volume: 20, Issue: 4
2021
REALMS study: real-world effectiveness and safety of fingolimod in patients with relapsing-remitting multiple sclerosis in Portugal.
Neurological sciences : official journal of the Italian Neurological Society and of the Italian Society of Clinical Neurophysiology, Volume: 42, Issue: 5
2021
Real-world evidence on the safety and effectiveness of fingolimod in patients with multiple sclerosis from Taiwan.
Journal of the Formosan Medical Association = Taiwan yi zhi, Volume: 120, Issue: 1 Pt 2
2021
The efficacy and safety of fingolimod in patients with relapsing multiple sclerosis: A meta-analysis.
British journal of clinical pharmacology, Volume: 86, Issue: 4
2020
Short term real-world Fingolimod efficacy and safety in Emirati patients with multiple sclerosis.
Journal of clinical neuroscience : official journal of the Neurosurgical Society of Australasia, Volume: 71
2020
Analysis of cardiac monitoring and safety data in patients initiating fingolimod treatment in the home or in clinic.
BMC neurology, Nov-15, Volume: 19, Issue: 1
2019
Identification of PP2A and S6 Kinase as Modifiers of Leucine-Rich Repeat Kinase-Induced Neurotoxicity.
Neuromolecular medicine, Volume: 22, Issue: 2
2020
A novel FTY720 analogue targets SET-PP2A interaction and inhibits growth of acute myeloid leukemia cells without inducing cardiac toxicity.
Cancer letters, 01-01, Volume: 468
2020
Promoting transitions of care, safety, and medication adherence for patients taking fingolimod in community pharmacies.
American journal of health-system pharmacy : AJHP : official journal of the American Society of Health-System Pharmacists, Jul-18, Volume: 76, Issue: 15
2019
Treatment with alemtuzumab or rituximab after fingolimod withdrawal in relapsing-remitting multiple sclerosis is effective and safe.
Journal of neurology, Volume: 266, Issue: 3
2019
Efficacy and safety of alemtuzumab versus fingolimod in RRMS after natalizumab cessation.
Journal of neurology, Volume: 266, Issue: 1
2019
[Efficacy and safety of fingolimod in routine clinical practice in patients with relapsing-remitting multiple sclerosis in Spain: an intermediate analysis of the MS NEXT study].
Revista de neurologia, Sep-01, Volume: 67, Issue: 5
2018
Clinical effectiveness and safety of fingolimod in relapsing remitting multiple sclerosis in Western Iran.
Neurosciences (Riyadh, Saudi Arabia), Volume: 23, Issue: 2
2018
Managing the side effects of multiple sclerosis therapy: pharmacotherapy options for patients.
Expert opinion on pharmacotherapy, Volume: 19, Issue: 5
2018
Efficacy and safety outcomes in vitamin D supplement users in the fingolimod phase 3 trials.
Journal of neurology, Volume: 265, Issue: 2
2018
Gender differences in safety issues during Fingolimod therapy: Evidence from a real-life Relapsing Multiple Sclerosis cohort.
Brain and behavior, Volume: 7, Issue: 10
2017
Comparison of efficacy and safety of oral agents for the treatment of relapsing-remitting multiple sclerosis.
Drug design, development and therapy, Volume: 11
2017
Effectiveness, safety and health-related quality of life of multiple sclerosis patients treated with fingolimod: results from a 12-month, real-world, observational PERFORMS study in the Middle East.
BMC neurology, Aug-07, Volume: 17, Issue: 1
2017
Phase IIa trial of fingolimod for amyotrophic lateral sclerosis demonstrates acceptable acute safety and tolerability.
Muscle & nerve, Volume: 56, Issue: 6
2017
Multiple sclerosis treatment with fingolimod: profile of non-cardiologic adverse events.
Acta neurologica Belgica, Volume: 117, Issue: 4
2017
Sphingosine Toxicity in EAE and MS: Evidence for Ceramide Generation via Serine-Palmitoyltransferase Activation.
Neurochemical research, Volume: 42, Issue: 10
2017
The real-world effectiveness and safety of fingolimod in relapsing-remitting multiple sclerosis patients: An observational study.
PloS one, Volume: 12, Issue: 4
2017
Fingolimod Limits Acute Aβ Neurotoxicity and Promotes Synaptic Versus Extrasynaptic NMDA Receptor Functionality in Hippocampal Neurons.
Scientific reports, 01-30, Volume: 7
2017
Long-term efficacy and safety of fingolimod in Japanese patients with relapsing multiple sclerosis: 3-year results of the phase 2 extension study.
BMC neurology, Jan-28, Volume: 17, Issue: 1
2017
Early safety and efficacy of fingolimod treatment in Denmark.
Acta neurologica Scandinavica, Volume: 135, Issue: 1
2017
Adverse Effect Profile of Topical Ocular Administration of Fingolimod for Treatment of Dry Eye Disease.
Basic & clinical pharmacology & toxicology, Volume: 120, Issue: 4
2017
Fingolimod: therapeutic mechanisms and ocular adverse effects.
Eye (London, England), Volume: 31, Issue: 2
2017
Safety and Efficacy of Fingolimod and Natalizumab in Multiple Sclerosis After the Failure of First-Line Therapy: Single Center Experience Based on the Treatment of Forty-Four Patients.
Medical science monitor : international medical journal of experimental and clinical research, Nov-10, Volume: 22
2016
Safety and tolerability of fingolimod in patients with relapsing-remitting multiple sclerosis: results of an open-label clinical trial in Italy.
Neurological sciences : official journal of the Italian Neurological Society and of the Italian Society of Clinical Neurophysiology, Volume: 38, Issue: 1
2017
Safety of switching from natalizumab straight into fingolimod in a group of JCV-positive patients with multiple sclerosis.
Arquivos de neuro-psiquiatria, Volume: 74, Issue: 8
2016
Update on monitoring and adverse effects of approved second-generation disease-modifying therapies in relapsing forms of multiple sclerosis.
Current opinion in neurology, Volume: 29, Issue: 3
2016
FTY720-induced enhancement of autophagy protects cells from FTY720 cytotoxicity in colorectal cancer.
Oncology reports, Volume: 35, Issue: 5
2016
A systematic evaluation of the safety and toxicity of fingolimod for its potential use in the treatment of acute myeloid leukaemia.
Anti-cancer drugs, Volume: 27, Issue: 6
2016
Efficacy and Safety of Fingolimod in an Unselected Patient Population.
PloS one, Volume: 11, Issue: 1
2016
Safety and efficacy of fingolimod in clinical practice: The experience of an academic center in the Middle East.
Journal of neuroimmunology, Dec-15, Volume: 289
2015
Fingolimod: a review of its mode of action in the context of its efficacy and safety profile in relapsing forms of multiple sclerosis.
Expert review of neurotherapeutics, Volume: 16, Issue: 1
2016
AKP-11 - A Novel S1P1 Agonist with Favorable Safety Profile Attenuates Experimental Autoimmune Encephalomyelitis in Rat Model of Multiple Sclerosis.
PloS one, Volume: 10, Issue: 10
2015
Fingolimod-Associated Peripheral Vascular Adverse Effects.
Mayo Clinic proceedings, Volume: 90, Issue: 10
2015
Safety and efficacy of reduced fingolimod dosage treatment.
Journal of neuroimmunology, Aug-15, Volume: 285
2015
Safety and Tolerability of Fingolimod in Latin American Patients with Relapsing-Remitting Multiple Sclerosis: The Open-Label FIRST LATAM Study.
Advances in therapy, Volume: 32, Issue: 7
2015
ROR1-targeted delivery of OSU-2S, a nonimmunosuppressive FTY720 derivative, exerts potent cytotoxicity in mantle-cell lymphoma in vitro and in vivo.
Experimental hematology, Volume: 43, Issue: 9
2015
Long-term cardiac safety and tolerability of fingolimod in multiple sclerosis: A postmarketing study.
Journal of clinical pharmacology, Volume: 55, Issue: 10
2015
ASP4058, a novel agonist for sphingosine 1-phosphate receptors 1 and 5, ameliorates rodent experimental autoimmune encephalomyelitis with a favorable safety profile.
PloS one, Volume: 9, Issue: 10
2014
Efficacy and safety of fingolimod in Hispanic patients with multiple sclerosis: pooled clinical trial analyses.
Advances in therapy, Volume: 31, Issue: 10
2014
Tumor antigen ROR1 targeted drug delivery mediated selective leukemic but not normal B-cell cytotoxicity in chronic lymphocytic leukemia.
Leukemia, Volume: 29, Issue: 2
2015
Overview and safety of fingolimod hydrochloride use in patients with multiple sclerosis.
Expert opinion on drug safety, Volume: 13, Issue: 7
2014
Safety of the first dose of fingolimod for multiple sclerosis: results of an open-label clinical trial.
BMC neurology, Apr-01, Volume: 14
2014
Safety and efficacy of fingolimod in patients with relapsing-remitting multiple sclerosis (FREEDOMS II): a double-blind, randomised, placebo-controlled, phase 3 trial.
The Lancet. Neurology, Volume: 13, Issue: 6
2014
Assessment of cardiac safety during fingolimod treatment initiation in a real-world relapsing multiple sclerosis population: a phase 3b, open-label study.
Journal of neurology, Volume: 261, Issue: 2
2014
Efficacy and safety of BG-12 (dimethyl fumarate) and other disease-modifying therapies for the treatment of relapsing-remitting multiple sclerosis: a systematic review and mixed treatment comparison.
Current medical research and opinion, Volume: 30, Issue: 4
2014
Protective effect of FTY720 against sevoflurane-induced developmental neurotoxicity in rats.
Cell biochemistry and biophysics, Volume: 67, Issue: 2
2013
Early tolerability and safety of fingolimod in clinical practice.
Journal of the neurological sciences, Dec-15, Volume: 323, Issue: 1-2
2012
4-aminopyridine toxicity: a case report and review of the literature.
Journal of medical toxicology : official journal of the American College of Medical Toxicology, Volume: 8, Issue: 3
2012
Mechanisms of fingolimod's efficacy and adverse effects in multiple sclerosis.
Annals of neurology, Volume: 69, Issue: 5
2011
FTY720, a novel immunomodulator: efficacy and safety results from the first phase 2A study in de novo renal transplantation.
Transplantation, Jun-15, Volume: 79, Issue: 11
2005
FTY720, a novel immunomodulator: efficacy and safety results from the first phase 2A study in de novo renal transplantation.
Transplantation, Jun-27, Volume: 77, Issue: 12
2004
Current immunosuppressive agents: efficacy, side effects, and utilization.
Pediatric clinics of North America, Volume: 50, Issue: 6
2003
Pharmacodynamics, pharmacokinetics, and safety of multiple doses of FTY720 in stable renal transplant patients: a multicenter, randomized, placebo-controlled, phase I study.
Transplantation, Oct-15, Volume: 76, Issue: 7
2003
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Long-term Use (14)

ArticleYear
Four-year safety and effectiveness data from patients with multiple sclerosis treated with fingolimod: The Spanish GILENYA registry.
PloS one, Volume: 16, Issue: 10
2021
Assessment of Macular Function by Multifocal Electroretinogram in Patients with Multiple Sclerosis Treated with Fingolimod.
Advances in therapy, Volume: 38, Issue: 7
2021
The protumorigenic potential of FTY720 by promoting extramedullary hematopoiesis and MDSC accumulation.
Oncogene, 06-29, Volume: 36, Issue: 26
2017
Immunologic mechanisms of fingolimod and the role of immunosenescence in the risk of cryptococcal infection: A case report and review of literature.
Multiple sclerosis and related disorders, Volume: 9
2016
Depletion of T lymphocytes ameliorates cardiac fibrosis in streptozotocin-induced diabetic cardiomyopathy.
International immunopharmacology, Volume: 39
2016
Fingolimod induces neurogenesis in adult mouse hippocampus and improves contextual fear memory.
Translational psychiatry, Nov-24, Volume: 5
2015
Fingolimod (FTY720) enhances hippocampal synaptic plasticity and memory in Huntington's disease by preventing p75NTR up-regulation and astrocyte-mediated inflammation.
Human molecular genetics, Sep-01, Volume: 24, Issue: 17
2015
Oral disease-modifying therapies for relapsing-remitting multiple sclerosis.
American journal of health-system pharmacy : AJHP : official journal of the American Society of Health-System Pharmacists, Jan-01, Volume: 72, Issue: 1
2015
Recurrence of disease activity after repeated Natalizumab withdrawals.
Neurological sciences : official journal of the Italian Neurological Society and of the Italian Society of Clinical Neurophysiology, Volume: 36, Issue: 3
2015
Overview and safety of fingolimod hydrochloride use in patients with multiple sclerosis.
Expert opinion on drug safety, Volume: 13, Issue: 7
2014
FTY720 (fingolimod) is a neuroprotective and disease-modifying agent in cellular and mouse models of Huntington disease.
Human molecular genetics, May-01, Volume: 23, Issue: 9
2014
Neurorestorative effect of FTY720 in a rat model of Alzheimer's disease: comparison with memantine.
Behavioural brain research, Sep-01, Volume: 252
2013
Effects of long-term administration of low-dose FTY720 on survival of murine cardiac allograft.
Journal of Huazhong University of Science and Technology. Medical sciences = Hua zhong ke ji da xue xue bao. Yi xue Ying De wen ban = Huazhong keji daxue xuebao. Yixue Yingdewen ban, Volume: 32, Issue: 2
2012
Prevention and cure of autoimmune diabetes in nonobese diabetic mice by continuous administration of FTY720.
Transplantation, May-15, Volume: 79, Issue: 9
2005
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Pharmacokinetics (47)

ArticleYear
Brain volume loss and physical and cognitive impairment in naive multiple sclerosis patients treated with fingolimod: prospective cohort study in Buenos Aires, Argentina.
Arquivos de neuro-psiquiatria, Volume: 80, Issue: 7
2022
Two-year macular volume assessment in multiple sclerosis patients treated with fingolimod.
Neurological sciences : official journal of the Italian Neurological Society and of the Italian Society of Clinical Neurophysiology, Volume: 42, Issue: 2
2021
A sensitive liquid chromatography-tandem mass spectrometry method for quantitative bioanalysis of fingolimod in human blood: Application to pharmacokinetic study.
Biomedical chromatography : BMC, Volume: 34, Issue: 6
2020
Two-year regional grey and white matter volume changes with natalizumab and fingolimod.
Journal of neurology, neurosurgery, and psychiatry, Volume: 91, Issue: 5
2020
Fingolimod's Impact on MRI Brain Volume Measures in Multiple Sclerosis: Results from MS-MRIUS.
Journal of neuroimaging : official journal of the American Society of Neuroimaging, Volume: 28, Issue: 4
2018
Pharmacokinetic Interaction Between Fingolimod and Carbamazepine in Healthy Subjects.
Clinical pharmacology in drug development, Volume: 7, Issue: 6
2018
A two-year study using cerebral gray matter volume to assess the response to fingolimod therapy in multiple sclerosis.
Journal of the neurological sciences, Dec-15, Volume: 383
2017
Effect of Fingolimod on Brain Volume Loss in Patients with Multiple Sclerosis.
CNS drugs, Volume: 31, Issue: 4
2017
Preclinical Metabolism, Pharmacokinetics and In Vivo Analysis of New Blood-Brain-Barrier Penetrant Fingolimod Analogues: FTY720-C2 and FTY720-Mitoxy.
PloS one, Volume: 11, Issue: 9
2016
Inclusion of brain volume loss in a revised measure of 'no evidence of disease activity' (NEDA-4) in relapsing-remitting multiple sclerosis.
Multiple sclerosis (Houndmills, Basingstoke, England), Volume: 22, Issue: 10
2016
The relationship between the rate of brain volume loss during first 24 months and disability progression over 24 and 48 months in relapsing MS.
Journal of neurology, Volume: 263, Issue: 2
2016
Pharmacokinetics of fingolimod and metabolites in subjects with severe renal impairment: An open-label, single-dose, parallel-group study.
International journal of clinical pharmacology and therapeutics, Volume: 53, Issue: 10
2015
Fingolimod effect on brain volume loss independently contributes to its effect on disability.
Multiple sclerosis (Houndmills, Basingstoke, England), Volume: 21, Issue: 7
2015
Correlation between brain volume loss and clinical and MRI outcomes in multiple sclerosis.
Neurology, Feb-24, Volume: 84, Issue: 8
2015
Translational pharmacokinetic modeling of fingolimod (FTY720) as a paradigm compound subject to sphingosine kinase-mediated phosphorylation.
Drug metabolism and disposition: the biological fate of chemicals, Volume: 42, Issue: 9
2014
Molecular pharmacodynamics of new oral drugs used in the treatment of multiple sclerosis.
Drug design, development and therapy, Volume: 8
2014
The influence of patient demographics, disease characteristics and treatment on brain volume loss in Trial Assessing Injectable Interferon vs FTY720 Oral in Relapsing-Remitting Multiple Sclerosis (TRANSFORMS), a phase 3 study of fingolimod in multiple scl
Multiple sclerosis (Houndmills, Basingstoke, England), Volume: 20, Issue: 13
2014
Pharmacokinetic evaluation of fingolimod for the treatment of multiple sclerosis.
Expert opinion on drug metabolism & toxicology, Volume: 10, Issue: 4
2014
Baseline retinal nerve fiber layer thickness and macular volume quantified by OCT in the North American phase 3 fingolimod trial for relapsing-remitting multiple sclerosis.
Journal of neuro-ophthalmology : the official journal of the North American Neuro-Ophthalmology Society, Volume: 33, Issue: 4
2013
Treatment with the sphingosine-1-phosphate analogue FTY 720 reduces loss of plasma volume during experimental sepsis in the rat.
Acta anaesthesiologica Scandinavica, Volume: 57, Issue: 6
2013
Fingolimod treatment in multiple sclerosis leads to increased macular volume.
Neurology, Jan-08, Volume: 80, Issue: 2
2013
Higher macular volume in patients with MS receiving fingolimod: positive outcome or side effect?
Neurology, Jan-08, Volume: 80, Issue: 2
2013
Pharmacokinetics of fingolimod (FTY720) and a combined oral contraceptive coadministered in healthy women: drug-drug interaction study results.
International journal of clinical pharmacology and therapeutics, Volume: 50, Issue: 8
2012
The sphingosine-1-phosphate analogue FTY720 impairs mucosal immunity and clearance of the enteric pathogen Citrobacter rodentium.
Infection and immunity, Volume: 80, Issue: 8
2012
The utility of pharmacokinetic-pharmacodynamic modeling in the discovery and optimization of selective S1P(1) agonists.
Xenobiotica; the fate of foreign compounds in biological systems, Volume: 42, Issue: 7
2012
Pharmacodynamic effects of steady-state fingolimod on antibody response in healthy volunteers: a 4-week, randomized, placebo-controlled, parallel-group, multiple-dose study.
Journal of clinical pharmacology, Volume: 52, Issue: 12
2012
Clinical pharmacokinetics of fingolimod.
Clinical pharmacokinetics, Jan-01, Volume: 51, Issue: 1
2012
Population pharmacokinetics of fingolimod phosphate in healthy participants.
Journal of clinical pharmacology, Volume: 52, Issue: 7
2012
Pharmacokinetics, pharmacodynamics, safety, and tolerability of single-dose fingolimod (FTY720) in adolescents with stable renal transplants.
Pediatric transplantation, Volume: 15, Issue: 4
2011
Transient FTY720 treatment promotes immune-mediated clearance of a chronic viral infection.
Nature, Aug-14, Volume: 454, Issue: 7206
2008
Oral-intravenous crossover study of fingolimod pharmacokinetics, lymphocyte responses and cardiac effects.
Biopharmaceutics & drug disposition, Volume: 28, Issue: 2
2007
Oral pharmacokinetic and pharmacodynamic effects of FTY720 in cats.
Journal of veterinary pharmacology and therapeutics, Volume: 30, Issue: 1
2007
Physiologically based pharmacokinetic modeling of FTY720 (2-amino-2[2-(-4-octylphenyl)ethyl]propane-1,3-diol hydrochloride) in rats after oral and intravenous doses.
Drug metabolism and disposition: the biological fate of chemicals, Volume: 34, Issue: 9
2006
Fingolimod (FTY720) in severe hepatic impairment: pharmacokinetics and relationship to markers of liver function.
Journal of clinical pharmacology, Volume: 46, Issue: 2
2006
FTY720, a novel immunomodulator in de novo kidney transplant patients: pharmacokinetics and exposure-response relationship.
Journal of clinical pharmacology, Volume: 45, Issue: 11
2005
Pharmacokinetic/pharmacodynamic relationships of FTY720 in kidney transplant recipients.
Brazilian journal of medical and biological research = Revista brasileira de pesquisas medicas e biologicas, Volume: 38, Issue: 5
2005
FTY720 pharmacokinetics in mild to moderate hepatic impairment.
Journal of clinical pharmacology, Volume: 45, Issue: 4
2005
On the prediction of the human response: a recycled mechanistic pharmacokinetic/pharmacodynamic approach.
Basic & clinical pharmacology & toxicology, Volume: 96, Issue: 3
2005
Multiple-dose FTY720: tolerability, pharmacokinetics, and lymphocyte responses in healthy subjects.
Journal of clinical pharmacology, Volume: 44, Issue: 5
2004
Single-dose FTY720 pharmacokinetics, food effect, and pharmacological responses in healthy subjects.
British journal of clinical pharmacology, Volume: 57, Issue: 5
2004
Pharmacodynamics of FTY720, the first member of a new class of immune-modulating therapeutics in transplantation medicine.
International journal of clinical pharmacology and therapeutics, Volume: 41, Issue: 10
2003
Pharmacodynamics, pharmacokinetics, and safety of multiple doses of FTY720 in stable renal transplant patients: a multicenter, randomized, placebo-controlled, phase I study.
Transplantation, Oct-15, Volume: 76, Issue: 7
2003
Pharmacodynamics of single doses of the novel immunosuppressant FTY720 in stable renal transplant patients.
American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons, Volume: 3, Issue: 7
2003
Pharmacokinetics and cell trafficking dynamics of 2-amino-2-[2-(4-octylphenyl)ethyl]propane-1,3-diol hydrochloride (FTY720) in cynomolgus monkeys after single oral and intravenous doses.
The Journal of pharmacology and experimental therapeutics, Volume: 301, Issue: 2
2002
Update on pharmacokinetic/pharmacodynamic studies with FTY720 and sirolimus.
Therapeutic drug monitoring, Volume: 24, Issue: 1
2002
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Bioavailability (14)

ArticleYear
Analogs of FTY720 inhibit TRPM7 but not S1PRs and exert multimodal anti-inflammatory effects.
The Journal of general physiology, Jan-01, Volume: 156, Issue: 1
2024
CDX-modified chitosan nanoparticles remarkably reduce therapeutic dose of fingolimod in the EAE model of mice.
International journal of pharmaceutics, Apr-05, Volume: 636
2023
Intranasal administration of fingolimod (FTY720) attenuates demyelination area in lysolecithin-induced demyelination model of rat optic chiasm.
Multiple sclerosis and related disorders, Volume: 59
2022
Preclinical Metabolism, Pharmacokinetics and In Vivo Analysis of New Blood-Brain-Barrier Penetrant Fingolimod Analogues: FTY720-C2 and FTY720-Mitoxy.
PloS one, Volume: 11, Issue: 9
2016
Lipid Nanosystems Enhance the Bioavailability and the Therapeutic Efficacy of FTY720 in Acute Myeloid Leukemia.
Journal of biomedical nanotechnology, Volume: 11, Issue: 4
2015
Assessing the potential impact of non-proprietary drug copies on quality of medicine and treatment in patients with relapsing multiple sclerosis: the experience with fingolimod.
Drug design, development and therapy, Volume: 8
2014
First-line disease-modifying therapies in paediatric multiple sclerosis: a comprehensive overview.
Drugs, Jun-18, Volume: 72, Issue: 9
2012
Clinical pharmacokinetics of fingolimod.
Clinical pharmacokinetics, Jan-01, Volume: 51, Issue: 1
2012
Population pharmacokinetics of fingolimod phosphate in healthy participants.
Journal of clinical pharmacology, Volume: 52, Issue: 7
2012
FTY720 and (S)-FTY720 vinylphosphonate inhibit sphingosine kinase 1 and promote its proteasomal degradation in human pulmonary artery smooth muscle, breast cancer and androgen-independent prostate cancer cells.
Cellular signalling, Volume: 22, Issue: 10
2010
FTY720: A new kid on the block for transplant immunosuppression.
Expert opinion on biological therapy, Volume: 3, Issue: 4
2003
Pharmacokinetics and cell trafficking dynamics of 2-amino-2-[2-(4-octylphenyl)ethyl]propane-1,3-diol hydrochloride (FTY720) in cynomolgus monkeys after single oral and intravenous doses.
The Journal of pharmacology and experimental therapeutics, Volume: 301, Issue: 2
2002
Immunosuppressive drugs in paediatric liver transplantation.
Paediatric drugs, Volume: 3, Issue: 1
2001
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Dosage (70)

ArticleYear
Effects of Crocin on brain neurotrophins, cognition, balance and pain in toxic-induced demyelination model.
Acta neurologica Taiwanica, Jun-30, Volume: 33(2)
2024
Comparative effectiveness of cladribine tablets versus fingolimod in the treatment of highly active multiple sclerosis: A real-world study.
Multiple sclerosis and related disorders, Volume: 76
2023
Using an animal model to predict the effective human dose for oral multiple sclerosis drugs.
Clinical and translational science, Volume: 16, Issue: 3
2023
Fingolimod attenuates gait deficits in mice subjected to experimental autoimmune encephalomyelitis.
Journal of neuroimmunology, 09-15, Volume: 370
2022
USPIO-SWI Shows Fingolimod Enhanced Alteplase Action on Angiographic Reperfusion in eMCAO Rats.
Journal of magnetic resonance imaging : JMRI, Volume: 55, Issue: 4
2022
Immunomodulation Eliminates Inflammation in the Hippocampus in Experimental Autoimmune Encephalomyelitis, but Does Not Ameliorate Anxiety-Like Behavior.
Frontiers in immunology, Volume: 12
2021
Benefit-harm balance of fingolimod in patients with MS: A modelling study based on FREEDOMS.
Multiple sclerosis and related disorders, Volume: 46
2020
The underpinning biology relating to multiple sclerosis disease modifying treatments during the COVID-19 pandemic.
Multiple sclerosis and related disorders, Volume: 43
2020
Repurposed molecules for antiepileptogenesis: Missing an opportunity to prevent epilepsy?
Epilepsia, Volume: 61, Issue: 3
2020
Dual dose-dependent effects of fingolimod in a mouse model of Alzheimer's disease.
Scientific reports, 07-29, Volume: 9, Issue: 1
2019
Effect of dosage reduction on peripheral blood lymphocyte count in patients with multiple sclerosis receiving long-term fingolimod therapy.
Journal of clinical neuroscience : official journal of the Neurosurgical Society of Australasia, Volume: 63
2019
When does economic model type become a decisive factor in health technology appraisals? Learning from the expanding treatment options for relapsing-remitting multiple sclerosis.
Journal of medical economics, Volume: 21, Issue: 10
2018
Beneficial Effects of the Calcium Channel Blocker CTK 01512-2 in a Mouse Model of Multiple Sclerosis.
Molecular neurobiology, Volume: 55, Issue: 12
2018
Predictors of hematological abnormalities in multiple sclerosis patients treated with fingolimod and dimethyl fumarate and impact of treatment switch on lymphocyte and leukocyte count.
Multiple sclerosis and related disorders, Volume: 20
2018
FTY720 ameliorates renal fibrosis by simultaneously affecting leucocyte recruitment and TGF-β signalling in fibroblasts.
Clinical and experimental immunology, Volume: 190, Issue: 1
2017
Local delivery of fingolimod from three-dimensional scaffolds impacts islet graft efficacy and microenvironment in a murine diabetic model.
Journal of tissue engineering and regenerative medicine, Volume: 12, Issue: 2
2018
Selective Sphingosine 1-Phosphate Receptor 1 Agonist Is Protective Against Ischemia/Reperfusion in Mice.
Stroke, Volume: 47, Issue: 12
2016
Comparative effectiveness using a matching-adjusted indirect comparison between delayed-release dimethyl fumarate and fingolimod for the treatment of multiple sclerosis.
Current medical research and opinion, Volume: 33, Issue: 2
2017
Fingolimod enhances myelin repair of hippocampus in pentylenetetrazol-induced kindling model.
European journal of pharmaceutical sciences : official journal of the European Federation for Pharmaceutical Sciences, Jan-01, Volume: 96
2017
Preclinical Metabolism, Pharmacokinetics and In Vivo Analysis of New Blood-Brain-Barrier Penetrant Fingolimod Analogues: FTY720-C2 and FTY720-Mitoxy.
PloS one, Volume: 11, Issue: 9
2016
Effect of fingolimod on cardiac autonomic regulation in patients with multiple sclerosis.
Multiple sclerosis (Houndmills, Basingstoke, England), Volume: 22, Issue: 8
2016
First-dose effects of fingolimod: Pooled safety data from three phase 3 studies.
Multiple sclerosis and related disorders, Volume: 3, Issue: 5
2014
Safety and efficacy of reduced fingolimod dosage treatment.
Journal of neuroimmunology, Aug-15, Volume: 285
2015
New FDA-Approved Disease-Modifying Therapies for Multiple Sclerosis.
Clinical therapeutics, Apr-01, Volume: 37, Issue: 4
2015
Fingolimod treatment promotes proliferation and differentiation of oligodendrocyte progenitor cells in mice with experimental autoimmune encephalomyelitis.
Neurobiology of disease, Volume: 76
2015
[Fingolimod treatment in multiple sclerosis].
Nihon rinsho. Japanese journal of clinical medicine, Volume: 72, Issue: 11
2014
Varicella-zoster virus infections in patients treated with fingolimod: risk assessment and consensus recommendations for management.
JAMA neurology, Volume: 72, Issue: 1
2015
Fingolimod: a review of its use in relapsing-remitting multiple sclerosis.
Drugs, Volume: 74, Issue: 12
2014
Fingolimod-associated amenorrhea: a report of three cases.
Multiple sclerosis (Houndmills, Basingstoke, England), Volume: 20, Issue: 12
2014
An update on new and emerging therapies for relapsing-remitting multiple sclerosis.
The American journal of managed care, Volume: 19, Issue: 17 Suppl
2013
Dual effects of daily FTY720 on human astrocytes in vitro: relevance for neuroinflammation.
Journal of neuroinflammation, Mar-19, Volume: 10
2013
Early administration of FTY720 prevents chronic airway as well as vascular destruction in experimental rat lung transplantation.
Transplantation proceedings, Volume: 45, Issue: 2
2013
Inhibition of de novo ceramide biosynthesis by FTY720 protects rat retina from light-induced degeneration.
Journal of lipid research, Volume: 54, Issue: 6
2013
Is this the dose for you?: the role of modeling.
Clinical pharmacology and therapeutics, Volume: 93, Issue: 2
2013
Fingolimod in the treatment algorithm of relapsing remitting multiple sclerosis: a statement of the Central and East European (CEE) MS Expert Group.
Wiener medizinische Wochenschrift (1946), Volume: 162, Issue: 15-16
2012
Cost-effectiveness of early initiation of fingolimod versus delayed initiation after 1 year of intramuscular interferon beta-1a in patients with multiple sclerosis.
Clinical therapeutics, Volume: 34, Issue: 7
2012
Pharmacokinetics of fingolimod (FTY720) and a combined oral contraceptive coadministered in healthy women: drug-drug interaction study results.
International journal of clinical pharmacology and therapeutics, Volume: 50, Issue: 8
2012
First-line disease-modifying therapies in paediatric multiple sclerosis: a comprehensive overview.
Drugs, Jun-18, Volume: 72, Issue: 9
2012
Sphingosine-1-phosphate-induced airway hyper-reactivity in rodents is mediated by the sphingosine-1-phosphate type 3 receptor.
The Journal of pharmacology and experimental therapeutics, Volume: 342, Issue: 2
2012
Annualized relapse rate of first-line treatments for multiple sclerosis: a meta-analysis, including indirect comparisons versus fingolimod.
Current medical research and opinion, Volume: 28, Issue: 5
2012
Clinical pharmacokinetics of fingolimod.
Clinical pharmacokinetics, Jan-01, Volume: 51, Issue: 1
2012
Therapeutic dosing of fingolimod (FTY720) prevents cell infiltration, rapidly suppresses ocular inflammation, and maintains the blood-ocular barrier.
The American journal of pathology, Volume: 180, Issue: 2
2012
S1P(1) receptor modulation with cyclical recovery from lymphopenia ameliorates mouse model of multiple sclerosis.
Molecular pharmacology, Volume: 81, Issue: 2
2012
Fingolimod: a review of its use in the management of relapsing-remitting multiple sclerosis.
CNS drugs, Volume: 25, Issue: 8
2011
Characterization of a sphingosine 1-phosphate receptor antagonist prodrug.
The Journal of pharmacology and experimental therapeutics, Volume: 338, Issue: 3
2011
Pharmacokinetics, pharmacodynamics, safety, and tolerability of single-dose fingolimod (FTY720) in adolescents with stable renal transplants.
Pediatric transplantation, Volume: 15, Issue: 4
2011
Fingolimod: an oral disease-modifying therapy for relapsing multiple sclerosis.
Advances in therapy, Volume: 28, Issue: 4
2011
Discovery of a novel series of potent S1P1 agonists.
Bioorganic & medicinal chemistry letters, Mar-01, Volume: 20, Issue: 5
2010
Treatment with a sphingosine analog does not alter the outcome of a persistent virus infection.
Virology, Feb-20, Volume: 397, Issue: 2
2010
FTY720 therapy exerts differential effects on T cell subsets in multiple sclerosis.
Neurology, Oct-14, Volume: 71, Issue: 16
2008
Attenuation of cell motility observed with high doses of sphingosine 1-phosphate or phosphorylated FTY720 involves RGS2 through its interactions with the receptor S1P.
Genes to cells : devoted to molecular & cellular mechanisms, Volume: 13, Issue: 7
2008
The effect on heart rate of combining single-dose fingolimod with steady-state atenolol or diltiazem in healthy subjects.
European journal of clinical pharmacology, Volume: 64, Issue: 5
2008
Brain penetration of the oral immunomodulatory drug FTY720 and its phosphorylation in the central nervous system during experimental autoimmune encephalomyelitis: consequences for mode of action in multiple sclerosis.
The Journal of pharmacology and experimental therapeutics, Volume: 323, Issue: 2
2007
Oral pharmacokinetic and pharmacodynamic effects of FTY720 in cats.
Journal of veterinary pharmacology and therapeutics, Volume: 30, Issue: 1
2007
Marked suppression of tumor growth by FTY720 in a rat liver tumor model: the significance of down-regulation of cell survival Akt pathway.
International journal of oncology, Volume: 30, Issue: 2
2007
Physiologically based pharmacokinetic modeling of FTY720 (2-amino-2[2-(-4-octylphenyl)ethyl]propane-1,3-diol hydrochloride) in rats after oral and intravenous doses.
Drug metabolism and disposition: the biological fate of chemicals, Volume: 34, Issue: 9
2006
Selective sphingosine 1-phosphate 1 receptor activation reduces ischemia-reperfusion injury in mouse kidney.
American journal of physiology. Renal physiology, Volume: 290, Issue: 6
2006
FTY720: a promising agent for treatment of metastatic hepatocellular carcinoma.
Clinical cancer research : an official journal of the American Association for Cancer Research, Dec-01, Volume: 11, Issue: 23
2005
Sphingosine kinase type 2 is essential for lymphopenia induced by the immunomodulatory drug FTY720.
Blood, Feb-15, Volume: 107, Issue: 4
2006
FTY720 immunomodulation: optimism for improved transplant regimens.
Transplantation proceedings, Volume: 36, Issue: 2 Suppl
2004
FTY720: early clinical experience.
Transplantation proceedings, Volume: 36, Issue: 2 Suppl
2004
Pharmacodynamics of single doses of the novel immunosuppressant FTY720 in stable renal transplant patients.
American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons, Volume: 3, Issue: 7
2003
Efficacy of mycophenolate sodium as monotherapy and in combination with FTY720 in a DA-to-Lewis-rat heart-transplantation model.
Transplantation, Nov-27, Volume: 74, Issue: 10
2002
Effects of immunosuppressant FTY720 on renal and hepatic hemodynamics in the rat.
Transplantation, Sep-15, Volume: 74, Issue: 5
2002
First human trial of FTY720, a novel immunomodulator, in stable renal transplant patients.
Journal of the American Society of Nephrology : JASN, Volume: 13, Issue: 4
2002
Marked prevention of tumor growth and metastasis by a novel immunosuppressive agent, FTY720, in mouse breast cancer models.
Cancer research, Mar-01, Volume: 62, Issue: 5
2002
[mTOR and FTY 720 inhibitors].
Presse medicale (Paris, France : 1983), Sep-01, Volume: 30, Issue: 24 Pt 2
2001
Coexpression of CD4 and CD8alpha on rat T-cells in whole blood: a sensitive marker for monitoring T-cell immunosuppressive drugs.
Journal of immunological methods, Aug-01, Volume: 254, Issue: 1-2
2001
Long-term graft acceptance in rat heart transplantation by CTLA4Ig gene transfection combined with FTY720 treatment.
World journal of surgery, Volume: 25, Issue: 4
2001
The peripheral lymphocyte count predicts graft survival in DA to Lewis heterotopic heart transplantation treated with FTY720 and SDZ RAD.
Transplant immunology, Volume: 8, Issue: 2
2000
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Interactions (22)

ArticleYear
Lysosome‑targeted drug combination induces multiple organelle dysfunctions and non‑canonical death in pancreatic cancer cells.
Oncology reports, Volume: 47, Issue: 2
2022
PP2A activation alone and in combination with cisplatin decreases cell growth and tumor formation in human HuH6 hepatoblastoma cells.
PloS one, Volume: 14, Issue: 4
2019
Efficacy of fingolimod combined with alteplase in acute ischemic stroke and rehabilitation nursing.
Pakistan journal of pharmaceutical sciences, Volume: 32, Issue: 1(Special)
2019
Inhibition of basal-like breast cancer growth by FTY720 in combination with epidermal growth factor receptor kinase blockade.
Breast cancer research : BCR, Aug-04, Volume: 19, Issue: 1
2017
Individual variation of human S1P₁ coding sequence leads to heterogeneity in receptor function and drug interactions.
Journal of lipid research, Volume: 55, Issue: 12
2014
Anti-TCR therapy combined with fingolimod for reversal of diabetic hyperglycemia by β cell regeneration in the LEW.1AR1-iddm rat model of type 1 diabetes.
Journal of molecular medicine (Berlin, Germany), Volume: 92, Issue: 7
2014
[Therapeutic approach to mite-induced intractable dermatitis using novel immunomodulator FTY720 (fingolimod) in combination with betamethasone ointment in NC/Nga mice].
Arerugi = [Allergy], Volume: 61, Issue: 7
2012
Therapeutic approach to steroid-resistant dermatitis using novel immunomodulator FTY720 (Fingolimod) in combination with betamethasone ointment in NC/Nga mice.
Biological & pharmaceutical bulletin, Volume: 35, Issue: 8
2012
Pharmacokinetics of fingolimod (FTY720) and a combined oral contraceptive coadministered in healthy women: drug-drug interaction study results.
International journal of clinical pharmacology and therapeutics, Volume: 50, Issue: 8
2012
FTY720 combined with tacrolimus in de novo renal transplantation: 1-year, multicenter, open-label randomized study.
Nephrology, dialysis, transplantation : official publication of the European Dialysis and Transplant Association - European Renal Association, Volume: 26, Issue: 11
2011
Sirolimus in combination with FTY720: analysis of urinary and serum parameters.
Transplantation proceedings, Volume: 42, Issue: 2
2010
Ketoconazole increases fingolimod blood levels in a drug interaction via CYP4F2 inhibition.
Journal of clinical pharmacology, Volume: 49, Issue: 2
2009
Treatment of chronic allograft nephropathy at late stages using everolimus or FTY720 in combination with cyclosporine.
Transplantation proceedings, Volume: 40, Issue: 10
2008
Tacrolimus in combination with FTY720--an analysis of renal and blood parameters.
International immunopharmacology, Dec-20, Volume: 6, Issue: 13-14
2006
FTY720 in combination with cyclosporine--an analysis of skin allograft survival and renal function.
International immunopharmacology, Dec-20, Volume: 6, Issue: 13-14
2006
Fat-derived hormone adiponectin combined with FTY720 significantly improves small-for-size fatty liver graft survival.
American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons, Volume: 6, Issue: 3
2006
Efficacy of mycophenolate sodium as monotherapy and in combination with FTY720 in a DA-to-Lewis-rat heart-transplantation model.
Transplantation, Nov-27, Volume: 74, Issue: 10
2002
Long-term graft acceptance in rat heart transplantation by CTLA4Ig gene transfection combined with FTY720 treatment.
World journal of surgery, Volume: 25, Issue: 4
2001
Immunosuppressive therapy using FTY720 combined with tacrolimus in rat liver transplantation.
Surgery, Volume: 127, Issue: 1
2000
Effect of peritransplant FTY720 alone or in combination with post-transplant tacrolimus in a rat model of cardiac allotransplantation.
Transplant international : official journal of the European Society for Organ Transplantation, Volume: 11, Issue: 4
1998
Immunosuppressive effects of FTY720 alone or in combination with cyclosporine and/or sirolimus.
Transplantation, Apr-15, Volume: 65, Issue: 7
1998
An immunosuppressive regimen using FTY720 combined with cyclosporin in canine kidney transplantation.
Transplant international : official journal of the European Society for Organ Transplantation, Volume: 11, Issue: 2
1998
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]