Small Molecule
C25H31N3O5
1262414-04-9
Cenerimod is an investigational, first-in-class, orally administered small molecule being developed for the treatment of Systemic Lupus Erythematosus (SLE).[1] It functions as a highly potent and selective modulator of the sphingosine-1-phosphate receptor subtype 1 (
S1P1), a validated therapeutic target for controlling lymphocyte trafficking in autoimmune diseases.[3] With a high functional potency (half-maximal effective concentration,
EC50, of approximately 1–2.7 nM), Cenerimod's primary mechanism involves inducing the internalization of S1P1 receptors on lymphocytes, which sequesters these pathogenic immune cells within secondary lymphoid organs and prevents their migration to sites of inflammation.[2]
The pharmacological profile of Cenerimod is distinguished by its unique signaling properties and exceptional selectivity, which contribute to a potentially superior safety profile compared to less selective agents in its class.[3] Its pharmacokinetic (PK) profile is characterized by a cytochrome P450 (CYP) enzyme-independent metabolism, minimizing the potential for drug-drug interactions, and an unusually long terminal half-life.[6] This long half-life results in a gradual accumulation to steady-state, creating a "built-in up-titration" effect that naturally mitigates the acute, first-dose cardiac effects commonly associated with
S1P receptor modulators.[6]
Clinical development has progressed to late-stage trials. The Phase 2b CARE study, while not meeting its primary endpoint after statistical adjustment for multiplicity, provided crucial insights. The 4 mg dose demonstrated clinically meaningful and statistically significant improvements in disease activity, particularly in a predefined subgroup of patients with a high Type 1 Interferon (IFN-1) gene expression signature—a marker of more severe disease.[10] These findings directly informed the design of the ongoing pivotal Phase 3 OPUS program, which is evaluating the 4 mg dose in a patient population enriched for this biomarker.[13]
Cenerimod has been generally well-tolerated in clinical trials, with an adverse event profile consistent with its mechanism of action, including a predictable and reversible reduction in lymphocyte count (lymphopenia).[1] Having received Fast Track designation from the U.S. Food and Drug Administration (FDA), Cenerimod is being co-developed through a global collaboration between Idorsia Pharmaceuticals and Viatris and is positioned as a potential blockbuster oral therapy for SLE.[1]
Systemic Lupus Erythematosus (SLE) is a complex, chronic, and heterogeneous autoimmune disease characterized by the loss of immunological tolerance, leading to the production of autoantibodies against nuclear antigens.[17] The pathophysiology involves the aberrant activation of both T and B lymphocytes, which drives the production of these autoantibodies by plasma cells.[10] The subsequent formation and deposition of immune complexes in various tissues trigger widespread inflammation, resulting in a diverse array of clinical manifestations and potentially irreversible organ damage, including lupus nephritis and central nervous system involvement.[10]
The current treatment landscape for SLE relies heavily on broad-spectrum immunosuppressants and corticosteroids, which, while effective at controlling flares, are associated with significant long-term toxicities.[2] More recent targeted biologic therapies have improved outcomes for some patients, but a substantial unmet medical need persists for novel, orally available treatments that offer a more targeted mechanism of action with an improved benefit-risk profile.[1]
The sphingosine-1-phosphate (S1P) signaling pathway has emerged as a key regulator of immune cell trafficking and a validated therapeutic target in autoimmunity.[3] S1P is a bioactive sphingolipid that signals through a family of five G protein-coupled receptors (
S1P1–S1P5).[3] The egress of lymphocytes—including pathogenic autoreactive T and B cells—from secondary lymphoid organs (such as lymph nodes and spleen) into the peripheral circulation is critically dependent on their response to an S1P concentration gradient.[5] Lymphocytes express high levels of the
S1P1 receptor and follow this gradient to exit the lymphoid tissues where S1P levels are low and enter the blood and lymph where S1P levels are high.[3]
Modulation of the S1P1 receptor disrupts this process. Agonists of the S1P1 receptor cause its sustained internalization and degradation, rendering the lymphocytes functionally insensitive to the S1P gradient.[3] This effectively traps them within the lymphoid organs, preventing their migration to target tissues where they would otherwise mediate inflammation and damage.[3] This therapeutic concept was first validated with the approval of fingolimod, a non-selective
S1P receptor modulator, for the treatment of multiple sclerosis, establishing a precedent for the development of more selective agents like Cenerimod for a range of autoimmune diseases.[3]
Cenerimod is a small molecule drug with a well-defined chemical identity, referenced across multiple chemical and pharmaceutical databases.
The chemical structure of Cenerimod is complex, incorporating several key functional groups that define its pharmacological activity and physicochemical properties.
The physicochemical properties of Cenerimod are critical determinants of its pharmacokinetic profile, including its slow absorption, high protein binding, and long half-life. Key properties are summarized in Table 1. While the molecule is predicted to violate certain empirical rules for oral drug-likeness, such as the Rule of Five and Ghose Filter, its clinical development confirms its viability as an oral agent, highlighting that these rules are guidelines rather than absolute barriers.[23]
Table 1: Key Physicochemical and Structural Properties of Cenerimod
Property | Value | Source/Method |
---|---|---|
Water Solubility | 0.0744 mg/mL | ALOGPS 23 |
logP | 3.98 / 5.05 | ALOGPS / Chemaxon 23 |
logS | -3.8 | ALOGPS 23 |
pKa (Strongest Acidic) | 13.62 | Chemaxon 23 |
pKa (Strongest Basic) | 0.62 | Chemaxon 23 |
Physiological Charge | 0 | Chemaxon 23 |
Hydrogen Acceptor Count | 7 | Chemaxon 23 |
Hydrogen Donor Count | 2 | Chemaxon 23 |
Polar Surface Area | 110.73 Ų | Chemaxon 23 |
Rotatable Bond Count | 9 | Chemaxon 23 |
Number of Rings | 4 | Chemaxon 23 |
Drug-Likeness Rules | Result | Source |
Rule of Five | No | Chemaxon 23 |
Ghose Filter | No | Chemaxon 23 |
Veber's Rule | No | Chemaxon 23 |
MDDR-like Rule | Yes | Chemaxon 23 |
Cenerimod is a potent, orally active agonist of the S1P1 receptor.[4] Functional assays have demonstrated its high potency, with reported
EC50 values ranging from 1 nM to 2.7 nM.[4] A defining characteristic of Cenerimod is its exceptional selectivity for the
S1P1 receptor subtype. This selectivity is a key element of its design, intended to maximize the desired immunomodulatory effects while minimizing off-target activities associated with other S1P receptor subtypes, which are linked to adverse effects.[4] The receptor binding profile, detailed in Table 2, illustrates this pronounced selectivity.
Table 2: Receptor Binding Profile and Selectivity of Cenerimod
Receptor Subtype | Cenerimod EC50 (nM) | S1P (Natural Ligand) EC50 (nM) | pFTY720 EC50 (nM) | Selectivity Fold (vs. S1P1) |
---|---|---|---|---|
S1P1 | 1 | 0.1 | 0.5 | 1x |
S1P2 | >10,000 | N/A | N/A | >10,000x |
S1P3 | 228 | 0.1 | 0.5 | 228x |
S1P4 | 2,137 | 60 | 64 | 2,137x |
S1P5 | 36 | 67 | 0.5 | 36x |
Data compiled from sources 4 and.28 |
The drug's high selectivity is particularly important for its safety profile. By being at least 2000-fold less active at the S1P3 receptor, Cenerimod is expected to have a lower propensity for causing the cardiovascular side effects, such as bradycardia and atrioventricular block, that are mediated by this subtype.[4] Similarly, its lack of activity at the
S1P2 receptor likely contributes to the preclinical findings showing an absence of vasoconstrictor effects.[3] This pharmacological precision represents a significant advancement over first-generation, non-selective
S1P modulators.
As a functional agonist, Cenerimod binds to the S1P1 receptor on lymphocytes and triggers its potent and sustained internalization.[5] This process removes the receptor from the cell surface, effectively rendering the lymphocyte unable to detect and respond to the natural S1P gradient that guides its exit from lymphoid organs.[3] The result is the sequestration of T and B lymphocytes within these tissues, leading to a profound and dose-dependent reduction of their numbers in the peripheral blood.[2] This sequestration of potentially autoreactive lymphocytes is the primary mechanism through which Cenerimod exerts its immunomodulatory effects.
The therapeutic impact of Cenerimod extends beyond simple lymphocyte sequestration. Preclinical data show that it also reduces the migration of dendritic cells, inhibits T cell proliferation, and decreases the secretion of pro-inflammatory cytokines.[17] Furthermore, by modulating auto-antigen transport to lymph nodes, Cenerimod may interrupt the cycle of autoimmune activation at a more fundamental stage, preventing the priming of new autoreactive T cells and halting the propagation of the autoimmune response.[10]
In clinical studies of SLE patients, these mechanisms translate into measurable changes in key disease biomarkers. Treatment with Cenerimod leads to a dose-dependent reduction in circulating antibody-secreting cells (ASCs), which are the primary producers of pathogenic autoantibodies.[18] It also significantly reduces plasma levels of IFN-α and downregulates the gene expression signatures associated with both Type 1 Interferon (IFN-1) and IFN-γ, two cytokine pathways central to SLE pathogenesis.[13] This effect on interferon pathways is particularly pronounced in patients who exhibit a high IFN-1 gene signature at baseline, providing a strong biological rationale for the enhanced clinical response observed in this subpopulation.[13]
The repeated description of Cenerimod possessing "unique signaling properties" or "biased signaling" points to a sophisticated mechanism beyond simple receptor subtype selectivity.[3] A standard agonist activates all signaling pathways coupled to its receptor. Biased agonism, however, allows a drug to selectively engage certain downstream pathways while ignoring others. The
S1P1 receptor is known to couple to multiple intracellular signaling cascades. The therapeutic goal is to activate the pathway leading to receptor internalization and lymphocyte sequestration, while avoiding others that might cause unwanted effects, such as those mediated by calcium signaling in endothelial cells. The observed "attenuated calcium response" and the absence of vasoconstrictor or bronchoconstrictor effects, despite potent S1P1 agonism, strongly suggest that Cenerimod is a biased agonist.[15] This property, combined with its high receptor subtype selectivity, represents a deliberate drug design strategy to pharmacologically separate efficacy from toxicity at the molecular level, underpinning its potentially differentiated safety profile.
Cenerimod is administered orally and is characterized by slow absorption, with the median time to reach maximum plasma concentration (tmax) occurring between 5.0 and 7.0 hours post-dose.[6] The presence of food does not have a clinically meaningful impact on the overall drug exposure (Area Under the Curve, AUC), although it may slightly delay the rate of absorption.[32] This allows for flexible dosing without regard to meals. As a highly lipophilic compound, Cenerimod exhibits extensive binding to plasma proteins, at over 99.9%.[7]
A pivotal and differentiating feature of Cenerimod is its metabolism, which occurs independently of the cytochrome P450 (CYP) enzyme system.[6] This is a significant advantage over many other
S1P modulators, as it greatly reduces the risk of drug-drug interactions with concomitant medications that are CYP substrates, inhibitors, or inducers—a common concern in SLE patients who often require polypharmacy.[9]
No major metabolites of Cenerimod are found circulating in the plasma.[6] The principal metabolite, designated M32, is formed non-enzymatically in the gastrointestinal tract through a reductive cleavage of the oxadiazole ring.[7] While M32 constitutes the majority of the excreted drug-related material, it is not a major circulating species and therefore does not contribute significantly to the systemic pharmacological effect or potential for interactions.[8]
The primary route of elimination for Cenerimod and its metabolites is through the feces. A human mass balance study using radiolabeled Cenerimod found that approximately 84% of the administered dose was recovered, with the vast majority (58–100%) excreted in feces and a minor fraction (4.6–12%) in urine.[8]
Cenerimod is distinguished by an exceptionally long terminal half-life (t1/2). After a single dose, the half-life ranges from 170 to 199 hours (approximately 7 to 8 days).[32] With repeated once-daily dosing, the apparent terminal half-life is even longer, ranging from 283 to 799 hours (approximately 12 to 33 days).[7]
This long half-life leads to substantial drug accumulation, with steady-state exposure being 5- to 9-fold higher than after a single dose.[32] Steady-state plasma concentrations are typically reached after 20 to 32 days of continuous once-daily administration.[32] The pharmacokinetic parameters across different dose levels have been shown to be dose-proportional, indicating predictable drug behavior.[32]
Table 3: Summary of Cenerimod Pharmacokinetic Parameters (Single and Multiple Doses)
Study/Dose | tmax (h) | Cmax (ng/mL) | AUC (h·ng/mL) | t1/2 (h) | Accumulation Ratio (Day 35/Day 1) |
---|---|---|---|---|---|
Single Dose | |||||
1 mg | 6.2 | 4.73 | AUC0–∞: 1160 | 199 | N/A |
3 mg | 5.0 | 15.1 | AUC0–∞: 3720 | 170 | N/A |
10 mg | 6.0 | 58.7 | AUC0–∞: 14400 | 186 | N/A |
Multiple Dose (o.d. for 35 days) | |||||
0.5 mg | 6.0 | Day 35: 16.5 | Day 35 AUC0–24: 369 | 283 | Cmax: 5.3; AUC0–24: 7.2 |
1 mg | 6.0 | Day 35: 31.0 | Day 35 AUC0–24: 688 | 436 | Cmax: 6.0; AUC0–24: 8.0 |
2 mg | 5.0 | Day 35: 57.0 | Day 35 AUC0–24: 1270 | 415 | Cmax: 6.0; AUC0–24: 8.2 |
4 mg | 4.3 | Day 35: 130 | Day 35 AUC0–24: 2860 | 539 | Cmax: 7.0; AUC0–24: 9.0 |
Data represent geometric means from sources 32 and.33 |
The distinct pharmacokinetic and pharmacological properties of Cenerimod synergize to create a highly differentiated clinical profile. The CYP-independent metabolism ensures predictable pharmacokinetics with a low risk of drug-drug interactions, a critical advantage for SLE patients on multiple medications.[6] The very long half-life results in a slow, gradual increase in plasma concentration over several weeks, a phenomenon described as a "built-in up-titration".[6] This slow onset naturally mitigates the acute, first-dose bradycardia that is a well-known class effect of
S1P modulators, potentially obviating the need for first-dose cardiac monitoring required for other drugs in this class.[6] When combined with its high selectivity for the
S1P1 receptor, which avoids the S1P3 subtype more directly implicated in cardiac adverse events, these features collectively suggest a "best-in-class" potential from a safety, tolerability, and clinical usability standpoint.[4]
The therapeutic potential of Cenerimod was robustly demonstrated in the MRL/lpr mouse model, which recapitulates many features of human SLE.[17] In this model, oral administration of Cenerimod led to significant and broad disease amelioration.[17] The treatment produced a marked reduction in circulating T and B lymphocytes, which correlated with decreased immune cell infiltration into key target organs, including the brain and kidneys.[17] This reduction in organ-specific inflammation was accompanied by improvements in clinical markers of disease, such as a significant decrease in proteinuria (an indicator of improved kidney function) and lower titers of pathogenic anti-dsDNA autoantibodies.[17] Ultimately, these systemic and organ-specific benefits translated into a significant increase in the survival of Cenerimod-treated mice compared to controls.[17]
The immunomodulatory efficacy of Cenerimod is not confined to lupus models. It has also been shown to effectively attenuate disease parameters in the experimental autoimmune encephalomyelitis (EAE) mouse model, a standard preclinical model for multiple sclerosis.[3] This finding supports the broader applicability of its mechanism for treating T cell-mediated autoimmune conditions. Additionally, Cenerimod demonstrated anti-fibrotic effects in a mouse model of sclerodermatous chronic graft versus host disease, suggesting a potential role in diseases with a fibrotic component.[28]
In vitro experiments have provided direct confirmation of Cenerimod's mechanism of action on human cells. Studies using primary T and B lymphocytes isolated from both healthy donors and patients with SLE confirmed that Cenerimod is potent and efficacious in inducing the internalization of the S1P1 receptor.[5] Furthermore, functional assays demonstrated that Cenerimod effectively blocks the migration of activated T cells along an S1P chemotactic gradient, providing a direct cellular correlate for its ability to sequester lymphocytes in vivo.[5]
The clinical development of Cenerimod began with Phase 1 studies involving over 150 healthy volunteers, which established the initial safety, pharmacokinetic, and pharmacodynamic profile of the drug. These studies confirmed the dose-dependent reduction in peripheral lymphocyte counts that is characteristic of its mechanism of action.[15]
Following this, a Phase 2a proof-of-concept study (NCT02472795) was conducted in 67 patients with SLE.[15] This 12-week study successfully translated the findings from healthy volunteers to the target patient population, demonstrating a dose-dependent, sustained, and reversible reduction in lymphocyte counts.[15] The drug was well-tolerated across all tested doses (0.5, 1, 2, and 4 mg).[39] Importantly, exploratory analyses of efficacy at the 4 mg dose showed early signals of clinical and biological improvement, including a reduction in the mSLEDAI-2K disease activity score and a decrease in anti-dsDNA antibody levels compared to placebo.[19]
The encouraging results from the Phase 2a study led to the initiation of the larger, more definitive Phase 2b CARE study (NCT03742037).
Table 4: Key Efficacy Results from the Phase 2b CARE Study (4 mg Dose vs. Placebo at Month 6)
Endpoint | Patient Population | Cenerimod 4 mg | Placebo | Difference vs. Placebo (95% CI) | p-value (nominal) |
---|---|---|---|---|---|
Change from Baseline in mSLEDAI-2K | Overall Population | -4.04 | -2.85 | -1.19 (-2.25, -0.12) | 0.029 |
Change from Baseline in mSLEDAI-2K | IFN-1 High Subgroup | -5.41 | -2.62 | -2.78 (-4.50, -1.08) | 0.0015 |
SRI-4 Response Rate (%) | Overall Population | N/A | N/A | N/A | N/A |
SRI-4 Response Rate (%) | IFN-1 High Subgroup | 70% | 46% | +24% | N/A |
Data compiled from sources.10 |
The results of the CARE study, particularly the strong signal in the 4 mg arm and the identification of the IFN-1 high subgroup as likely high-responders, provided a clear and scientifically-driven path forward. The top-line "failure" of the CARE study was a result of a statistical penalty for testing multiple doses, not a lack of a biological or clinical signal. The study successfully fulfilled its purpose as a dose-finding and biomarker discovery trial. This led to a strategic pivot in the development program, focusing on a targeted patient population with a single, optimized dose.
This data-driven strategy culminated in the initiation of the pivotal Phase 3 OPUS (Oral S1P1 Receptor ModUlation in SLE) program in December 2022.[10]
Table 5: Summary of Key Cenerimod Clinical Trials in SLE
Trial Acronym/ID | Phase | Status | N (Patients) | Primary Endpoint |
---|---|---|---|---|
Phase 2a (NCT02472795) | 2a | Completed | 67 | Change in total lymphocyte count |
CARE (NCT03742037) | 2b | Completed | 427 | Change from baseline in mSLEDAI-2K at Month 6 |
OPUS-1 (NCT05648500) | 3 | Recruiting | ~420 | SRI-4 response rate at Month 12 |
OPUS-2 (NCT05672576) | 3 | Recruiting | ~420 | SRI-4 response rate at Month 12 |
OPUS OLE (NCT06475742) | 3 (Extension) | Active, not recruiting | ~680 | Long-term safety and tolerability |
Data compiled from sources.10 |
Across the clinical development program, Cenerimod has been consistently described as generally well-tolerated.[10] The majority of treatment-emergent adverse events (TEAEs) reported in studies have been mild to moderate in severity.[10]
Data from the Phase 2 program provide a detailed view of the safety profile. In the Phase 2b CARE study, the overall rates of AEs were similar across all Cenerimod dose groups and the placebo group over six months of treatment. The most frequent TEAEs that occurred more often in any Cenerimod group than in the placebo group were abdominal pain, headache, and the expected finding of lymphopenia.[14] A critical safety finding was that there was
no increased rate of infections in the Cenerimod arms compared to placebo, a significant concern for immunomodulatory therapies.[47] While a higher percentage of hypertension AEs was reported in some Cenerimod groups, objective monthly blood pressure measurements did not show any mean increases.[46] In the CARE study, two deaths occurred in the 1 mg group; however, both were investigated and determined to be unrelated to the study treatment.[11]
Table 6: Summary of Treatment-Emergent Adverse Events (TEAEs) in the CARE Study (6-Month Data)
Adverse Event (%) | Placebo (n=86) | Cenerimod 0.5 mg (n=85) | Cenerimod 1 mg (n=85) | Cenerimod 2 mg (n=86) | Cenerimod 4 mg (n=84) |
---|---|---|---|---|---|
Any TEAE | 54.7% | 49.4% | 64.7% | 59.3% | 58.3% |
Lymphopenia | 0% | 1% | 6% | 10% | 14% |
Headache | >5%* | >5%* | >5%* | >5%* | >5%* |
Abdominal Pain | >5%* | >5%* | >5%* | >5%* | >5%* |
Infections | 18.6% | 23.5% | 11.8% | 19.8% | 20.2% |
Specific percentages for headache and abdominal pain were not consistently reported across all sources but were noted as the most frequent AEs with >5% incidence in any group and higher than placebo.14 Data compiled from sources.11 |
The combination of high selectivity for the S1P1 receptor and unique signaling properties appears to confer a differentiated safety profile. Preclinical studies specifically noted an absence of bronchoconstrictor or vasoconstrictor effects, which are known potential liabilities for less selective S1P modulators acting on S1P2 and S1P3 receptors.[3] This targeted pharmacological action, together with the favorable clinical data gathered to date, supports the potential for a highly differentiated and favorable benefit-risk profile for Cenerimod in the treatment of SLE.[1]
Cenerimod is an investigational drug and is not yet approved for marketing in any country. However, it is on an expedited development track with major global health authorities.
Cenerimod was discovered and initially developed by Actelion Pharmaceuticals, with the program continuing under Idorsia Pharmaceuticals following a corporate spin-out.[3] In a significant strategic move in February 2024, Idorsia entered into a global research and development collaboration with Viatris Inc..[16] Under this agreement, Viatris acquired the development program and holds worldwide commercialization rights for Cenerimod, with the exception of Japan, South Korea, and certain other countries in the Asia-Pacific region.[16]
Cenerimod is positioned as a potential "blockbuster" oral therapy with a highly differentiated profile for SLE.[1] Commercial projections from Viatris anticipate a potential launch in the United States in 2028, with a potential loss of exclusivity (LOE) in 2036.[1] The drug is expected to command a premium value proposition and be positioned as an attractive oral option for use prior to injectable biologics in the SLE treatment paradigm.[1]
Cenerimod represents a sophisticated, third-generation approach to therapeutic immunomodulation via the S1P1 receptor. Its development reflects a deliberate, multi-parameter optimization strategy aimed at engineering a superior clinical profile. The combination of high receptor selectivity, unique biased signaling properties, a predictable CYP-independent pharmacokinetic profile, and a long half-life that inherently mitigates first-dose cardiac effects collectively distinguishes it from earlier agents in its class. This profile is designed to maximize the desired therapeutic effect of lymphocyte sequestration while minimizing off-target and mechanism-based toxicities.
If approved, Cenerimod would be the first S1P1 receptor modulator for SLE, introducing a novel oral mechanism of action to the treatment armamentarium.[1] It would compete with established biologics such as belimumab (Benlysta®) and anifrolumab (Saphnelo®).[1] Its key competitive advantages would be its oral route of administration and its targeted mechanism, which intervenes at the fundamental level of lymphocyte trafficking. The biomarker-driven strategy, focusing on patients with a high IFN-1 signature, could further allow Cenerimod to be positioned as a precision medicine for a well-defined patient subgroup often associated with more severe and immunologically active disease.
The ultimate success of the Cenerimod program now rests on the outcomes of the pivotal Phase 3 OPUS trials. The critical questions that these trials will answer are:
In conclusion, Cenerimod is a promising and scientifically well-rationalized therapeutic candidate for SLE. Its development path exemplifies a modern, biomarker-informed strategy that successfully navigated the complexities of a heterogeneous disease in mid-stage trials. The results of the OPUS program are highly anticipated by the clinical and scientific communities and will be decisive in determining whether Cenerimod's unique pharmacological profile translates into a valuable new treatment option for patients living with lupus.
Published at: September 26, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.