Skip to main content
Erschienen in: Drugs in R&D 4/2023

Open Access 28.08.2023 | Review Article

The Benefits and Risks of Switching from Fingolimod to Siponimod for the Treatment of Relapsing–Remitting and Secondary Progressive Multiple Sclerosis

verfasst von: Martin Vališ, Anat Achiron, Hans Peter Hartung, Jan Mareš, Veronika Tichá, Pavel Štourač, Simona Halusková, Francesco Angelucci, Zbyšek Pavelek

Erschienen in: Drugs in R&D | Ausgabe 4/2023

Abstract

Multiple sclerosis (MS) is a chronic neurodegenerative disease that affects the central nervous system (CNS). Currently, MS treatment is limited to several Food and Drug Administration (FDA)- and European Medicines Agency (EMA)-approved medications that slow disease progression by immunomodulatory action. Fingolimod and siponimod have similar mechanisms of action, and consequently, their therapeutic effects may be comparable. However, while fingolimod is mainly used for relapsing–remitting MS (RRMS), siponimod, according to EMA label, is recommended for active secondary progressive MS (SPMS). Clinicians and scientists are analysing whether patients can switch from fingolimod to siponimod and identifying the advantages or disadvantages of such a switch from a therapeutic point of view. In this review, we aim to discuss the therapeutic effects of these two drugs and the advantages/disadvantages of switching treatment from fingolimod to siponimod in patients with the most common forms of MS, RRMS and SPMS.
Key Points
Siponimod and fingolimod have different mechanisms of action in SPMS patients.
Differences in receptor profiles can explain therapeutic and side effects of these two drugs.
Switching from fingolimod to siponimod for the treatment of patients with active SPMS may have benefits and risks.

1 Introduction

Multiple sclerosis (MS) is a chronic neurodegenerative disease that affects the central nervous system (CNS) [16]. The main feature of this disease is the aggressive attack on the CNS by the immune system, damaging myelin [7, 8].
There are different forms of MS, each of them reflecting different disease phases and severity at the neurological level: clinically isolated syndrome (CIS), relapsing–remitting MS (RRMS), secondary progressive MS (SPMS) and primary progressive multiple sclerosis (PPMS) [9]. The most common form of MS is RRMS, in which acute episodes (‘attacks’ or ‘relapses’), characterized by the sudden worsening of neurological function, alternate with phases of remission typified by partial or complete recovery [9]. In many cases, RRMS progresses to SPMS [8], which is characterized by the constant worsening of neurological function and progressive disability in absence of noteworthy episodes of remission. Conversion rate to SPMS within 10 years of the first MS-related episode varies from 15% to 50%, with lower SPMS incidence linked to active management of MS with disease-modifying treatments [811].

2 MS Treatment

The goals of current MS treatments are threefold: pathologically, to reduce the ongoing autoimmune inflammatory process and to promote remyelination [1] and, clinically, to postpone the transition from RRMS to SPMS [12]. Currently, there is no treatment that achieves complete recovery from MS. Available treatments are limited to several Food and Drug Administration (FDA)- and European Medicines Agency (EMA)-approved medications that have been shown to reduce relapse rate, slow disease progression and diminish magnetic resonance imaging (MRI) activity [12]. The availability of different disease-modifying drugs (DMDs) for MS allows patients, in collaboration with their physicians, to try different treatment regimens based on individual efficacy and side effects to identify the best individual approach. Among recently developed drugs, two of them have similar mechanism of action: fingolimod, the first-in-class sphingosine 1-phosphate receptor modulator approved by the FDA and EMA for the treatment of RRMS in 2010 and 2011, respectively [13], and siponimod, approved by the FDA for RRMS, SPMS, or CIS in 2019 [14] and by the EMA [15] for active SPMS. It is commonly assumed that their therapeutic effects may be comparable. However, while fingolimod is mainly used for RRMS, siponimod in most countries is recommended for the treatment of SPMS. Clinicians and scientists are analysing whether patients can switch from fingolimod to siponimod and identifying the advantages or disadvantages of such a switch from a therapeutic point of view.
In this review, we aim to discuss the differences between therapeutic and side effects of these two DMDs and the advantages and disadvantages of switching treatment from fingolimod to siponimod in patients with the most common forms of MS, RRMS and SPMS.

2.1 Fingolimod in RRMS and SPMS

Fingolimod (Gilenya) was the first sphingosine 1-phosphate (S1P) receptor 1 (S1P1) modulator [13]. The primary activity of fingolimod associated with therapeutic outcomes is the functional inhibition S1P1 receptor on T cells. [16]. Fingolimod prevents autoaggressive T cells from exiting lymph nodes and entering the CNS to attack the myelin sheath. Thereby, fingolimod reduces inflammatory damage in the CNS [17]. The sequestration of autoaggressive T cells is reversible upon discontinuation of fingolimod treatment.
In Europe, the use of fingolimod is indicated in patients with highly active RRMS, defined as two or more disabling relapses in 1 year, and with one or more gadolinium-enhancing lesions on brain MRI, or with a significant increase in lesion load on T2-weighted imaging [18]. However, in the USA and Australia, fingolimod can also be administered to SPMS patients with superimposed relapses to reduce relapse activity [19, 20].
The efficacy of fingolimod in SPMS has been investigated in some clinical trials, but none of them were randomized, and the results are not conclusive [21, 22]. In the INFORMS study [23], when compared with placebo, fingolimod was not able to delay sustained disability progression in patients with primary progressive MS (PPMS). A recent study analysing different treatments for SPMS noted that fingolimod is superior to low-efficacy therapy in reducing disease progression in patients with active relapse associated with SPMS but not in those with inactive (no more relapses) SPMS [24].
In line with human data, fingolimod did not prevent secondary progressive neurodegeneration in the MS animal model of experimental allergic encephalomyelitis (EAE) [25]. These data indicate that the therapeutic action of fingolimod in SPMS patients is limited, especially in patients who are not in an active phase of the disease, suggesting that, once RRMS patients progress to the SPMS disease phase, it would be advised to switch the treatment to siponimod.

2.1.1 Side Effects

Potential side effects of fingolimod include bradycardia, infection, macular oedema, teratogenicity and progressive multifocal leukoencephalopathy [26]. In addition, suspension of fingolimod treatment in patients with RRMS has been associated with disease reactivation [27] or even disease exacerbation [28]. Therefore, patients should be closely monitored for MS activity for several months after stopping fingolimod. Moreover, another MS therapy, such as siponimod, should be started as soon as possible after discontinuing fingolimod, preferably after the lymphocyte count has normalized.

2.2 Siponimod in RRMS and SPMS

Siponimod (Mayzent) is a second-generation S1PR modulator that binds with high affinity to S1PR1 and S1PR5. It was approved by the FDA in 2019 for adults with RRMS, SPMS or CIS [14, 29]. In Europe, based on the pivotal phase 3 trial, the authorized therapeutic indication for siponimod is for adults with SPMS with active disease, evidenced by relapses or imaging features of inflammatory activity [30]. Siponimod is currently the only approved therapy for SPMS [31]. In RRMS, it was shown that siponimod reduced the frequency of relapses and the number of brain lesions at MRI [32].
Similar to fingolimod, siponimod acts as a functional antagonist of S1P1 on circulating T cells. Specifically, it induces receptor internalization and degradation (also called downmodulation) to inhibit cell egress from lymph nodes, and therefore, it prevents autoreactive lymphocyte infiltration into the CNS [33].
Siponimod may also exert therapeutic activity within the CNS [34]. In the EAE mouse model, siponimod prevented synaptic neurodegeneration, independently of its effects on circulating lymphocytes [35], and promoted remyelination in the Xenopus laevis model [3639].

2.2.1 Side Effects

The safety profile of siponimod in SPMS has been investigated in many studies and is comparable to that of other S1PR-modulating drugs, including the typical side effects of this class of agents.[33]. The most common adverse events associated with siponimod are headache, back pain, bradycardia, dizziness, fatigue, influenza, urinary tract infection, lymphopenia, nausea, alanine amino transferase elevation and upper respiratory tract infection. These side effects are dose-dependent, and only in rare cases can they lead to treatment interruption [40].

2.3 Switching from Fingolimod to Siponimod for the Treatment of RRMS

There are currently no data on the effects of switching from fingolimod to siponimod for the treatment of RRMS [41]. It was shown that siponimod has therapeutic effects in RRMS. A phase 2 clinical trial of patients with RRMS in 2013 showed that siponimod reduced MRI lesion activity at 3 months in a dose-dependent manner (from 0.25 to 10 mg) [32]. Although no serious side effects were reported at 3 months, some of them (headache, bradycardia, dizziness and nasopharyngitis) were observed after 6 months in a small percentage of patients (from 5% to 19% in the dose groups studied). Similar efficacy and safety results were reported in the extension of this phase 2 clinical trial in 2016 [42].
At present, there are not phase 3 studies on the effect of siponimod in RRMS. Thus, a comparison with phase 3 studies of fingolimod in RRMS is not feasible. The term ‘relapsing multiple sclerosis’ (RMS) includes relapsing–remitting multiple sclerosis (RRMS) and SPMS with relapses. The latter is termed ‘active’ SPMS, for which RMS- or active-SPMS-approved disease modifying therapies (DMTs) can be used. However, the level of evidence supporting efficacy and safety in SPMS differs between DMTs approved for RMS and SPMS [43]. In the aforementioned phase 2 clinical trials of patients with RRMS, it is concluded that evaluation of therapeutic effects and tolerability in RRMS warrant investigation in phase 3 trials [32, 42].
Although S1PR modulators have better efficacy and safety profiles than other neuroimmunomodulatory drugs for the treatment of RRMS [44], there is currently insufficient evidence indicating that switching from fingolimod to siponimod for the treatment of RRMS has potential benefit in terms of therapeutic efficacy and side effects. Thus, further preclinical and clinical studies are necessary to evaluate the advantages and disadvantages of this treatment switch in patients with RRMS.

2.4 Switching from Fingolimod to Siponimod for the Treatment of SPMS

Recently reported cost-effectiveness analyses of siponimod use in SPMS concluded that, in patients with active SPMS, switching to siponimod is a cost-effective and clinically beneficial treatment approach as compared with the continuation with other DMDs [45], including fingolimod [46]. Therefore, in patients with active SPMS, switching from fingolimod to siponimod may have both clinical and economic benefits, especially when this switching is supported by appropriate analyses to diagnose active SPMS.
Despite these indications on the potential benefit of switching from fingolimod to siponimod, this translation is likely not without risk. Some recent case reports have evidenced possible initial complications that need to be closely monitored. A SPMS patient who switched from fingolimod to siponimod experienced disease reactivation in the absence of medication during a gap between the two treatments [47]. Another patient developed acute optic neuritis after a few weeks of treatment with siponimod, which persisted after treatment discontinuation [48]. Furthermore, another SPMS patient developed severe lymphopenia 1 month after switching from fingolimod to siponimod [49].
In a recent case series with 13 SPMS patients switching from fingolimod to siponimod, it was reported that three patients experienced disease reactivation in the first 3 months after switching [50]. Based on pharmacokinetics of these two drugs, the authors suggest that disease reactivation after switching from fingolimod to siponimod may be due to upregulation of S1P3, which triggers a pro-inflammatory cytokine cascade via activation of NFκB [51].
These case reports highlight the potential risks of switching from fingolimod to siponimod for the treatment of SPMS. Careful monitoring of disease activity is therefore recommended before and after switching these treatment regimens.

2.5 Advantages and Disadvantages of Siponimod over Fingolimod, a Comparison of Their Mechanisms of Action, and the Therapeutic Implications of Switching Treatment

The current knowledge on the mechanisms of action of fingolimod and siponimod mostly comes from studies using animal and cell culture models [26, 33, 34] (Table 1). These studies have shown that, although these two drugs have a similar mechanism of action, they can activate different receptors in the sphingolipid-regulated signalling pathway. Sphingolipids constitute a highly complex class of molecules that serve as structural components of cellular membranes and as signalling molecules capable of eliciting apoptosis, differentiation, chemotaxis and other responses in mammalian cells [52].
Table 1
Preclinical in vitro, in vivo and human data on the mechanism of action of fingolimod and siponimod
 
Fingolimod
Siponimod
Action
Functional agonist of S1P5 and S1P3 receptors; functional antagonist of S1P1 receptor
Functional agonist of S1P5 receptor; functional antagonist of S1P1 receptor
Preclinical data
Prevention of autoaggressive T cells from exiting lymph nodes and entering the CNS to attack the myelin sheath [17]
Amelioration of experimental autoimmune encephalomyelitis in Lewis rats [16]
Neuroprotective action in cell culture and preclinical models [26]
Inhibition of T cell egress from lymph nodes, therefore preventing autoreactive lymphocyte infiltration into the CNS [33]
Neuroprotective action in cell culture and preclinical models [26]
Prevention of synaptic neurodegeneration in the EAE mouse model [35]
Promotion of remyelination and attenuation of demyelination in the Xenopus laevis model [3639]
Partial restore of cortical neuronal circuit function, lowering inflammatory cell infiltration in cortical grey matter lesions and restoring cortical network functionality in the EAE mouse model [57]
Human data
Significant reduction of active lesions detected on MRI in RRMS patients [18]
Reduction of relapses in SPMS patients with active relapse but not in those with inactive SPMS [24]
Reduction of relapses in adults with SPMS with active phase [30, 31]
Reduction of frequency of relapses and the number of brain lesions on MRI in RRMS [32]
S1P sphingosine 1-phosphate receptor, CNS central nervous system, MRI magnetic resonance imaging, RRMS relapsing–remitting multiple sclerosis, SPMS secondary progressive multiple sclerosis, EAE experimental allergic encephalomyelitis
One important sphingolipid is S1P, a physiological signalling molecule that acts as a ligand for a group of cell surface receptors, the S1P receptors (S1PRs). These receptors are expressed in various tissues and regulate diverse physiological and pathological cellular responses involved in innate and adaptive immunity as well as in cardiovascular and neurological functions [53]. In particular, S1PR1 is expressed on the cell surface of lymphocytes, which are well known for their major role in the pathogenesis of MS. Moreover, S1PR1 plays an important regulatory role in the egress of lymphocytes from lymphoid organs into lymphatic circulation [33].
Both fingolimod and siponimod target S1PR1 and thus modulate immune cell trafficking through the sequestration of autoreactive lymphocytes in lymphoid organs. This sequestration reduces their recirculation and subsequent infiltration into the CNS [33]. Therefore, these two drugs reduce MS immuno-inflammation.
Upon entry into the CNS, fingolimod and siponimod also reduce inflammatory signalling in glial cells and astrocytes expressing S1P1 [54]. In addition, both drugs can bind S1P5, which is predominantly expressed in oligodendrocytes and brain endothelial cells [55]. S1P5 activation protects adult oligodendrocytes from apoptosis and contributes to the maintenance of blood–brain barrier (BBB) integrity [56]. Moreover, S1P5 activation may support reparative action by promoting oligodendrocyte maturation and survival and myelination.
However, siponimod seems to have a more specific and selective activity than fingolimod in the CNS, at least in cellular cultures and animal models [34, 57]. Experiments in the Xenopus tadpole model showed that spontaneous remyelination does not occur in S1P5-knockout tadpoles and that remyelination increases in response to highly selective S1P5 agonists, but not to S1P1 agonists [39]. In EAE mice, siponimod administration induced partial restoration of cortical neuronal circuit function, lowering inflammatory cell infiltration in cortical grey matter lesions and restoring cortical network functionality [57]. Although a neuroprotective action of both fingolimod and siponimod has been observed in cell culture and preclinical models [26], clinical data for fingolimod have not conclusively shown a consistent effect on disease progression or other clinically relevant outcome measures of neurodegeneration [41]. The effect of siponimod on MS progression is more robust than that of fingolimod, resulting in greater efficacy for the treatment of SPMS. Siponimod appears to be more selective than fingolimod in targeting S1P5 [34]. Therefore, its action at S1P5 may be more potent in the CNS [34, 56]. In in vitro functional assays with astrocytes generated from human fibroblasts, it was shown that both siponimod and fingolimod have a comparable neuroprotective action on S1P1 receptor in glial cells [37]. Nonetheless, the specific target selectivity (S1P1,5) of siponimod and its CNS/blood pharmacokinetic (PK)/pharmacodynamic (PD) profiles allow optimal realization of a dual mode of action: inhibiting peripheral/central inflammation and promoting remyelination. This could also explain the different effects of siponimod and fingolimod in SPMS patients [33, 41].
When discussing the pharmacokinetic/pharmacodynamic (and side effects) of fingolimod and siponimod, it should be noted that fingolimod is a nonselective functional antagonist of four of the five S1PRs (1, 3, 4, and 5) [26]. In contrast, siponimod exhibits high-affinity binding to only S1PR1 and S1PR5, resulting in a favourable side effect profile compared with that of fingolimod.
At the S1P1-receptor level, both drugs act as functional antagonists and cause the internalization and degradation of this receptor [22]. The S1P3 receptor is instead only internalized by fingolimod, but not by siponimod [22]. These pharmacokinetic differences explain the more manageable cardiac effects of siponimod compared with fingolimod. In fact, the S1P3 receptor appears to be a key direct regulator of cardiac rhythm and is involved in the risk of bradycardia and type I and type II atrioventricular block, as demonstrated in mouse models [58].
The interaction of fingolimod with S1P3 may be responsible for serious side effects [59], such as atrioventricular nodal block [60] and macular oedema [61]. Nonetheless, similar side effects were also reported with siponimod [34, 62], bringing into question the effective role of the S1P3 receptor in such complications. It should be noted that the lack of action of siponimod on the S1P3 receptor, while bringing benefits in terms of cardiac complications, could also be the reason for disease reactivation after switching from fingolimod to siponimod, as previously stated. When fingolimod is withdrawn, there may be an upregulation of the S1P3 receptor which is present in astrocytes. This upregulation is able to trigger a pro-inflammatory response through the ability to activate RhoA [51].
In addition to these pharmacokinetic differences, there are also pharmacodynamic differences between fingolimod and siponimod [22, 63]. It was shown that fingolimod is able to penetrate the CNS to a greater extent than siponimod [64]. Siponimod, however, has a much faster elimination half-life (2–5 h) than fingolimod (12–16 h) [22, 63]. However, the bioavailability of the drug in the CNS may not correspond to the levels of free drug required to act on the receptors. Thus, therapeutic efficacy depends on a combination of factors including molecular composition, half-life, ability to cross nervous tissue membranes, and CNS transport and metabolism mechanisms (phosphorylation, oxidation) [65].
A representative scheme of the interactions of these two drugs with S1PRs is shown in Fig. 1.

3 Conclusions and Future Directions

In conclusion, switching from fingolimod to siponimod for the treatment of MS may have benefits, especially in patients with active SPMS. Siponimod also appears to be more potent than fingolimod in promoting myelin repair (or remyelination) in the CNS. This increased potency of siponimod may be due to its specific agonist action on the S1P5 receptor. Furthermore, the lack of activation of other S1PRs by siponimod, such as S1P3R, limits the incidence of side effects.
Given siponimod’s pharmacological profile at the receptor level, it is reasonable to conclude that the promyelination and neuroprotective effects are more beneficial when neuronal damage increases in more advanced MS forms, such as SPMS.
Further studies at preclinical and clinical levels are needed to determine the therapeutic efficacy caused by the modulation of different receptors in the S1P pathway of these two drugs in MS.

Declarations

Funding

This study was partially supported by the grant projects of the Ministry of Health of the Czech Republic (FN HK 00179906) and by Charles University in Prague, Czech Republic (PROGRES Q40/15).

Conflict of interest

The authors declare no conflicts of interest.

Ethics approval

Not applicable.
Not applicable.
Not applicable.

Availability of data and material

Not applicable.

Code availability

Not applicable.
Open AccessThis article is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License, which permits any non-commercial use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by-nc/​4.​0/​.
Literatur
1.
Zurück zum Zitat Ghasemi N, Razavi S, Nikzad E. Multiple sclerosis: pathogenesis, symptoms, diagnoses and cell-based therapy. Cell J. 2017;19:1–10.PubMed Ghasemi N, Razavi S, Nikzad E. Multiple sclerosis: pathogenesis, symptoms, diagnoses and cell-based therapy. Cell J. 2017;19:1–10.PubMed
2.
Zurück zum Zitat Lublin FD, Reingold SC, Cohen JA, Cutter GR, Sørensen PS, Thompson AJ, et al. Defining the clinical course of multiple sclerosis: the 2013 revisions. Neurology. 2014;83:278–86.PubMedPubMedCentralCrossRef Lublin FD, Reingold SC, Cohen JA, Cutter GR, Sørensen PS, Thompson AJ, et al. Defining the clinical course of multiple sclerosis: the 2013 revisions. Neurology. 2014;83:278–86.PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Lublin FD, Coetzee T, Cohen JA, Marrie RA, Thompson AJ, International Advisory Committee on Clinical Trials in MS. The 2013 clinical course descriptors for multiple sclerosis: a clarification. Neurology. 2020;94:1088–92.PubMedPubMedCentralCrossRef Lublin FD, Coetzee T, Cohen JA, Marrie RA, Thompson AJ, International Advisory Committee on Clinical Trials in MS. The 2013 clinical course descriptors for multiple sclerosis: a clarification. Neurology. 2020;94:1088–92.PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Lassmann H. Pathogenic mechanisms associated with different clinical courses of multiple sclerosis. Front Immunol. 2018;9:3116.PubMedCrossRef Lassmann H. Pathogenic mechanisms associated with different clinical courses of multiple sclerosis. Front Immunol. 2018;9:3116.PubMedCrossRef
6.
Zurück zum Zitat Iaffaldano P, Lucisano G, Guerra T, Patti F, Onofrj M, Brescia Morra V, et al. Towards a validated definition of the clinical transition to secondary progressive multiple sclerosis: a study from the Italian MS Register. Mult Scler [Internet]. 2022;13524585221114008. Available from: http://www.ncbi.nlm.nih.gov/pubmed/35971322. Iaffaldano P, Lucisano G, Guerra T, Patti F, Onofrj M, Brescia Morra V, et al. Towards a validated definition of the clinical transition to secondary progressive multiple sclerosis: a study from the Italian MS Register. Mult Scler [Internet]. 2022;13524585221114008. Available from: http://​www.​ncbi.​nlm.​nih.​gov/​pubmed/​35971322.
7.
Zurück zum Zitat Hemmer B, Kerschensteiner M, Korn T. Role of the innate and adaptive immune responses in the course of multiple sclerosis. Lancet Neurol. 2015;14:406–19.PubMedCrossRef Hemmer B, Kerschensteiner M, Korn T. Role of the innate and adaptive immune responses in the course of multiple sclerosis. Lancet Neurol. 2015;14:406–19.PubMedCrossRef
9.
Zurück zum Zitat Thompson AJ, Banwell BL, Barkhof F, Carroll WM, Coetzee T, Comi G, et al. Diagnosis of multiple sclerosis: 2017 revisions of the McDonald criteria. Lancet Neurol. 2018;17:162–73.PubMedCrossRef Thompson AJ, Banwell BL, Barkhof F, Carroll WM, Coetzee T, Comi G, et al. Diagnosis of multiple sclerosis: 2017 revisions of the McDonald criteria. Lancet Neurol. 2018;17:162–73.PubMedCrossRef
12.
Zurück zum Zitat Hart FM, Bainbridge J. Current and emerging treatment of multiple sclerosis. Am J Manag Care. 2016;22:s159–70.PubMed Hart FM, Bainbridge J. Current and emerging treatment of multiple sclerosis. Am J Manag Care. 2016;22:s159–70.PubMed
13.
Zurück zum Zitat Willis MA, Cohen JA. Fingolimod therapy for multiple sclerosis. Semin Neurol. 2013;33(1):37–44.PubMedCrossRef Willis MA, Cohen JA. Fingolimod therapy for multiple sclerosis. Semin Neurol. 2013;33(1):37–44.PubMedCrossRef
14.
Zurück zum Zitat Goodman AD, Anadani N, Gerwitz L. Siponimod in the treatment of multiple sclerosis. Expert Opin Investig Drugs. 2019;28:1051–7.PubMedCrossRef Goodman AD, Anadani N, Gerwitz L. Siponimod in the treatment of multiple sclerosis. Expert Opin Investig Drugs. 2019;28:1051–7.PubMedCrossRef
16.
Zurück zum Zitat Huwiler A, Zangemeister-Wittke U. The sphingosine 1-phosphate receptor modulator fingolimod as a therapeutic agent: recent findings and new perspectives. Pharmacol Ther. 2018;185:34–49.PubMedCrossRef Huwiler A, Zangemeister-Wittke U. The sphingosine 1-phosphate receptor modulator fingolimod as a therapeutic agent: recent findings and new perspectives. Pharmacol Ther. 2018;185:34–49.PubMedCrossRef
18.
Zurück zum Zitat Sanford M. Fingolimod: a review of its use in relapsing–remitting multiple sclerosis. Drugs. 2014;74:1411–33.PubMedCrossRef Sanford M. Fingolimod: a review of its use in relapsing–remitting multiple sclerosis. Drugs. 2014;74:1411–33.PubMedCrossRef
19.
Zurück zum Zitat Haas J, Jeffery D, Silva D, Meier DP, Meinert R, Cohen J, et al. Early initiation of fingolimod reduces the rate of severe relapses over the long term: post hoc analysis from the FREEDOMS, FREEDOMS II, and TRANSFORMS studies. Mult Scler Relat Disord. 2019;36: 101335.PubMedCrossRef Haas J, Jeffery D, Silva D, Meier DP, Meinert R, Cohen J, et al. Early initiation of fingolimod reduces the rate of severe relapses over the long term: post hoc analysis from the FREEDOMS, FREEDOMS II, and TRANSFORMS studies. Mult Scler Relat Disord. 2019;36: 101335.PubMedCrossRef
21.
Zurück zum Zitat Comi G, Hartung H-P, Bakshi R, Williams IM, Wiendl H. Benefit-risk profile of sphingosine-1-phosphate receptor modulators in relapsing and secondary progressive multiple sclerosis. Drugs. 2017;77:1755–68.PubMedPubMedCentralCrossRef Comi G, Hartung H-P, Bakshi R, Williams IM, Wiendl H. Benefit-risk profile of sphingosine-1-phosphate receptor modulators in relapsing and secondary progressive multiple sclerosis. Drugs. 2017;77:1755–68.PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Chun J, Giovannoni G, Hunter SF. Sphingosine 1-phosphate receptor modulator therapy for multiple sclerosis: differential downstream receptor signalling and clinical profile effects. Drugs. 2021;81:207–31.PubMedCrossRef Chun J, Giovannoni G, Hunter SF. Sphingosine 1-phosphate receptor modulator therapy for multiple sclerosis: differential downstream receptor signalling and clinical profile effects. Drugs. 2021;81:207–31.PubMedCrossRef
23.
Zurück zum Zitat Lublin F, Miller DH, Freedman MS, Cree BAC, Wolinsky JS, Weiner H, et al. Oral fingolimod in primary progressive multiple sclerosis (INFORMS): a phase 3, randomised, double-blind, placebo-controlled trial. Lancet (London, England). 2016;387:1075–84.PubMedCrossRef Lublin F, Miller DH, Freedman MS, Cree BAC, Wolinsky JS, Weiner H, et al. Oral fingolimod in primary progressive multiple sclerosis (INFORMS): a phase 3, randomised, double-blind, placebo-controlled trial. Lancet (London, England). 2016;387:1075–84.PubMedCrossRef
27.
Zurück zum Zitat Frau J, Sormani MP, Signori A, Realmuto S, Baroncini D, Annovazzi P, et al. Clinical activity after fingolimod cessation: disease reactivation or rebound? Eur J Neurol. 2018;25:1270–5.PubMedCrossRef Frau J, Sormani MP, Signori A, Realmuto S, Baroncini D, Annovazzi P, et al. Clinical activity after fingolimod cessation: disease reactivation or rebound? Eur J Neurol. 2018;25:1270–5.PubMedCrossRef
28.
Zurück zum Zitat Litwin T, Smoliński Ł, Członkowka A. Substantial disease exacerbation in a patient with relapsing–remitting multiple sclerosis after withdrawal from siponimod. Neurol Neurochir Pol. 2018;52:98–101.PubMedCrossRef Litwin T, Smoliński Ł, Członkowka A. Substantial disease exacerbation in a patient with relapsing–remitting multiple sclerosis after withdrawal from siponimod. Neurol Neurochir Pol. 2018;52:98–101.PubMedCrossRef
31.
Zurück zum Zitat Kappos L, Bar-Or A, Cree BAC, Fox RJ, Giovannoni G, Gold R, et al. Siponimod versus placebo in secondary progressive multiple sclerosis (EXPAND): a double-blind, randomised, phase 3 study. Lancet. 2018;391:1263–73.PubMedCrossRef Kappos L, Bar-Or A, Cree BAC, Fox RJ, Giovannoni G, Gold R, et al. Siponimod versus placebo in secondary progressive multiple sclerosis (EXPAND): a double-blind, randomised, phase 3 study. Lancet. 2018;391:1263–73.PubMedCrossRef
32.
Zurück zum Zitat Selmaj K, Li DKB, Hartung H-P, Hemmer B, Kappos L, Freedman MS, et al. Siponimod for patients with relapsing–remitting multiple sclerosis (BOLD): an adaptive, dose-ranging, randomised, phase 2 study. Lancet Neurol. 2013;12:756–67.PubMedCrossRef Selmaj K, Li DKB, Hartung H-P, Hemmer B, Kappos L, Freedman MS, et al. Siponimod for patients with relapsing–remitting multiple sclerosis (BOLD): an adaptive, dose-ranging, randomised, phase 2 study. Lancet Neurol. 2013;12:756–67.PubMedCrossRef
33.
Zurück zum Zitat Roy R, Alotaibi AA, Freedman MS. Sphingosine 1-phosphate receptor modulators for multiple sclerosis. CNS Drugs. 2021;35:385–402.PubMedCrossRef Roy R, Alotaibi AA, Freedman MS. Sphingosine 1-phosphate receptor modulators for multiple sclerosis. CNS Drugs. 2021;35:385–402.PubMedCrossRef
36.
38.
Zurück zum Zitat O’Sullivan C, Schubart A, Mir AK, Dev KK. The dual S1PR1/S1PR5 drug BAF312 (siponimod) attenuates demyelination in organotypic slice cultures. J Neuroinflammation. 2016;13:31.PubMedPubMedCentralCrossRef O’Sullivan C, Schubart A, Mir AK, Dev KK. The dual S1PR1/S1PR5 drug BAF312 (siponimod) attenuates demyelination in organotypic slice cultures. J Neuroinflammation. 2016;13:31.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Cao L, Li M, Yao L, Yan P, Wang X, Yang Z, et al. Siponimod for multiple sclerosis. Cochrane database Syst Rev. 2021;11:CD013647.PubMed Cao L, Li M, Yao L, Yan P, Wang X, Yang Z, et al. Siponimod for multiple sclerosis. Cochrane database Syst Rev. 2021;11:CD013647.PubMed
41.
Zurück zum Zitat Samjoo IA, Worthington E, Haltner A, Spin P, Drudge C, Cameron C, et al. Indirect comparisons of siponimod with fingolimod and ofatumumab in multiple sclerosis: assessing the feasibility of propensity score matching analyses. Curr Med Res Opin. 2021;37:1933–44.PubMedCrossRef Samjoo IA, Worthington E, Haltner A, Spin P, Drudge C, Cameron C, et al. Indirect comparisons of siponimod with fingolimod and ofatumumab in multiple sclerosis: assessing the feasibility of propensity score matching analyses. Curr Med Res Opin. 2021;37:1933–44.PubMedCrossRef
42.
Zurück zum Zitat Kappos L, Li DKB, Stüve O, Hartung H-P, Freedman MS, Hemmer B, et al. Safety and efficacy of siponimod (BAF312) in patients with relapsing–remitting multiple sclerosis: dose-blinded, randomized extension of the phase 2 BOLD study. JAMA Neurol. 2016;73:1089–98.PubMedCrossRef Kappos L, Li DKB, Stüve O, Hartung H-P, Freedman MS, Hemmer B, et al. Safety and efficacy of siponimod (BAF312) in patients with relapsing–remitting multiple sclerosis: dose-blinded, randomized extension of the phase 2 BOLD study. JAMA Neurol. 2016;73:1089–98.PubMedCrossRef
43.
Zurück zum Zitat Bayas A, Christ M, Faissner S, Klehmet J, Pul R, Skripuletz T, et al. Disease-modifying therapies for relapsing/active secondary progressive multiple sclerosis—a review of population-specific evidence from randomized clinical trials. Ther Adv Neurol Disord. 2023;16:17562864221146836.PubMedPubMedCentralCrossRef Bayas A, Christ M, Faissner S, Klehmet J, Pul R, Skripuletz T, et al. Disease-modifying therapies for relapsing/active secondary progressive multiple sclerosis—a review of population-specific evidence from randomized clinical trials. Ther Adv Neurol Disord. 2023;16:17562864221146836.PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Yang S, Li X, Wang J, Wang T, Xu Z, Gao H, et al. Sphingosine-1-phosphate receptor modulators versus interferon beta for the treatment of relapsing–remitting multiple sclerosis: findings from randomized controlled trials. Neurol Sci. 2022;43:3565–81.PubMedCrossRef Yang S, Li X, Wang J, Wang T, Xu Z, Gao H, et al. Sphingosine-1-phosphate receptor modulators versus interferon beta for the treatment of relapsing–remitting multiple sclerosis: findings from randomized controlled trials. Neurol Sci. 2022;43:3565–81.PubMedCrossRef
45.
Zurück zum Zitat Schur N, Gudala K, Vudumula U, Vadapalle S, Bhadhuri A, Casanova A, et al. Cost effectiveness and budget impact of siponimod compared to interferon beta-1a in the treatment of adult patients with secondary progressive multiple sclerosis with active disease in Switzerland. Pharmacoeconomics. 2021;39:563–77.PubMedPubMedCentralCrossRef Schur N, Gudala K, Vudumula U, Vadapalle S, Bhadhuri A, Casanova A, et al. Cost effectiveness and budget impact of siponimod compared to interferon beta-1a in the treatment of adult patients with secondary progressive multiple sclerosis with active disease in Switzerland. Pharmacoeconomics. 2021;39:563–77.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Montgomery S, Woodhouse F, Vudumula U, Gudala K, Duddy M, Kroes M. Stick or twist? Cost-effectiveness of siponimod compared with continuing existing disease-modifying therapies in the treatment of active secondary progressive multiple sclerosis in the UK. J Med Econ. 2022;25:669–78.PubMedCrossRef Montgomery S, Woodhouse F, Vudumula U, Gudala K, Duddy M, Kroes M. Stick or twist? Cost-effectiveness of siponimod compared with continuing existing disease-modifying therapies in the treatment of active secondary progressive multiple sclerosis in the UK. J Med Econ. 2022;25:669–78.PubMedCrossRef
47.
Zurück zum Zitat Senzaki K, Ochi H, Ochi M, Okada Y, Miura S, Ohyagi Y. Disease reactivation in a patient with secondary progressive multiple sclerosis after switching treatment from fingolimod to siponimod. eNeurologicalSci. 2021;23:100346.PubMedPubMedCentralCrossRef Senzaki K, Ochi H, Ochi M, Okada Y, Miura S, Ohyagi Y. Disease reactivation in a patient with secondary progressive multiple sclerosis after switching treatment from fingolimod to siponimod. eNeurologicalSci. 2021;23:100346.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Hussain R, O’Leary S, Pacheco FM, Zacharias TE, Litvak P, Sguigna P, et al. Acute relapse after initiation of siponimod in a patient with secondary progressive MS. J Neurol. 2016;263:606–10.PubMedCrossRef Hussain R, O’Leary S, Pacheco FM, Zacharias TE, Litvak P, Sguigna P, et al. Acute relapse after initiation of siponimod in a patient with secondary progressive MS. J Neurol. 2016;263:606–10.PubMedCrossRef
49.
Zurück zum Zitat Sparaco M, Miele G, Bonavita S. Severe lymphopenia switching from fingolimod to siponimod. Neurol Sci. 2021;42:4837–8.PubMedCrossRef Sparaco M, Miele G, Bonavita S. Severe lymphopenia switching from fingolimod to siponimod. Neurol Sci. 2021;42:4837–8.PubMedCrossRef
50.
51.
Zurück zum Zitat Van Doorn R, Van Horssen J, Verzijl D, Witte M, Ronken E, Van Het Hof B, et al. Sphingosine 1-phosphate receptor 1 and 3 are upregulated in multiple sclerosis lesions. Glia. 2010;58:1465–76.PubMedCrossRef Van Doorn R, Van Horssen J, Verzijl D, Witte M, Ronken E, Van Het Hof B, et al. Sphingosine 1-phosphate receptor 1 and 3 are upregulated in multiple sclerosis lesions. Glia. 2010;58:1465–76.PubMedCrossRef
52.
Zurück zum Zitat Hannun YA, Obeid LM. Sphingolipids and their metabolism in physiology and disease. Nat Rev Mol Cell Biol. 2018;19:175–91.PubMedCrossRef Hannun YA, Obeid LM. Sphingolipids and their metabolism in physiology and disease. Nat Rev Mol Cell Biol. 2018;19:175–91.PubMedCrossRef
53.
Zurück zum Zitat Therond P, Chapman MJ. Sphingosine-1-phosphate: metabolism, transport, atheroprotection and effect of statin treatment. Curr Opin Lipidol. 2022;33:199–207.PubMedCrossRef Therond P, Chapman MJ. Sphingosine-1-phosphate: metabolism, transport, atheroprotection and effect of statin treatment. Curr Opin Lipidol. 2022;33:199–207.PubMedCrossRef
54.
Zurück zum Zitat O’Sullivan S, Dev KK. Sphingosine-1-phosphate receptor therapies: advances in clinical trials for CNS-related diseases. Neuropharmacology. 2017;113:597–607.PubMedCrossRef O’Sullivan S, Dev KK. Sphingosine-1-phosphate receptor therapies: advances in clinical trials for CNS-related diseases. Neuropharmacology. 2017;113:597–607.PubMedCrossRef
56.
Zurück zum Zitat Spampinato SF, Merlo S, Sano Y, Kanda T, Sortino MA. Protective effect of the sphingosine-1 phosphate receptor agonist siponimod on disrupted blood brain barrier function. Biochem Pharmacol. 2021;186: 114465.PubMedCrossRef Spampinato SF, Merlo S, Sano Y, Kanda T, Sortino MA. Protective effect of the sphingosine-1 phosphate receptor agonist siponimod on disrupted blood brain barrier function. Biochem Pharmacol. 2021;186: 114465.PubMedCrossRef
57.
Zurück zum Zitat Hundehege P, Cerina M, Eichler S, Thomas C, Herrmann AM, Göbel K, et al. The next-generation sphingosine-1 receptor modulator BAF312 (siponimod) improves cortical network functionality in focal autoimmune encephalomyelitis. Neural Regen Res. 2019;14:1950–60.PubMedPubMedCentralCrossRef Hundehege P, Cerina M, Eichler S, Thomas C, Herrmann AM, Göbel K, et al. The next-generation sphingosine-1 receptor modulator BAF312 (siponimod) improves cortical network functionality in focal autoimmune encephalomyelitis. Neural Regen Res. 2019;14:1950–60.PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Sanna MG, Vincent KP, Repetto E, Nguyen N, Brown SJ, Abgaryan L, et al. Bitopic sphingosine 1-phosphate receptor 3 (S1P3) antagonist rescue from complete heart block: pharmacological and genetic evidence for direct S1P3 regulation of mouse cardiac conduction. Mol Pharmacol. 2016;89:176–86.PubMedPubMedCentralCrossRef Sanna MG, Vincent KP, Repetto E, Nguyen N, Brown SJ, Abgaryan L, et al. Bitopic sphingosine 1-phosphate receptor 3 (S1P3) antagonist rescue from complete heart block: pharmacological and genetic evidence for direct S1P3 regulation of mouse cardiac conduction. Mol Pharmacol. 2016;89:176–86.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Gajofatto A. Spotlight on siponimod and its potential in the treatment of secondary progressive multiple sclerosis: the evidence to date. Drug Des Dev Ther. 2017;11:3153–7.CrossRef Gajofatto A. Spotlight on siponimod and its potential in the treatment of secondary progressive multiple sclerosis: the evidence to date. Drug Des Dev Ther. 2017;11:3153–7.CrossRef
60.
Zurück zum Zitat Egom EE, Kruzliak P, Rotrekl V, Lei M. The effect of the sphingosine-1-phosphate analogue FTY720 on atrioventricular nodal tissue. J Cell Mol Med. 2015;19:1729–34.PubMedPubMedCentralCrossRef Egom EE, Kruzliak P, Rotrekl V, Lei M. The effect of the sphingosine-1-phosphate analogue FTY720 on atrioventricular nodal tissue. J Cell Mol Med. 2015;19:1729–34.PubMedPubMedCentralCrossRef
62.
Zurück zum Zitat Muñoz-Ortiz J, Reyes-Guanes J, Zapata-Bravo E, Mora-Muñoz L, Reyes-Hurtado JA, Tierradentro-García LO, et al. Ocular adverse events from pharmacological treatment in patients with multiple sclerosis—a systematic review of the literature. Syst Rev. 2021;10:280.PubMedPubMedCentralCrossRef Muñoz-Ortiz J, Reyes-Guanes J, Zapata-Bravo E, Mora-Muñoz L, Reyes-Hurtado JA, Tierradentro-García LO, et al. Ocular adverse events from pharmacological treatment in patients with multiple sclerosis—a systematic review of the literature. Syst Rev. 2021;10:280.PubMedPubMedCentralCrossRef
63.
Zurück zum Zitat Chaudhry BZ, Cohen JA, Conway DS. Sphingosine 1-phosphate receptor modulators for the treatment of multiple sclerosis. Neurotherapeutics. 2017;14:859–73.PubMedPubMedCentralCrossRef Chaudhry BZ, Cohen JA, Conway DS. Sphingosine 1-phosphate receptor modulators for the treatment of multiple sclerosis. Neurotherapeutics. 2017;14:859–73.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Bigaud M, Rudolph B, Briard E, Beerli C, Hofmann A, Hermes E, et al. Siponimod (BAF312) penetrates, distributes, and acts in the central nervous system: preclinical insights. Mult Scler J Exp Transl Clin. 2021;7:20552173211049170. Bigaud M, Rudolph B, Briard E, Beerli C, Hofmann A, Hermes E, et al. Siponimod (BAF312) penetrates, distributes, and acts in the central nervous system: preclinical insights. Mult Scler J Exp Transl Clin. 2021;7:20552173211049170.
65.
Zurück zum Zitat Hammarlund-Udenaes M, Fridén M, Syvänen S, Gupta A. On the rate and extent of drug delivery to the brain. Pharm Res. 2008;25:1737–50.PubMedCrossRef Hammarlund-Udenaes M, Fridén M, Syvänen S, Gupta A. On the rate and extent of drug delivery to the brain. Pharm Res. 2008;25:1737–50.PubMedCrossRef
Metadaten
Titel
The Benefits and Risks of Switching from Fingolimod to Siponimod for the Treatment of Relapsing–Remitting and Secondary Progressive Multiple Sclerosis
verfasst von
Martin Vališ
Anat Achiron
Hans Peter Hartung
Jan Mareš
Veronika Tichá
Pavel Štourač
Simona Halusková
Francesco Angelucci
Zbyšek Pavelek
Publikationsdatum
28.08.2023
Verlag
Springer International Publishing
Erschienen in
Drugs in R&D / Ausgabe 4/2023
Print ISSN: 1174-5886
Elektronische ISSN: 1179-6901
DOI
https://doi.org/10.1007/s40268-023-00434-6

Weitere Artikel der Ausgabe 4/2023

Drugs in R&D 4/2023 Zur Ausgabe

Acknowledgement to Referees

Acknowledgement to Referees