molecular formula C19H33NO2 B1672674 Fingolimod CAS No. 162359-55-9

Fingolimod

Cat. No.: B1672674
CAS No.: 162359-55-9
M. Wt: 307.5 g/mol
InChI Key: KKGQTZUTZRNORY-UHFFFAOYSA-N
Attention: For research use only. Not for human or veterinary use.
In Stock
  • Click on QUICK INQUIRY to receive a quote from our team of experts.
  • With the quality product at a COMPETITIVE price, you can focus more on your research.

Description

Fingolimod is an aminodiol that consists of propane-1,3-diol having amino and 2-(4-octylphenyl)ethyl substituents at the 2-position. It is a sphingosine 1-phosphate receptor modulator used for the treatment of relapsing-remitting multiple sclerosis. A prodrug, this compound is phosphorylated by sphingosine kinase to active metabolite this compound-phosphate, a structural analogue of sphingosine 1-phosphate. It has a role as an immunosuppressive agent, a prodrug, an antineoplastic agent, a sphingosine-1-phosphate receptor agonist and a CB1 receptor antagonist. It is an aminodiol and a primary amino compound. It is a conjugate base of a this compound(1+).
Multiple sclerosis, or MS, is a devastating inflammatory disease that often progresses and causes severe neurological, physical, and cognitive effects. This compound is a sphingosine 1-phosphate receptor modulator for the treatment of relapsing-remitting multiple sclerosis. It was developed by Novartis and initially approved by the FDA in 2010. This compound was also studied for the treatment of COVID-19, the disease caused by infection with the SARS-CoV-2 virus.
This compound is a Sphingosine 1-phosphate Receptor Modulator. The mechanism of action of this compound is as a Sphingosine 1-Phosphate Receptor Modulator.
This compound is an orally available immunomodulatory drug used to treat relapsing multiple sclerosis. This compound is associated with transient serum enzyme elevations during therapy and with rare instances of clinically apparent, acute liver injury that can be severe and even fatal.
This compound is a natural product found in Isaria sinclairii with data available.
This compound is an orally available derivate of myriocin and sphingosine-1-phosphate receptor 1 (S1PR1, S1P1) modulator, with potential anti-inflammatory and immunomodulating activities. Upon oral administration, this compound, as a structural analogue of sphingosine, selectively targets and binds to S1PR1 on lymphocytes and causes transient receptor activation followed by S1PR1 internalization and degradation. This results in the sequestration of lymphocytes in lymph nodes. By preventing egress of lymphocytes. This compound reduces both the amount of circulating peripheral lymphocytes and the infiltration of lymphocytes into target tissues. This prevents a lymphocyte-mediated immune response and may reduce inflammation. S1PR1, a G-protein coupled receptor, plays a key role in lymphocyte migration from lymphoid tissues. This compound also shifts macrophages to an anti-inflammatory M2 phenotype, and modulates their proliferation, morphology, and cytokine release via inhibition of the transient receptor potential cation channel, subfamily M, member 7 (TRPM7).
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.
See also: this compound Hydrochloride (has salt form);  this compound Lauryl Sulfate (active moiety of).

Properties

IUPAC Name

2-amino-2-[2-(4-octylphenyl)ethyl]propane-1,3-diol
Source PubChem
URL https://pubchem.ncbi.nlm.nih.gov
Description Data deposited in or computed by PubChem

InChI

InChI=1S/C19H33NO2/c1-2-3-4-5-6-7-8-17-9-11-18(12-10-17)13-14-19(20,15-21)16-22/h9-12,21-22H,2-8,13-16,20H2,1H3
Source PubChem
URL https://pubchem.ncbi.nlm.nih.gov
Description Data deposited in or computed by PubChem

InChI Key

KKGQTZUTZRNORY-UHFFFAOYSA-N
Source PubChem
URL https://pubchem.ncbi.nlm.nih.gov
Description Data deposited in or computed by PubChem

Canonical SMILES

CCCCCCCCC1=CC=C(C=C1)CCC(CO)(CO)N
Source PubChem
URL https://pubchem.ncbi.nlm.nih.gov
Description Data deposited in or computed by PubChem

Molecular Formula

C19H33NO2
Source PubChem
URL https://pubchem.ncbi.nlm.nih.gov
Description Data deposited in or computed by PubChem

DSSTOX Substance ID

DTXSID40167363
Record name Fingolimod
Source EPA DSSTox
URL https://comptox.epa.gov/dashboard/DTXSID40167363
Description DSSTox provides a high quality public chemistry resource for supporting improved predictive toxicology.

Molecular Weight

307.5 g/mol
Source PubChem
URL https://pubchem.ncbi.nlm.nih.gov
Description Data deposited in or computed by PubChem

Solubility

Soluble
Record name Fingolimod
Source DrugBank
URL https://www.drugbank.ca/drugs/DB08868
Description The DrugBank database is a unique bioinformatics and cheminformatics resource that combines detailed drug (i.e. chemical, pharmacological and pharmaceutical) data with comprehensive drug target (i.e. sequence, structure, and pathway) information.
Explanation Creative Common's Attribution-NonCommercial 4.0 International License (http://creativecommons.org/licenses/by-nc/4.0/legalcode)

CAS No.

162359-55-9
Record name Fingolimod
Source CAS Common Chemistry
URL https://commonchemistry.cas.org/detail?cas_rn=162359-55-9
Description CAS Common Chemistry is an open community resource for accessing chemical information. Nearly 500,000 chemical substances from CAS REGISTRY cover areas of community interest, including common and frequently regulated chemicals, and those relevant to high school and undergraduate chemistry classes. This chemical information, curated by our expert scientists, is provided in alignment with our mission as a division of the American Chemical Society.
Explanation The data from CAS Common Chemistry is provided under a CC-BY-NC 4.0 license, unless otherwise stated.
Record name Fingolimod [INN]
Source ChemIDplus
URL https://pubchem.ncbi.nlm.nih.gov/substance/?source=chemidplus&sourceid=0162359559
Description ChemIDplus is a free, web search system that provides access to the structure and nomenclature authority files used for the identification of chemical substances cited in National Library of Medicine (NLM) databases, including the TOXNET system.
Record name Fingolimod
Source DrugBank
URL https://www.drugbank.ca/drugs/DB08868
Description The DrugBank database is a unique bioinformatics and cheminformatics resource that combines detailed drug (i.e. chemical, pharmacological and pharmaceutical) data with comprehensive drug target (i.e. sequence, structure, and pathway) information.
Explanation Creative Common's Attribution-NonCommercial 4.0 International License (http://creativecommons.org/licenses/by-nc/4.0/legalcode)
Record name Fingolimod
Source EPA DSSTox
URL https://comptox.epa.gov/dashboard/DTXSID40167363
Description DSSTox provides a high quality public chemistry resource for supporting improved predictive toxicology.
Record name FINGOLIMOD
Source FDA Global Substance Registration System (GSRS)
URL https://gsrs.ncats.nih.gov/ginas/app/beta/substances/3QN8BYN5QF
Description The FDA Global Substance Registration System (GSRS) enables the efficient and accurate exchange of information on what substances are in regulated products. Instead of relying on names, which vary across regulatory domains, countries, and regions, the GSRS knowledge base makes it possible for substances to be defined by standardized, scientific descriptions.
Explanation Unless otherwise noted, the contents of the FDA website (www.fda.gov), both text and graphics, are not copyrighted. They are in the public domain and may be republished, reprinted and otherwise used freely by anyone without the need to obtain permission from FDA. Credit to the U.S. Food and Drug Administration as the source is appreciated but not required.

Melting Point

102-107
Record name Fingolimod
Source DrugBank
URL https://www.drugbank.ca/drugs/DB08868
Description The DrugBank database is a unique bioinformatics and cheminformatics resource that combines detailed drug (i.e. chemical, pharmacological and pharmaceutical) data with comprehensive drug target (i.e. sequence, structure, and pathway) information.
Explanation Creative Common's Attribution-NonCommercial 4.0 International License (http://creativecommons.org/licenses/by-nc/4.0/legalcode)

Mechanisms of Action and Molecular Pharmacology

Immunological Mechanisms of Action

The primary immunological effect of fingolimod-phosphate is its interaction with S1P1 receptors on lymphocytes, which is crucial for their egress from lymphoid organs. aimspress.comscripps.edudiva-portal.orgresearchgate.netfrontiersin.org

This compound-phosphate's binding to S1P1 receptors on lymphocytes leads to the internalization and degradation of these receptors. aimspress.comfrontiersin.orgneurology.orgjsir.gr.jp This functional antagonism prevents lymphocytes from responding to the S1P gradient, which normally facilitates their exit from lymphoid tissues. aimspress.comfrontiersin.orgjsir.gr.jp

The core mechanism of this compound involves the reversible retention of T and B cells within secondary lymphoid organs, such as lymph nodes and the spleen. frontiersin.orgaimspress.comnih.govnih.govscripps.edudiva-portal.orgresearchgate.netfrontiersin.orgneurology.org This sequestration prevents these cells from re-entering the peripheral circulation. aimspress.comnih.govscripps.edudiva-portal.orgresearchgate.netfrontiersin.orgneurology.org

By trapping lymphocytes in lymphoid organs, this compound significantly reduces the number of autoreactive lymphocytes, including T and B cells, that can recirculate and infiltrate the central nervous system (CNS). aimspress.comnih.govnih.govscripps.edudiva-portal.orgfrontiersin.orgneurology.orgjsir.gr.jpplos.orgfrontiersin.orgiomcworld.orgnih.gov This reduction in CNS infiltration is a key aspect of its therapeutic benefit in autoimmune diseases like MS, where immune cells contribute to inflammation and tissue damage within the CNS. nih.govnih.govscripps.edudiva-portal.orgresearchgate.netfrontiersin.orgneurology.orgiomcworld.org

This compound exhibits a selective effect on different T-cell subpopulations. It primarily prevents the egress of C-C chemokine receptor type 7 (CCR7)-positive naïve T cells and central memory T cells (TCM) from lymph nodes. diva-portal.orgresearchgate.netfrontiersin.orgplos.orgiomcworld.orgnih.govmdpi.com In contrast, effector memory T cells (TEM), which are generally CCR7-negative, are largely unaffected and retain their ability to circulate, thereby preserving a significant portion of immune surveillance and defense against pathogens. diva-portal.orgresearchgate.netfrontiersin.orgplos.orgiomcworld.orgnih.govmdpi.com Studies have shown a pronounced reduction in naïve and central memory T cells, while effector memory cells show a less pronounced reduction. plos.org

Table 1: Differential Impact of this compound on T-cell Subpopulations

T-cell SubpopulationEffect of this compound on EgressCCR7 ExpressionFunctional Implication
Naïve T cellsSignificantly inhibitedCCR7+Retained in lymphoid organs diva-portal.orgresearchgate.netfrontiersin.orgplos.orgiomcworld.orgnih.govmdpi.com
Central Memory T cells (TCM)Significantly inhibitedCCR7+Retained in lymphoid organs diva-portal.orgresearchgate.netfrontiersin.orgplos.orgiomcworld.orgnih.govmdpi.com
Effector Memory T cells (TEM)Largely unaffectedCCR7-Continue to circulate, preserving immune surveillance diva-portal.orgresearchgate.netfrontiersin.orgplos.orgiomcworld.orgnih.govmdpi.com

This compound's active form, this compound-phosphate, acts as a functional antagonist by binding to S1P1 receptors on the surface of lymphocytes. aimspress.comscripps.eduresearchgate.netneurology.orgplos.orgfrontiersin.org This binding leads to the internalization and subsequent degradation of the S1P1 receptor, effectively reducing the number of available receptors on the cell surface. aimspress.comfrontiersin.orgneurology.orgjsir.gr.jp This process, known as receptor down-modulation, renders the lymphocytes unresponsive to the natural S1P gradient, which is essential for their migration out of lymphoid tissues. aimspress.comfrontiersin.orgjsir.gr.jp

The sequestration of lymphocytes in lymphoid organs results in a notable and dose-dependent reduction in absolute lymphocyte counts in the peripheral blood. diva-portal.orgplos.orgfrontiersin.orgnih.govfrontiersin.orgmims.comneurology.org This lymphopenia is a well-known pharmacodynamic effect of this compound. frontiersin.orgfrontiersin.orgneurology.org Studies have shown a significant decrease in total lymphocyte count, with the most pronounced reductions observed in CD4+ T helper cells and B cell populations. plos.orgfrontiersin.orgfrontiersin.org While CD8+ cytotoxic T cells are also reduced, natural killer (NK) cells are only moderately affected. plos.orgfrontiersin.orgfrontiersin.org This reduction in circulating lymphocytes is a redistribution to lymphoid tissues rather than a destruction of the cells, and it is reversible upon discontinuation of the drug. diva-portal.orgneurology.org Early immunological changes, including a decrease in absolute lymphocyte count, CD3+, CD3+CD4+, and CD19+ cells, can be detected as early as 6 hours after the first administration of this compound. frontiersin.org

Table 2: Changes in Peripheral Blood Lymphocyte Subsets Following this compound Treatment

Lymphocyte SubsetChange in Absolute Count (Example Data)Reference
Total LymphocytesMarked decrease (e.g., ~70% reduction reported) plos.orgneurology.org
CD3+ T cellsSignificant decrease frontiersin.org
CD4+ T helper cellsMost pronounced decrease plos.orgfrontiersin.orgfrontiersin.org
CD8+ T cellsReduced, but less pronounced than CD4+ T cells plos.orgfrontiersin.orgfrontiersin.org
B cellsPronounced decrease plos.orgfrontiersin.org
NK cellsModerately affected plos.orgfrontiersin.org

Preclinical Research and Animal Models

Studies in Experimental Autoimmune Encephalomyelitis (EAE) Models

Experimental autoimmune encephalomyelitis (EAE) serves as a widely accepted animal model for multiple sclerosis (MS), and preclinical studies using this model have been crucial in understanding fingolimod's efficacy and mechanisms ufrgs.brtranspharmation.com.

Efficacy in Prophylaxis and Therapy of EAE

This compound has demonstrated significant efficacy in EAE models, both when administered prophylactically (before disease onset) and therapeutically (after disease onset) ufrgs.brtranspharmation.comscripps.edutandfonline.comunipg.itnih.gov. In various mouse and rat EAE models, this compound treatment effectively reduced clinical symptoms, inflammation, and demyelination ufrgs.brtandfonline.comd-nb.infoimmunologyresearchjournal.com. It has been shown to inhibit subsequent relapses and axonal loss in the spinal cord in a relapsing-progressive EAE model in mice, facilitating motor recovery scripps.edu. Additionally, this compound reversed blood-brain barrier leakiness and improved neurological function in the DA rat model of EAE scripps.edu. Studies also indicate that prophylactic administration of this compound can lead to better disease reduction compared to therapeutic regimens researchgate.net.

The therapeutic effects observed in EAE models include:

  • Reduced Clinical Severity : Significant alleviation of clinical symptoms, such as tail paralysis and limb weakness ufrgs.brimmunologyresearchjournal.comscantox.com.
  • Reduced Inflammation : Decreased infiltration of inflammatory cells, including T cells and macrophages/microglia, into the spinal cord d-nb.infoimmunologyresearchjournal.com.
  • Reduced Demyelination : Attenuation of demyelination in the spinal cord and preservation of myelin ufrgs.brscripps.edud-nb.inforesearchgate.net.
  • Axonal Protection : Reduction of axonal loss scripps.edupnas.orgnih.gov.
  • Functional Improvement : Improved neurological function and motor coordination scripps.eduscantox.com.
  • EAE Model (Species) Treatment Regimen Key Efficacy Findings Citation
    Mouse (various) Prophylactic & Therapeutic Reduced clinical symptoms, inflammation, demyelination, axonal loss ufrgs.brscripps.edutandfonline.comd-nb.infoimmunologyresearchjournal.com
    Rat (DA) Therapeutic Reversed blood-brain barrier leakiness, improved neurological function scripps.edu
    Canine Therapeutic Resolution of clinical signs and inflammatory lesions ufrgs.br

    Confirmation of T-cell Recirculation Inhibition

    A primary mechanism of action attributed to this compound is its immunomodulatory effect, specifically the inhibition of T-cell recirculation to the CNS tandfonline.comunipg.it. This compound is a prodrug that is phosphorylated in vivo to this compound-phosphate (this compound-P) scripps.eduunipg.it. This compound-P acts as a functional antagonist of sphingosine 1-phosphate receptor 1 (S1P1), which is highly expressed on lymphocytes scripps.eduaimspress.comneurology.org. This binding induces the internalization and degradation of S1P1 receptors on lymphocytes, particularly naive and central memory T cells tandfonline.comunipg.itresearchgate.net. Consequently, it prevents their egress from lymph nodes and thymus, leading to a reduction in circulating lymphocytes and limiting their infiltration into the CNS scripps.edutandfonline.comunipg.itaimspress.comresearchgate.net.

    Preclinical studies in EAE models have confirmed this mechanism:

  • This compound significantly inhibits the development of EAE and markedly reduces the infiltration of encephalitogenic T cells, including Th17 and Th1 cells, into the spinal cords of EAE mice aimspress.com.
  • The prophylactic effects of this compound on EAE are largely due to the inhibition of S1P1-dependent egress of autoreactive T cells from draining lymph nodes aimspress.com.
  • Conditional deletion of S1P1 receptors in T lymphocytes and hematopoietic cells has been shown to mimic the in vivo this compound effect by decreasing the counts of circulating T cells tandfonline.comresearchgate.net.
  • Assessment of Direct CNS Effects in EAE Models

    Beyond its well-established immunomodulatory effects, emerging evidence from preclinical studies indicates that this compound exerts direct effects within the CNS scripps.edunih.govoup.com. This compound, being lipophilic, can cross the blood-brain barrier and its active metabolite, this compound-P, has been detected in the cerebrospinal fluid scripps.edu.

    Direct CNS effects observed in EAE models include:

  • Neuroprotection : this compound has shown neuroprotective effects, reducing injury to oligodendrocytes, myelin, and axons scripps.eduresearchgate.netmdpi.com. It has been suggested to promote myelin preservation and repair, although some studies indicate it attenuates injury rather than promoting remyelination directly scripps.edu.
  • Astrogliosis Attenuation : this compound treatment reduced astrogliosis, a histopathological feature of EAE, and the production of proinflammatory cytokines by astrocytes tandfonline.comd-nb.infooup.com.
  • Microglia Modulation : It reduces microglial activation tandfonline.com.
  • Neuronal Protection : this compound reduces dendritic spine loss and protects neurons from excitotoxic cell death tandfonline.comoup.com. It can also restore the permeability of the blood-brain barrier in EAE rats tandfonline.com.
  • Glutamate Modulation : this compound has been proposed to modulate glutamate transmission in the CNS of EAE mice researchgate.netresearchgate.net.
  • Role of Astrocyte S1P1 Deficiency in this compound Efficacy in EAE

    Crucially, preclinical studies have identified a significant role for S1P1 receptors expressed on astrocytes in the efficacy of this compound in EAE scripps.edupnas.orgnih.govaimspress.comoup.comresearchgate.neteneuro.orgcambridge.org. While this compound-P is known to modulate S1P1 on lymphocytes, studies using conditional null mouse mutants have revealed that the efficacy of this compound in EAE is lost or significantly attenuated in animals lacking S1P1 on glial fibrillary acidic protein (GFAP)-expressing astrocytes, but not on neurons scripps.edupnas.orgnih.govaimspress.comresearchgate.neteneuro.orgcambridge.org. This occurs despite maintained normal lymphocyte trafficking and S1P-mediated immune activities in these mutants scripps.edunih.gov.

    Key findings include:

  • EAE severity was attenuated in mutants lacking S1P1 on astrocytes compared to controls scripps.edunih.gov.
  • Reductions in EAE clinical scores were paralleled by reductions in demyelination, axonal loss, and astrogliosis in these astrocyte-specific S1P1 deficient mutants pnas.orgnih.govresearchgate.net.
  • Functional antagonism of S1P1 on astrocytes by this compound-P is considered a primary non-immunological CNS mechanism contributing to this compound's efficacy scripps.edupnas.orgnih.govaimspress.comresearchgate.neteneuro.org.
  • This suggests that S1P signaling pathways within the CNS, particularly involving astrocytes, are important targets for this compound's therapeutic effects pnas.orgnih.gov.
  • Investigations in Other Neurodegenerative Disease Models

    The observed direct CNS effects of this compound in EAE models have prompted investigations into its potential therapeutic utility in other neurodegenerative diseases, where neuroinflammation and neuronal damage are key pathological features nih.govd-nb.info.

    Alzheimer's Disease Models (Reduction of Amyloid-β Production, Microglia/Astrocyte Activation)

    This compound has shown promising neuroprotective effects in various animal models of Alzheimer's disease (AD), including the 5xFAD transgenic mouse model, 3xTg-AD mice, and models involving intracerebral Aβ injections nih.govd-nb.infoharvard.eduuio.nonih.govresearchgate.net.

    Key findings in AD models include:

  • Reduction of Amyloid-β (Aβ) Pathology : this compound treatment has been shown to decrease amyloid-beta plaque density and reduce levels of both soluble and insoluble Aβ nih.govd-nb.infoharvard.eduuio.noresearchgate.net. In intracerebral Aβ injection models, it reduced Aβ concentration and neuronal loss nih.govd-nb.info.
  • Modulation of Microglia and Astrocyte Activation : this compound attenuates neuroinflammatory signs by decreasing the number of activated microglia (e.g., Iba1-positive cells) and astrocytes (e.g., GFAP staining) nih.govd-nb.infoharvard.eduuio.nonih.govresearchgate.netnih.gov. In 3xTg-AD mice, it reduced microglial cell number and promoted a resting microglial morphology nih.gov. It also inhibits the secretion of pro-inflammatory molecules from astrocytes nih.gov.
  • Cognitive Improvement : These pathological improvements are often accompanied by an attenuation of memory and learning deficits and cognitive improvement in AD animal models oup.comnih.govd-nb.infoharvard.edunih.govresearchgate.netnih.gov.
  • Neurotrophic Factor Induction : this compound has been correlated with increased brain-derived neurotrophic factor (BDNF) levels in AD models, which may contribute to its neuroprotective effects and improved disease phenotype nih.govuio.nonih.govnih.gov.
  • The efficacy of this compound in AD models can be time- and sex-dependent, with some studies showing reduced Aβ load and microglia activation primarily in male animals with late treatment initiation uio.no.

    AD Model (Species) Key Pathological Hallmarks Addressed Observed Effects Citation
    5xFAD Mouse Aβ plaques, Soluble/Insoluble Aβ, Microglia/Astrocyte activation Decreased plaque density, Aβ levels, GFAP staining, activated microglia nih.govd-nb.infoharvard.edu
    3xTg-AD Mouse Microglial activation, Phosphorylated tau, APP levels Reduced Iba1-positive cells, less ramified microglia, reduced phosphorylated tau and APP nih.gov
    Intracerebral Aβ Injection Models Neuronal loss, Aβ concentration, Caspase-3 activation Reduced neuronal loss, Aβ concentration, attenuated memory/learning deficits nih.govd-nb.info

    Clinical Research and Therapeutic Efficacy Excluding Dosage/administration

    Efficacy in Relapsing-Remitting Multiple Sclerosis (RRMS)

    Reduction in Annualized Relapse Rate (ARR)

    Fingolimod has shown significant efficacy in reducing the Annualized Relapse Rate (ARR) in patients with RRMS. In the 24-month placebo-controlled FREEDOMS trial, this compound 0.5 mg significantly reduced the ARR to 0.18 compared to 0.40 with placebo (p < 0.001), representing a 54% relative reduction. neurology.orgnih.govfrontiersin.org The FREEDOMS II trial also reported a 48% relative reduction in ARR at 24 months compared to placebo. fiercepharma.com

    In the 12-month TRANSFORMS study, this compound 0.5 mg demonstrated superior efficacy over intramuscular interferon-β-1a, reducing the ARR by 52% at 1 year. neurology.orgnih.govfrontiersin.org Long-term extensions of these trials have indicated that these reductions in ARR are sustained with continued this compound treatment. For instance, in the extension of the TRANSFORMS study, the ARR in patients continuously on this compound 0.5 mg was significantly lower (0.17) compared to those who switched from interferon-β-1a (0.27) over the long-term observation period. Patients switching from interferon-β-1a to this compound experienced a 50% reduction in ARR (from 0.40 to 0.20) after the switch. bmj.com

    Table 1: Annualized Relapse Rate (ARR) Reduction in Key Clinical Trials

    Study (Duration)This compound 0.5 mg ARRComparator ARRRelative Reduction (%)p-value
    FREEDOMS (24 months) neurology.orgnih.gov0.180.40 (Placebo)54%< 0.001
    FREEDOMS II (24 months) fiercepharma.com-- (Placebo)48%< 0.0001
    TRANSFORMS (12 months) neurology.orgnih.gov0.160.33 (IM IFNβ-1a)52%< 0.001
    TRANSFORMS Extension (up to 4.5 years) bmj.com0.17 (Continuous)0.27 (IFN-switch)35% (vs IFN-switch)< 0.001

    Impact on Disability Progression

    This compound has demonstrated a significant impact on delaying disability progression in patients with RRMS. In the FREEDOMS trial, this compound 0.5 mg reduced the risk of disability progression by 30% (p = 0.02) confirmed after 3 months and by 37% (p = 0.01) confirmed after 6 months compared to placebo over 2 years. frontiersin.org Long-term data from the ACROSS study, an observational study evaluating 10-year disability outcomes, showed that patients continuously treated with this compound had significantly lower disability progression compared to those whose treatment was interrupted. novartis.com

    Table 2: Impact on Disability Progression in the FREEDOMS Trial

    OutcomeThis compound 0.5 mgPlaceboRelative Risk Reduction (%)p-value
    Risk of Disability Progression (3-month confirmed) frontiersin.org--30%0.02
    Risk of Disability Progression (6-month confirmed) frontiersin.org--37%0.01
    Confirmed Disability Progression (CDP)

    Specifically, regarding Confirmed Disability Progression (CDP), the FREEDOMS trial showed that this compound 0.5 mg significantly reduced the risk of 3-month CDP. neurology.orgnih.gov In the extension of the FREEDOMS study, a lower proportion of patients in the continuous-fingolimod groups experienced 3-month CDP compared to a group comprising all placebo-fingolimod patients. neurology.org The risk of 3-month CDP was reduced by 27% and 6-month CDP by 31% in the continuous this compound 0.5 mg group compared to the combined switch group. neurology.orgnih.gov

    Table 3: Confirmed Disability Progression (CDP) in FREEDOMS Extension

    OutcomeContinuous this compound 0.5 mg (Proportion Free) neurology.orgCombined Switch Group (Proportion Free) neurology.orgHazard Ratio (HR) neurology.orgp-value neurology.org
    3-month CDP74%66%0.730.0189
    6-month CDP80%73%0.690.0140

    Magnetic Resonance Imaging (MRI) Outcomes

    This compound has consistently shown significant benefits on various Magnetic Resonance Imaging (MRI) outcomes, indicating a reduction in inflammatory disease activity and brain volume loss. neurology.orgnih.govfrontiersin.orgsmw.ch

    Reduction in New or Enlarged T2 Lesions

    This compound treatment leads to a substantial reduction in the number of new or enlarged T2 lesions. In the FREEDOMS study, this compound 0.5 mg significantly reduced the number of new or enlarged lesions on T2-weighted images (p < 0.001). neurology.orgnih.gov After 2 years, the relative reduction in new or enlarged T2-lesions was 74% compared to placebo. frontiersin.org In the TRANSFORMS study, this compound 0.5 mg reduced new or enlarged T2 lesions by 35% at 12 months compared to intramuscular interferon-β-1a. frontiersin.org

    Table 4: Reduction in New or Enlarged T2 Lesions

    Study (Duration)This compound 0.5 mg (Relative Reduction)Comparator (Relative Reduction)p-value
    FREEDOMS (2 years) frontiersin.org74% vs Placebo-< 0.001
    TRANSFORMS (12 months) frontiersin.org35% vs IM IFNβ-1a-< 0.004
    Reduction in Gadolinium-Enhanced Lesions (GELs)

    This compound also significantly reduces the number of Gadolinium-Enhanced Lesions (GELs), which are indicative of active inflammation. In the FREEDOMS trial, this compound 0.5 mg significantly reduced Gd-enhancing lesions (p < 0.001). neurology.orgnih.gov After 2 years, the relative reduction in Gd-enhancing T1 lesions was 82% compared to placebo. frontiersin.org In the TRANSFORMS study, GELs were reduced by 55% in this compound 0.5 mg-treated patients compared to those treated with intramuscular interferon-β-1a.

    Table 5: Reduction in Gadolinium-Enhanced Lesions (GELs)

    Study (Duration)This compound 0.5 mg (Relative Reduction)Comparator (Relative Reduction)p-value
    FREEDOMS (2 years) frontiersin.org82% vs Placebo-< 0.001
    TRANSFORMS (12 months) 55% vs IM IFNβ-1a-< 0.001
    Preservation of CNS Tissue

    Long-Term Efficacy Studies

    Persistence and Adherence in Real-World Settings

    Real-world studies have demonstrated that this compound exhibits higher rates of persistence and adherence compared to injectable and infusible disease-modifying therapies (DMTs) used for multiple sclerosis. A retrospective analysis of a US claims database indicated that patients treated with this compound had significantly higher rates of persistence and adherence than those on glatiramer acetate, interferons, and natalizumab tandfonline.com. The risk of discontinuing treatment was notably lower with this compound across these comparisons tandfonline.com. For instance, one study reported that 72.1% of patients in the this compound cohort remained persistent on their index therapy after 360 days, compared to 60.5% for glatiramer acetate, 56.3% for interferon, and 61% for natalizumab jmcp.org. Furthermore, a long-term, non-interventional study in Hungary observed a treatment persistence rate of 73.4% for this compound after 60 months plos.org. These findings suggest that the once-daily oral administration of this compound may contribute to improved medication-taking behaviors in clinical practice tandfonline.com.

    Table 1: Persistence Rates of this compound vs. Other DMTs (12 Months)

    DMT CategoryPersistence Rate (%) jmcp.org
    This compound72.1
    Glatiramer Acetate60.5
    Interferon56.3
    Natalizumab61.0

    Comparison with Other Disease-Modifying Therapies (DMTs)

    This compound has been extensively compared with other established DMTs, revealing distinct efficacy profiles.

    This compound has shown superior efficacy when compared to interferons in reducing disease activity in both adult and pediatric MS populations. In the TRANSFORMS study, a 12-month Phase 3 trial, this compound demonstrated greater efficacy on relapse rates and MRI outcomes compared to interferon beta-1a nih.gov. An extension of this study confirmed that switching from interferon beta-1a to this compound led to enhanced efficacy nih.gov. In a real-world setting, a retrospective US claims database analysis found that, after adjusting for baseline differences, this compound was associated with a 52% reduction in the probability of having a relapse and a 50% reduction in annualized relapse rate (ARR) compared with combined interferon/glatiramer acetate treatment tandfonline.com. Specifically, the adjusted ARR was 0.32 for this compound compared to 0.64 for the interferon/glatiramer acetate cohort tandfonline.com. For pediatric MS, the PARADIGMS study demonstrated an 82% superiority of this compound over interferon beta-1a in reducing relapses over a 2-year period, with an adjusted ARR of 0.12 for this compound versus 0.67 for interferon beta-1a dovepress.comneurologylive.com2minutemedicine.com. This compound also showed superiority in reducing new and enhancing or enlarging lesions on MRI and in reducing brain volume loss neurologylive.com.

    Table 2: Efficacy Comparison: this compound vs. Interferon Beta-1a in Pediatric MS (PARADIGMS Study) dovepress.comneurologylive.com2minutemedicine.com

    Efficacy MeasureThis compound (0.5 mg)Interferon Beta-1aRelative Reduction (this compound vs. IFN)
    Adjusted Annualized Relapse Rate (ARR)0.120.6782%
    Median Time to First Relapse (days)720488N/A
    Patients Relapse-Free at 24 Months (%)85.738.8N/A
    Mean Number of Gadolinium-Enhancing Lesions0.441.2863%
    Annualized Rate of New or Enlarging T2 Lesions4.399.2753%

    In head-to-head comparisons, this compound has demonstrated superior efficacy over glatiramer acetate. The Phase IIIb ASSESS study, which evaluated this compound (0.5 mg and 0.25 mg) versus glatiramer acetate (20 mg) in relapsing-remitting MS (RRMS) patients, found that this compound 0.5 mg significantly reduced the annualized relapse rate (ARR) compared to glatiramer acetate novartis.commultiplesclerosisnewstoday.commdedge.comneurology.org. Treatment with this compound 0.5 mg resulted in a 40.7% relative reduction in the rate of relapses over one year compared to glatiramer acetate (ARR estimates of 0.153 vs. 0.258, respectively) novartis.commdedge.comneurology.org. Both this compound doses (0.5 mg and 0.25 mg) also significantly reduced new or newly enlarging T2 and gadolinium-enhancing T1 lesions compared with glatiramer acetate multiplesclerosisnewstoday.comneurology.orgnih.gov.

    Table 3: Efficacy Comparison: this compound vs. Glatiramer Acetate (ASSESS Study) novartis.commdedge.comneurology.org

    Efficacy MeasureThis compound (0.5 mg)Glatiramer Acetate (20 mg)Relative Reduction (this compound vs. GA)
    Annualized Relapse Rate (ARR)0.1530.25840.7%
    Reduction in New/Newly Enlarged T2 Lesions (%)54.4N/AN/A
    Reduction in Gadolinium-Enhancing T1 Lesions (%)55.6N/AN/A

    The comparative efficacy between this compound and natalizumab in relapsing-remitting MS has been a subject of several studies, with some suggesting natalizumab may be more efficacious, especially in highly active disease. A prospective, randomized head-to-head study (REVEAL) indicated that natalizumab was more efficacious than this compound in reducing MS relapses and T1 gadolinium-enhancing (Gd+) lesion accumulation in patients with active disease bmj.comnih.gov. In this study, the cumulative probability of relapse over follow-up was 1.9% with natalizumab versus 22.3% with this compound bmj.comnih.gov. The on-treatment ARR was 0.02 in the natalizumab group (a 99% reduction) and 0.20 in the this compound group (an 89% reduction) bmj.com. Another analysis from the MSBase observational cohort study, involving patients who switched from injectable DMTs due to continued disease activity, found that while both agents reduced relapse activity and disability, natalizumab was more effective, with a decrease in ARR from 1.5 to 0.2 for natalizumab and from 1.3 to 0.3 for this compound mscare.org. However, some real-world systematic reviews have noted inconsistent trends, with some studies reporting similar effectiveness between this compound and natalizumab researchgate.net.

    Table 4: Efficacy Comparison: Natalizumab vs. This compound bmj.commscare.org

    Efficacy MeasureNatalizumabThis compound
    Cumulative Probability of Relapse (%) bmj.com1.922.3
    On-Treatment Annualized Relapse Rate (ARR) bmj.com0.020.20
    Reduction in Pre-treatment ARR mscare.org1.5 to 0.21.3 to 0.3
    Glatiramer Acetate

    Efficacy in Pediatric MS Patients

    This compound holds a unique position as the only FDA-approved disease-modifying therapy for pediatric MS patients, specifically for those aged 10 to 17 years dovepress.comneurologylive.commdpi.com. The pivotal Phase 3 PARADIGMS study demonstrated its robust efficacy in this population. The study showed a significant and clinically meaningful reduction in the annualized relapse rate (ARR) in children and adolescents with MS over a period of up to two years, compared to interferon beta-1a intramuscular injections dovepress.comnovartis.com. The adjusted ARR was 0.12 in the this compound-treated group, meaning these patients experienced a relapse less than every 8 years, a dramatic difference compared to the interferon beta-1a group where the relapse rate was significantly higher neurologylive.com. This compound was also superior in reducing the number of new and enhancing or enlarging lesions on MRI, as well as in reducing brain volume loss or atrophy neurologylive.com. Beyond clinical and radiological outcomes, this compound also improved health-related quality of life (HRQoL) in pediatric patients with MS, as evidenced by both self-reported and parent-reported Pediatric Quality of Life (PedsQL) scores bmj.com.

    Biomarkers of Efficacy (e.g., Neurofilaments in CSF)

    Biomarkers play an increasingly important role in assessing disease activity and treatment efficacy in multiple sclerosis. Neurofilament light chain (NfL), a neuronal structural protein, has emerged as a promising biomarker for axonal injury in MS bmj.comnih.goveasp.esfrontiersin.org. Elevated levels of NfL in cerebrospinal fluid (CSF) correlate with MS activity and reflect axonal damage bmj.comfrontiersin.org. Studies have shown that this compound treatment can lead to a reduction in CSF NfL levels, indicating a beneficial effect on axonal injury nih.govucl.ac.uk. For instance, in a subset of patients from the FREEDOMS study, median changes from baseline in CSF NfL levels after 12 months of this compound treatment (0.5 mg and 1.25 mg) were significantly lower than zero, unlike the placebo group nih.gov. This reduction in NfL levels correlated with improvements in relapse and MRI outcomes nih.gov. Beyond NfL, other CSF biomarkers such as C-X-C motif chemokine 13 (CXCL13), a marker of B-cell activity, and chitinase-3-like protein 1 (CHI3L1) and chitotriosidase (CHIT1), markers of glial activation, have also been observed to decrease after this compound treatment, particularly in patients switching from first-line DMTs ucl.ac.uk. These findings suggest that CSF biomarkers can provide an additional measure of treatment efficacy by reflecting the influence of DMTs on different aspects of MS immunopathogenesis ucl.ac.uk.

    Table 5: Effect of this compound on CSF Biomarkers nih.govucl.ac.uk

    BiomarkerEffect of this compound TreatmentIndication
    Neurofilament Light Chain (NfL)Decreased levelsReduced axonal injury and disease activity
    C-X-C motif chemokine 13 (CXCL13)Decreased levelsReduced B-cell activity
    Chitinase-3-like protein 1 (CHI3L1)Decreased levelsReduced glial activation
    Chitotriosidase (CHIT1)Decreased levelsReduced glial activation

    Adverse Event Mechanisms and Management Excluding Dosage/administration

    Cardiac Adverse Event Mechanisms

    Bradycardia

    Initiation of fingolimod treatment commonly leads to a transient decrease in heart rate, with the maximal decline typically occurring within 6 hours of the first dose. europa.eueuropa.eu This effect is attributed to the activation of inwardly rectifying Gα1-protein-regulated potassium channels (GIRK/IKACh) in atrial myocytes and endothelial cells, a mechanism similar to vagally-mediated cardiac effects. nih.gov This results in inhibited cardiac pacemaker activity. nih.gov The bradycardia is usually transient and often asymptomatic, with heart rate generally returning towards baseline within one month of continued administration. europa.eueuropa.eu In cases where significant symptoms develop, the bradycardia induced by this compound can be reversed by parenteral administration of atropine or isoprenaline. revportcardiol.orgnih.goveuropa.eueuropa.eu

    Atrioventricular Block

    This compound can also cause delays in atrioventricular conduction, including the occurrence of AV block. revportcardiol.orgeuropa.eueuropa.eu This is also related to the modulation of S1P receptors in the AV node. revportcardiol.org First-degree AV block (prolonged PR interval) has been observed, and less frequently, second-degree AV block. revportcardiol.orgeuropa.eu Isolated reports of transient, spontaneously resolving complete AV block have also occurred, typically within the first 24 hours after the initial dose. europa.eueuropa.eu These conduction abnormalities are generally transient and asymptomatic, often resolving without intervention within the first 24 hours of treatment. revportcardiol.orgeuropa.eu The mechanism may involve the activation of GIRK channels and potentially L-type calcium channel blockade effects. researchgate.netresearchgate.net

    Hypertension

    A modest increase in blood pressure has been observed in some patients treated with this compound, typically detected around one month after treatment initiation and persisting with continued therapy. europa.eu This effect is thought to be related to the functional antagonism of S1PR1, which favors the binding of S1P to S1PR2 and S1PR3 in arterial smooth muscle cells. revportcardiol.org Activation of S1PR2 and S1PR3 can lead to increased intracellular calcium concentrations and subsequent vascular contraction, contributing to increased vascular tone and blood pressure. revportcardiol.orgnih.gov

    Ocular Adverse Event Mechanisms

    Ocular adverse events, particularly macular edema, are a recognized complication of this compound therapy. nih.goveyewiki.org

    Macular Edema (FAME) and its Risk Factors (e.g., Uveitis, Diabetes Mellitus)

    This compound-associated macular edema (FAME) is characterized by the accumulation of fluid in the layers of the retina, primarily in the outer plexiform and inner nuclear layers. eyewiki.org The exact mechanism is not fully elucidated, but it is likely related to the disruption of the inner blood-retinal barrier (BRB). eyewiki.orgfrontiersin.orgnih.gov this compound's active metabolite, this compound-phosphate, binds to S1P receptors, including those on vascular endothelial cells. eyewiki.orgfrontiersin.org Antagonism of S1P1 receptors by this compound may decrease pericytes and disrupt tight junctions between endothelial cells, increasing vascular permeability at the BRB and leading to fluid leakage and edema formation. eyewiki.orgresearchgate.net

    Several factors have been identified that increase the risk of developing FAME. These include a history of uveitis and diabetes mellitus. eyewiki.orgresearchgate.netnih.govnih.govdroracle.ai In conditions like diabetes and uveitis, there may be pre-existing inflammation or compromise of the retinal vasculature, which could be exacerbated by this compound's effects on vascular permeability. nih.gov Other risk factors include patient age (particularly >41 years) and a history of retinal vascular disease or past ocular surgery. eyewiki.orgnih.gov

    Retinal Hemorrhages and Retinal Vein Occlusion

    While less common than macular edema, retinal hemorrhages and retinal vein occlusion have also been reported in patients receiving this compound. nih.govnih.gov The mechanism underlying these events may also involve the effects of this compound on vascular integrity and permeability, similar to the proposed mechanism for FAME. nih.govnih.gov Disruption of the blood-retinal barrier and increased vascular permeability could potentially predispose individuals to vascular events such as hemorrhages and occlusions. nih.gov

    Dermatological Adverse Event Mechanisms

    Malignant Melanoma

    Cases of malignant melanoma have also been reported in patients treated with this compound, although the incidence in clinical trials was rare. nih.govnih.govmdpi.com Similar to other skin cancers, the mechanism by which this compound might contribute to melanoma development is not clearly understood. nih.govnih.gov Some research suggests that while this compound has shown anti-tumor effects in certain melanoma models in vitro and in vivo, acting as an antiangiogenic drug and inhibiting tumor growth and metastatic spreading, a protumorigenic role has also been proposed, potentially by enhancing the accumulation of myeloid-derived suppressor cells (MDSCs) around the tumor lesion. nih.govmdpi.com In vitro studies have shown varied effects on melanoma cell lines, with both inhibition and promotion of cell migration depending on the cell line examined. mdpi.com

    Management of malignant melanoma in patients on this compound involves prompt diagnosis and appropriate oncological treatment. Enhanced dermatological monitoring is recommended for patients receiving this compound to facilitate early detection of skin cancers, including melanoma. nih.gov Physicians should discuss the possible risk of melanoma with patients when initiating this compound treatment. nih.gov

    Other Systemic Adverse Event Mechanisms (e.g., Liver Enzyme Elevation)

    Elevated liver enzyme levels, particularly alanine transaminase (ALT) and aspartate transaminase (AST), are commonly observed adverse events associated with this compound treatment. nih.gove-century.us In large clinical trials, serum ALT elevations above 3 times the upper limit of normal (ULN) were reported more frequently in this compound recipients compared to placebo recipients. nih.goveuropa.eu While the exact mechanism by which this compound causes liver injury is not fully known, it is extensively metabolized by the liver, primarily via the cytochrome P450 system, and liver injury may potentially be caused by a toxic or immunogenic intermediate of its metabolism. nih.gov In vitro studies using human hepatocellular carcinoma cell lines (HepG2) suggest that the active form, this compound (S)-phosphate, can induce time- and dose-dependent decreases in cell viability, mitochondrial membrane potential, and ATP levels, along with the induction of oxidative stress. oup.comlarvol.com It is hypothesized that this toxic profile may originate from the interaction of this compound (S)-phosphate with sirtuin proteins, specifically SIRT3 and SIRT5. oup.comlarvol.com

    Management of liver enzyme elevation involves monitoring liver function tests. Routine liver tests are recommended before initiating treatment and at specified intervals thereafter, and periodically until two months after discontinuation. nih.goveuropa.eu More frequent monitoring is advised if serum enzyme levels exceed 3 times the ULN. nih.gov If there are symptoms or signs of liver injury or if aminotransferase levels rise above 5 times ULN, discontinuation of therapy is recommended. nih.goveuropa.eu Liver enzyme levels typically return to normal within approximately two months after discontinuing this compound. nih.gov In cases of confirmed significant liver injury, treatment should be discontinued and generally not resumed unless an alternative cause can be established. europa.euwww.gov.uk Screening for hepatitis B virus (HBV) infection is also recommended before starting therapy, as this compound can cause reactivation of hepatitis B in susceptible patients. nih.gov

    Other systemic adverse events with known or hypothesized mechanisms include:

    Bradycardia and Atrioventricular Block: this compound phosphate binds to S1P receptors in cardiac tissue, including the sinoatrial and atrioventricular nodes and atrial myocytes. This binding can induce inward potassium flow, leading to a negative chronotropic effect on the heart, resulting in transient bradycardia and AV conduction delays, typically observed upon treatment initiation. nih.govmedcentral.com

    Macular Edema: A dose-dependent increase in the risk of macular edema has been observed with this compound. medcentral.com While the precise mechanism is not fully understood, S1P receptors are present in the retina, and modulation of these receptors by this compound may affect vascular permeability and fluid balance, contributing to macular edema. scripps.edu Ophthalmologic evaluations are recommended at baseline and after treatment initiation, with more frequent checks for patients with risk factors like diabetes mellitus or a history of uveitis. europa.eu

    Infections: The immunosuppressive properties of this compound, primarily due to lymphocyte sequestration in lymph nodes, increase the risk of various infections, including viral, bacterial, and fungal opportunistic infections. nih.gov This is a direct consequence of its intended mechanism of reducing circulating lymphocytes. bmj.comdrugbank.com Management involves monitoring for signs and symptoms of infection and considering therapy interruption in case of serious infections. sfda.gov.sa

    Future Directions and Research Gaps

    Refining Understanding of Precise Mechanisms of Action

    While Fingolimod's main pharmacological effect is immunomodulation through lymphocyte homing, its full spectrum of action is still being elucidated. nih.gov

    Beyond its well-known S1P receptor modulation, emerging evidence suggests that this compound may exert effects through mechanisms independent of S1P receptors. larvol.complos.org These non-S1P receptor mediated effects could contribute to its diverse biological functions, including potential anti-tumor activities, angiogenesis inhibition, calcium mobilization, and apoptosis. plos.org Candidate cellular targets for these effects include cPLA2, 14-3-3, PKC, and PP2A. plos.org Furthermore, this compound has been shown to inhibit S1P lyase activity, an enzyme responsible for the irreversible degradation of S1P, which could alter S1P chemotactic gradients and influence various cellular functions like survival, migration, and inflammation. nih.gov A deeper understanding of these non-S1P receptor mediated pathways could uncover additional therapeutic avenues and explain some of its broader pharmacological effects.

    Clarifying the Role of S1P1 Modulation in CNS vs. Immune System

    Exploring Novel Therapeutic Applications Beyond MS

    The multifaceted actions of this compound, including its neuroprotective and anti-inflammatory properties, have generated significant interest in exploring its therapeutic potential beyond MS. nih.govlarvol.comfrontiersin.org Preclinical studies have shown promising results in various neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), and epilepsy. nih.govfrontiersin.orgresearchgate.net this compound has demonstrated neuroprotective effects in animal models, correlating with increased brain-derived neurotrophic factor (BDNF) levels and improved cognitive and/or motor abilities. nih.govfrontiersin.orgresearchgate.net It has also been shown to reduce neuroinflammatory markers by inhibiting lymphocytes and directly affecting astrocytes and microglia. nih.govresearchgate.net Specific effects observed include the reduction of amyloid-beta production in AD models and antiepileptogenic properties. nih.govresearchgate.net Beyond neurodegenerative conditions, this compound is being investigated for other autoimmune and inflammatory disorders, including plaque psoriasis, dermatomyositis, Crohn's disease, ulcerative colitis, polymyositis, liver failure, renal failure, acute stroke, graft-versus-host disease (GVHD), and transplant rejection. biomolther.org Its potential in organ transplantation, oncological conditions, viral infections, and hypersensitivity diseases is also under review. larvol.com

    Development of More Selective S1P Receptor Modulators

    This compound is a non-selective S1P receptor modulator, binding with high affinity to S1P1, S1P3, S1P4, and S1P5 receptors. tandfonline.comnih.govresearchgate.net While this broad activity contributes to its efficacy, it is also associated with certain adverse effects, particularly cardiac-related side effects like bradycardia, which necessitate first-dose monitoring. nih.govnih.govresearchgate.net Consequently, a significant area of research focuses on developing more selective S1P receptor modulators, primarily targeting S1P1, to minimize off-target effects and improve safety profiles. biomolther.orgnih.govresearchgate.netnih.gov Second-generation S1PR modulators, such as Siponimod, Ozanimod, and Ponesimod, have been developed with greater selectivity for S1P1 and improved pharmacokinetic profiles, including shorter half-lives and no requirement for phosphorylation for activation, potentially leading to a faster onset of action and quicker reversal of pharmacological effects if needed. biomolther.orgnih.govnih.govresearchgate.net These newer agents aim to retain efficacy while reducing safety concerns associated with broader S1PR modulation. nih.govresearchgate.net

    Long-Term Safety and Efficacy in Diverse Patient Populations

    While clinical trials have demonstrated this compound's efficacy and safety in the short to medium term, ongoing research is crucial to assess its long-term safety and effectiveness in diverse patient populations. tandfonline.comneurology.orgnih.gov Real-world data studies and long-term extensions of pivotal trials continue to provide valuable insights into the sustained benefits of this compound in reducing relapse rates, disability progression, and brain volume loss. neurology.orgnih.govfrontiersin.orgd-nb.info These studies also monitor for late-onset safety events and assess the drug's profile in specific subgroups, such as young adults and pediatric patients. neurology.orgfrontiersin.org Understanding the impact of comorbidities and prior treatments on this compound outcomes in varied patient demographics remains an important area of investigation. frontiersin.org

    Patient-Reported Outcomes and Quality of Life Studies

    Patient-reported outcomes (PROs) and quality of life (QoL) are increasingly recognized as crucial measures of treatment success, reflecting the patient's perspective on their health status and the impact of therapy. Studies investigating this compound have consistently demonstrated positive effects on these parameters.

    In the phase 3 PARADIGMS Study, which evaluated this compound in pediatric patients with multiple sclerosis (PoMS), treatment with this compound showed improvements compared to interferon β-1a on the 23-item Pediatric Quality of Life (PedsQL) scale bmj.com. Specifically, self-reported Total Scale Scores improved by 4.66 with this compound versus -1.16 with interferon β-1a (p≤0.001). Parent-reported Total Scale Scores also favored this compound (2.71 vs -1.02, p≤0.05) bmj.com. The proportion of patients achieving a clinically meaningful improvement in the PedsQL Total Scale Score was notably higher with this compound (47.5%) compared to interferon β-1a (24.2%) based on self-reported scores (p=0.001) bmj.com.

    Table 1: PedsQL Total Scale Score Changes in PARADIGMS Study

    Outcome MeasureThis compound (Change from Baseline)Interferon β-1a (Change from Baseline)p-valueProportion with Clinically Meaningful Improvement (this compound)Proportion with Clinically Meaningful Improvement (IFN β-1a)p-value
    Self-reported Total Scale Score4.66-1.16≤0.00147.5%24.2%0.001
    Parent-reported Total Scale Score2.71-1.02≤0.0537.8%24.7%Non-significant

    Real-world evidence further supports these findings. A prospective observational study in Greece indicated that this compound treatment contributed to high patient-reported treatment satisfaction and improvements in the QoL of MS patients nih.gov. Significant increases were observed in patients' global satisfaction and effectiveness domain scores from enrollment to 24 months nih.gov. Similarly, a survey found that the majority of patients treated with this compound reported an improvement in their QoL and an enhanced capacity to plan for the future dovepress.com. These perceptions were more frequent among patients satisfied with their treatment, aligning with findings from other interventional and observational studies dovepress.com. The EPOC (Evaluate Patient OutComes) study, a phase 4, open-label, multi-center study, also aimed to assess patient-reported treatment satisfaction, perceived effectiveness, activities of daily living, fatigue, and depression in patients switching to this compound tandfonline.com.

    Combined Clinical Outcomes and Overall Treatment Effectiveness

    This compound has demonstrated robust effectiveness across various clinical outcomes, both in controlled trials and real-world settings. Pivotal clinical trials have shown this compound's superior efficacy in reducing the annualized relapse rate (ARR) and improving brain magnetic resonance imaging (MRI) outcomes compared to placebo or intramuscular interferon nsj.org.sa.

    Real-world studies consistently confirm these benefits. In a study of 60 patients with relapsing-remitting multiple sclerosis (RRMS) in Western Iran, a significant 85% reduction in the annualized relapse rate was observed, decreasing from 1.8±1.35 at baseline to 0.27±0.58 after 12 months of treatment (p=0.001) nsj.org.sa. Furthermore, 76.66% of these patients were relapse-free after the 12-month intervention nsj.org.sa. Another multivariate pooled analysis of two observational studies in Spain, involving 988 RRMS patients, reported a decrease in mean ARR by 76.5% after 1 year, 82.4% after 2 years, and 86.3% after 3 years, compared to the year prior to this compound initiation (all p<0.0001) neurology.org. This analysis also showed that at 12 months, 89.6% of patients had stable or improved Expanded Disability Status Scale (EDSS) scores, which was maintained in 84.4% of patients at 24 months neurology.org.

    Table 2: Annualized Relapse Rate (ARR) Reduction with this compound

    Study/CohortBaseline ARR12-Month ARR24-Month ARR36-Month ARRARR Reduction (%)Relapse-Free Patients (12 Months)
    Western Iran RRMS (N=60) nsj.org.sa1.8 ± 1.350.27 ± 0.58--85%76.66%
    Spanish RRMS (N=988) neurology.org1.190.280.210.1676.5% (1yr), 82.4% (2yr), 86.3% (3yr)-

    A systematic review of real-world data for this compound highlighted that treatment improved outcomes compared to the period before treatment initiation and was more effective than interferons or glatiramer acetate researchgate.netnih.gov. Long-term extension studies of the FREEDOMS trial also demonstrated sustained effectiveness, with the ARR remaining low and a high proportion of patients remaining free of relapse and confirmed disease progression over several years dovepress.com. MRI outcomes were also favorable, with a high percentage of patients free of gadolinium-enhancing lesions (GELs) and new or enlarging T2 lesions dovepress.com.

    Neurofilament Light Chain as a Biomarker for Neurodegeneration

    Neurofilament light chain (NfL) is a neuronal structural protein that has emerged as a promising biomarker for axonal injury and neurodegeneration in multiple sclerosis frontiersin.orgnih.govmdpi.com. Elevated levels of NfL in cerebrospinal fluid (CSF) and blood are associated with MS activity, relapses, disability progression, and MRI lesions, reflecting ongoing neuronal damage mdpi.com.

    Studies have investigated the impact of this compound on NfL levels. In a subset of patients from the phase 3 FREEDOMS study, CSF NfL levels were assessed at baseline and after 12 months of treatment nih.govneurology.org. Median NfL levels did not significantly differ between treatment groups at baseline. However, following 12 months of treatment, median changes from baseline in NfL levels were significantly reduced in the this compound groups compared to placebo nih.govneurology.org. For the pooled 0.5/1.25 mg this compound group, the median change was -326 pg/mL (83.3% with reduction, p=0.002), whereas the placebo group showed a reduction of -214 pg/mL (66.7% with reduction, p=0.388) nih.govneurology.org. Importantly, these reductions in NfL levels at month 12 correlated with an improvement in relapse and MRI outcomes, suggesting a beneficial effect of this compound on axonal integrity and supporting NfL's utility as a quantitative biomarker in MS nih.govneurology.org.

    Table 3: Median Neurofilament Light Chain (NfL) Levels and Changes in FREEDOMS Study (12 Months)

    Treatment GroupBaseline Median NfL (pg/mL)12-Month Median NfL (pg/mL)Median Change from Baseline (pg/mL)% with Reductionp-value (Change vs. Zero)
    This compound 0.5 mg644--346-0.039
    This compound 1.25 mg659--313-0.035
    Pooled this compound652335-32683.3%0.002
    Placebo886738-21466.7%0.388

    While NfL is a non-specific marker of neurodegeneration, its consistent elevation in MS activity and its reduction with effective therapies like this compound underscore its potential as a prognostic biomarker for identifying early biochemical signs of disease progression and treatment response mdpi.compracticalneurology.com.

    Q & A

    Q. What experimental models are commonly used to investigate Fingolimod’s mechanism of action in multiple sclerosis (MS)?

    this compound’s mechanism is studied using in vitro lymphocyte migration assays and in vivo models like experimental autoimmune encephalomyelitis (EAE). These models assess immune modulation via sphingosine-1-phosphate receptor (S1PR) internalization, which traps lymphocytes in lymph nodes . CNS effects are evaluated using brain tissue analysis to measure direct S1PR modulation on astrocytes and oligodendrocytes .

    Q. How are clinical trial endpoints (e.g., relapse rate, MRI activity) standardized in this compound studies?

    Phase III trials (e.g., FREEDOMS, TRANSFORMS) use annualized relapse rate (ARR), gadolinium-enhancing MRI lesions, and disability progression (EDSS) as primary endpoints. Standardized protocols include double-blinded designs, placebo or active comparators (e.g., interferon-β), and 24-month follow-ups to assess sustained efficacy .

    Q. What methodologies are employed to validate this compound’s pharmacokinetics and hepatic metabolism?

    Pharmacokinetic studies use liquid chromatography-mass spectrometry (LC-MS) to measure plasma concentrations, with a focus on CYP4F2-mediated metabolism. Severe hepatic impairment cohorts are monitored for altered exposure using terminal half-life calculations (6–9 days) and dose adjustments .

    Advanced Research Questions

    Q. How can researchers resolve contradictory findings on this compound’s infection risk in meta-analyses?

    Systematic reviews (e.g., Zhao et al., 2021) address heterogeneity by stratifying trials based on patient demographics, prior DMT use, and infection definitions. Sensitivity analyses exclude studies with high bias risk, while subgroup analyses differentiate between herpesviruses (e.g., VZV) and bacterial infections. Pooled odds ratios are adjusted for confounders like lymphocyte count thresholds .

    Q. What advanced bioinformatics tools identify this compound’s oncological targets, such as PLK1 in head and neck squamous cell carcinoma (HNSC)?

    TCGA data mining combined with Bayesian differential expression analysis (limma R package) identifies PLK1 as a key target. Validation includes in vitro lentiviral PLK1 overexpression in SCC9 cells, CCK-8 proliferation assays, and survival analysis via Kaplan-Meier curves. GSEA confirms enrichment in cell cycle pathways (NES=1.53, P<0.05) .

    Q. What methodologies mitigate bias in real-world this compound studies comparing clinical trial vs. practice populations?

    Real-world studies (e.g., PANGAEA) use propensity score matching to balance cohorts for disease severity and prior DMT exposure. Sensitivity analyses restrict populations to EU label criteria (highly active MS). Missing data are handled via multiple imputation or observed-case frameworks .

    Q. How is this compound’s neuroprotective potential evaluated in Alzheimer’s disease models?

    The 3xTg-AD mouse model undergoes Novel Object Location (NOL) tests to assess spatial memory. This compound’s efficacy is quantified by discrimination index improvements (e.g., from 0.2 to 0.5 at 12 months) and Western blot validation of amyloid-β reduction. CNS penetration is confirmed via LC-MS of brain tissue .

    Q. What strategies address rebound syndrome after this compound discontinuation in MS patients?

    Rebound risk is managed by overlapping this compound cessation with B-cell depleting therapies (e.g., rituximab) or corticosteroids. MRI monitoring at 4-week intervals tracks gadolinium lesion surges (median >30 new lesions). Washout periods are minimized (<2 weeks) to reduce relapse severity .

    Methodological Considerations

    Q. How are Bayesian statistical frameworks applied in TCGA-based studies of this compound’s anticancer effects?

    Bayesian analysis with the limma package identifies differentially expressed genes (|log2FC|≥1, P<0.05). Heatmaps and volcano plots visualize tumor vs. paracancerous tissue differences. Survival significance is validated via log-rank tests (HR=1.34 for PLK1) .

    Q. What experimental designs validate this compound’s biomarker-driven efficacy, such as serum Sema4A levels?

    Retrospective cohorts stratify MS patients by baseline Sema4A (ELISA-measured). Efficacy endpoints (ARR, EDSS) are compared between high/low Sema4A subgroups. Preclinical validation uses EAE mice injected with recombinant Sema4A-Fc, with this compound response assessed via clinical score reductions .

    Retrosynthesis Analysis

    AI-Powered Synthesis Planning: Our tool employs the Template_relevance Pistachio, Template_relevance Bkms_metabolic, Template_relevance Pistachio_ringbreaker, Template_relevance Reaxys, Template_relevance Reaxys_biocatalysis model, leveraging a vast database of chemical reactions to predict feasible synthetic routes.

    One-Step Synthesis Focus: Specifically designed for one-step synthesis, it provides concise and direct routes for your target compounds, streamlining the synthesis process.

    Accurate Predictions: Utilizing the extensive PISTACHIO, BKMS_METABOLIC, PISTACHIO_RINGBREAKER, REAXYS, REAXYS_BIOCATALYSIS database, our tool offers high-accuracy predictions, reflecting the latest in chemical research and data.

    Strategy Settings

    Precursor scoring Relevance Heuristic
    Min. plausibility 0.01
    Model Template_relevance
    Template Set Pistachio/Bkms_metabolic/Pistachio_ringbreaker/Reaxys/Reaxys_biocatalysis
    Top-N result to add to graph 6

    Feasible Synthetic Routes

    Reactant of Route 1
    Fingolimod
    Reactant of Route 2
    Fingolimod

    Disclaimer and Information on In-Vitro Research Products

    Please be aware that all articles and product information presented on BenchChem are intended solely for informational purposes. The products available for purchase on BenchChem are specifically designed for in-vitro studies, which are conducted outside of living organisms. In-vitro studies, derived from the Latin term "in glass," involve experiments performed in controlled laboratory settings using cells or tissues. It is important to note that these products are not categorized as medicines or drugs, and they have not received approval from the FDA for the prevention, treatment, or cure of any medical condition, ailment, or disease. We must emphasize that any form of bodily introduction of these products into humans or animals is strictly prohibited by law. It is essential to adhere to these guidelines to ensure compliance with legal and ethical standards in research and experimentation.