Small Molecule
C26H32N6O
2171019-55-7
Afimetoran (BMS-986256) is an investigational, first-in-class, orally bioavailable small molecule designed as an equipotent dual antagonist of Toll-like receptor 7 (TLR7) and Toll-like receptor 8 (TLR8). These endosomal receptors are key components of the innate immune system, and their aberrant activation by self-nucleic acids is a central driver in the pathophysiology of systemic autoimmune diseases, most notably Systemic Lupus Erythematosus (SLE). By selectively blocking this upstream signaling node, Afimetoran aims to inhibit the downstream production of pathogenic Type I interferons and other pro-inflammatory cytokines, offering a highly targeted immunomodulatory approach.
The preclinical evidence supporting Afimetoran is exceptionally robust. In validated murine models of advanced lupus, the compound demonstrated not only the ability to control disease but also to reverse established kidney damage, leading to a dramatic improvement in survival. A cornerstone of its preclinical value proposition is a pronounced steroid-sparing effect; Afimetoran was shown to act synergistically with corticosteroids, reversing TLR7-mediated resistance to steroid-induced apoptosis. This finding directly addresses a critical unmet need in lupus management: the reduction of cumulative toxicity from long-term glucocorticoid use.
The clinical development program, sponsored by Bristol Myers Squibb, has been methodical and strategically de-risked. A comprehensive suite of Phase 1 studies in healthy volunteers established a favorable safety, tolerability, and pharmacokinetic profile, supporting once-daily oral dosing. Crucially, early and thorough drug-drug interaction studies have suggested a low risk of clinically significant interactions, a vital characteristic for a drug intended for a patient population often on multiple concurrent medications.
The first evidence of clinical activity emerged from a Phase 1b study in patients with Cutaneous Lupus Erythematosus (CLE). In this trial, Afimetoran was safe, well-tolerated, and demonstrated rapid and sustained pharmacodynamic effects, including a significant reduction of the pathogenic interferon gene signature. Exploratory efficacy endpoints were met, with patients showing clinically meaningful improvements in skin disease activity. These promising results provided a strong rationale for advancing into a larger, ongoing Phase 2, placebo-controlled trial in patients with active SLE (NCT04895696). The future of Afimetoran is contingent upon the results of this pivotal study, which will determine if its targeted mechanism can translate into a safe and effective oral therapy capable of redefining the standard of care for patients with lupus.
Toll-like receptors (TLRs) are a class of evolutionarily conserved proteins that serve as a cornerstone of the innate immune system, acting as sentinel receptors that recognize conserved pathogen-associated molecular patterns (PAMPs).[1] Among these, the endosomally-located TLR7 and TLR8 are specialized in the detection of single-stranded RNA (ssRNA), a common signature of viral pathogens.[2] In a healthy immune response, the engagement of TLR7 and TLR8 by viral ssRNA within immune cells—such as plasmacytoid dendritic cells (pDCs), B cells, and monocytes—triggers a protective inflammatory cascade.[2]
However, in the context of systemic autoimmune diseases like Systemic Lupus Erythematosus (SLE), this protective mechanism becomes a central driver of pathology. SLE is characterized by defects in the clearance of apoptotic cellular debris, leading to the accumulation of self-derived nucleic acids.[3] When these self-RNAs form immune complexes with autoantibodies, they are internalized by immune cells and aberrantly activate TLR7 and TLR8 in the endosomes.[1] This chronic, inappropriate activation initiates a self-perpetuating feedback loop of inflammation. TLR7 activation in pDCs drives the massive production of Type I interferons (IFNs), creating a pro-inflammatory "IFN signature" that is a molecular hallmark of lupus.[1] Concurrently, TLR8 activation in monocytes and neutrophils contributes to the secretion of other pro-inflammatory cytokines.[1] This sustained signaling promotes the maturation of autoreactive B cells, increases the production of autoantibodies, and contributes to the development of steroid resistance, culminating in widespread inflammation and tissue damage.[1]
The decision to develop a compound that antagonizes both TLR7 and TLR8, rather than targeting only one, reflects a strategic understanding of this complex pathophysiology. While TLR7 is a primary driver of the IFN signature from pDCs, TLR8 plays a distinct but complementary role in activating other myeloid cells.[1] A dual antagonist, therefore, offers the potential for a more comprehensive blockade of the innate immune pathways fueled by self-RNA, addressing multiple cellular drivers of the disease simultaneously and potentially yielding greater therapeutic benefit than a more selective inhibitor.
In response to the clear pathogenic role of TLR7 and TLR8, Afimetoran (also known as BMS-986256) was developed as a first-in-class, orally bioavailable, selective small molecule inhibitor designed to disrupt this pathological cycle.[2] It is characterized as an equipotent dual antagonist of human TLR7 and TLR8, positioning it as a highly targeted immunomodulator.[2] Unlike broad-spectrum immunosuppressants that dampen the immune system globally, Afimetoran's mechanism is precise, aiming to selectively silence the specific receptors responsible for sensing pathogenic self-RNA. This targeted approach seeks to interrupt the inflammatory cascade at its origin while preserving other essential immune functions. Currently, Afimetoran is in clinical development for the treatment of inflammatory and autoimmune diseases, with a primary focus on SLE and its cutaneous manifestation, Cutaneous Lupus Erythematosus (CLE).[6]
Afimetoran is a small molecule drug classified as an indole-based compound.[2] Its identity is established through a comprehensive set of chemical names, development codes, and registry numbers that ensure unambiguous identification across scientific literature, patent filings, and regulatory databases. The complete identifiers for Afimetoran are consolidated in Table 1.
The physicochemical properties of Afimetoran are critical determinants of its absorption, distribution, metabolism, and excretion (ADME) profile, and thus its suitability as an oral therapeutic agent. As a small molecule, it is designed for oral bioavailability.[2]
Its lipophilicity is indicated by a calculated logP value of 4.26 (Chemaxon) or 3.9 (XLogP3), suggesting good membrane permeability but also a potential for non-specific binding or partitioning into adipose tissue.[2] The molecule possesses both weakly acidic and basic functional groups, with a strongest acidic pKa of 15.77 and a strongest basic pKa of 7.59.[2] The basic pKa near physiological pH results in a predicted physiological charge of +1, which can influence its solubility and interactions with biological targets.[2]
For non-clinical research, Afimetoran is soluble in dimethyl sulfoxide (DMSO), often requiring sonication and pH adjustment to achieve high concentrations (e.g., 100 mg/mL).[10] For in vivo animal studies, specific formulations using co-solvents such as polyethylene glycol 300 (PEG300), Tween-80, and saline are required to achieve adequate solubility for administration.[6]
Table 1: Chemical and Physicochemical Properties of Afimetoran
| Property | Value | Source(s) |
|---|---|---|
| Identification | ||
| Generic Name | Afimetoran | 2 |
| Development Code | BMS-986256 | 2 |
| CAS Number | 2171019-55-7 | 2 |
| IUPAC Name | 2-[4-(2-{7,8-dimethyl-triazolo[1,5-a]pyridin-6-yl}-3-(propan-2-yl)-1H-indol-5-yl)piperidin-1-yl]acetamide | 2 |
| Molecular Attributes | ||
| Molecular Formula | $C_{26}H_{32}N_{6}O$ | 2 |
| Average Molecular Weight | 444.58 g/mol | 2 |
| Monoisotopic Mass | 444.263759674 Da | 2 |
| Physicochemical Properties | ||
| Modality | Small Molecule | 2 |
| logP | 4.26 | 2 |
| pKa (Strongest Acidic) | 15.77 | 2 |
| pKa (Strongest Basic) | 7.59 | 2 |
| Physiological Charge | 1 | 2 |
| Polar Surface Area | 92.31 Ų | 2 |
| Hydrogen Bond Donors | 2 | 2 |
| Hydrogen Bond Acceptors | 4 | 2 |
| Rotatable Bonds | 5 | 2 |
| Solubility Notes | Soluble in DMSO (up to 100 mg/mL with adjustments); requires co-solvents for aqueous solutions. | 6 |
The primary mechanism of action of Afimetoran is its function as a direct, selective, and equipotent antagonist of human Toll-like receptor 7 and Toll-like receptor 8.[2] It is designed to physically interact with these receptors, likely at or near the orthosteric ligand-binding site within the endosome, thereby preventing the binding of their natural agonists, such as GU-rich or uridine-containing ssRNA.[1] This competitive inhibition precludes the agonist-induced conformational changes necessary for receptor dimerization, which is the initial and obligatory step for signal transduction.[2] The designation of Afimetoran as an "equipotent" antagonist is a key feature, indicating that it inhibits both TLR7 and TLR8 with comparable high potency.[4] This ensures a balanced and comprehensive blockade of the pathological ssRNA-sensing pathway. It is noted, however, that the precise in vitro potency measured for each receptor can exhibit some variability depending on the specific cell line, agonist, and endpoint used in the experimental assay.[11]
By preventing the initial activation and dimerization of TLR7 and TLR8, Afimetoran effectively halts the recruitment of essential downstream signaling components.[2] Upon agonist binding, activated TLRs recruit the adaptor protein Myeloid Differentiation Primary Response 88 (MYD88) through interactions between their Toll-interleukin-1 receptor (TIR) domains. This initiates the assembly of a higher-order signaling platform known as the "Myddosome," which includes IRAK4, IRAK1, and TRAF6.[2] The formation of this complex is the critical juncture that connects receptor activation to downstream cellular responses.
Afimetoran's antagonism at the receptor level prevents Myddosome assembly, thereby blocking the activation of two major downstream signaling arms:
By acting at the apex of this cascade, Afimetoran provides a complete shutdown of signals emanating from TLR7 and TLR8.[6]
The ultimate pharmacodynamic effect of Afimetoran is the suppression of the pathological inflammatory mediators that drive autoimmune disease. The blockade of the NF-κB and IRF pathways translates directly into a marked reduction in the production and secretion of key cytokines and chemokines. Pharmacodynamic analyses from both preclinical models and clinical trials have confirmed that treatment with Afimetoran leads to a significant decrease in NF-κB-dependent cytokines, such as Interleukin-6 (IL-6) and Tumor Necrosis Factor-alpha (TNFα), as well as IRF7-dependent Type I interferons like IFN-α.[3] In patients with lupus, this manifests as a rapid and sustained reduction in the characteristic Type I interferon gene signature, a key biomarker of disease activity.[17]
This highly specific mechanism of action distinguishes Afimetoran from conventional, broader-acting immunosuppressants. Standard therapies for lupus often involve agents like corticosteroids or mycophenolate mofetil, which have widespread effects across the immune system. In contrast, Afimetoran's activity is precisely focused on the TLR7/8 pathway, which is pathologically overactive in lupus, while leaving other components of the immune system, such as TLR3 and TLR9, functionally intact.[11] This targeted approach aims to normalize the specific dysregulated pathway driving the disease, which may preserve necessary immune functions and potentially lead to a more favorable safety profile, particularly concerning the risk of opportunistic infections, compared to less selective agents.[5]
Before advancing to animal models, Afimetoran's activity was rigorously characterized in a variety of in vitro cellular assays. These studies confirmed its high potency and selectivity. In assays using human whole blood from both healthy volunteers and patients with SLE, Afimetoran effectively inhibited the induction of IL-6 mediated by either TLR7 or TLR8 agonists.[9] The potency was further quantified by its ability to inhibit the upregulation of cell surface activation markers, demonstrating single-digit nanomolar ($nM$) half-maximal inhibitory concentration ($IC_{50}$) values for the suppression of CD69 on B cells (a TLR7-mediated response) and CD319 on monocytes (a TLR8-mediated response).[9] A critical aspect of its profile is its selectivity; parallel assays confirmed that Afimetoran had no inhibitory effect on other endosomal Toll-like receptors, such as TLR3 and TLR9, underscoring its highly targeted mechanism of action.[11]
The therapeutic potential of Afimetoran was compellingly demonstrated in the NZB/W F1 hybrid mouse model, a well-established and highly relevant preclinical model that spontaneously develops an autoimmune disease closely resembling human SLE, including severe lupus nephritis. A significant strength of these studies is that treatment was initiated in mice with established, advanced disease (e.g., significant proteinuria), a much higher and more clinically relevant bar for efficacy than prophylactic treatment.[3]
Oral administration of Afimetoran (at doses such as 0.25 mg/kg once daily) produced robust and multifaceted therapeutic effects [3]:
One of the most significant challenges in the long-term management of SLE is the cumulative toxicity associated with chronic glucocorticoid (steroid) therapy. Furthermore, some patients develop resistance to the therapeutic effects of steroids.[3] The preclinical development of Afimetoran identified a strong mechanistic rationale and compelling evidence for a steroid-sparing potential, establishing this as a core component of the drug's value proposition.
The activation of the TLR7/8-NFκB pathway has been directly implicated in promoting resistance to the apoptotic effects of steroids.[3] Preclinical studies demonstrated that Afimetoran could directly counteract this phenomenon. Both in vitro and in vivo, Afimetoran reversed the TLR7-agonist-induced resistance to prednisolone-induced apoptosis in key immune cells, including pDCs and B cells.[4]
This mechanistic synergy translated into superior efficacy in animal models. When Afimetoran was co-administered with a low dose of prednisolone in the NZB/W lupus model, the combination was more effective than either monotherapy. This combination therapy achieved 100% survival and demonstrated enhanced suppression of splenomegaly and pathogenic ABCs.[3] This powerful preclinical evidence provides a strong foundation for a clinical development strategy focused not only on direct disease modification but also on enabling a significant reduction in steroid burden for patients—a highly desirable clinical outcome for both patients and physicians. This preclinical finding directly informed the design of later-stage clinical trials, which incorporate corticosteroid dose reduction as a key secondary endpoint.[22]
The clinical development of Afimetoran has progressed through a series of methodically planned studies, moving from initial safety and pharmacokinetic assessments in healthy volunteers to proof-of-concept efficacy trials in patient populations. The program, sponsored by Bristol Myers Squibb, has been characterized by a proactive approach to de-risking, particularly regarding the drug's interaction potential, before embarking on large-scale patient studies.
Table 2: Overview of Afimetoran Clinical Trials
| NCT Identifier | Phase | Study Title/Purpose | Population | Status |
|---|---|---|---|---|
| NCT03634995 | 1 | Safety, tolerability, PK, and immunologic effects of single and multiple ascending doses | Healthy Participants | Completed |
| NCT04269356 | 1 | Assess ADME of radio-labeled BMS-986256 | Healthy Male Participants | Completed |
| NCT03950960 | 1 | DDI: Effect of itraconazole (CYP3A4 inhibitor) on Afimetoran PK | Healthy Participants | Completed |
| NCT04470778 | 1 | DDI: Effect of famotidine (acid-reducing agent) on Afimetoran drug levels | Healthy Participants | Completed |
| NCT05901714 | 1 | DDI: Effect of phenytoin (CYP inducer) on Afimetoran PK & effect of Afimetoran on midazolam (CYP3A4 substrate) PK | Healthy Participants | Completed |
| NCT04493541 | 1b | Safety, tolerability, and exploratory efficacy in patients with active CLE | Patients with CLE | Completed |
| NCT04895696 | 2 | Efficacy and safety of Afimetoran in patients with active SLE | Patients with SLE | Active, Not Recruiting |
The initial phase of clinical testing focused on establishing the fundamental characteristics of Afimetoran in humans.
Recognizing that SLE patients are often treated with multiple medications (polypharmacy), a series of DDI studies were conducted early in development to characterize Afimetoran's potential for interactions. This proactive strategy is crucial for ensuring a new drug can be safely and effectively integrated into existing treatment regimens.
This study served as a critical bridge from healthy volunteer data to patient treatment, providing the first human proof-of-concept for Afimetoran's therapeutic potential. The design, focusing on a measurable and visible manifestation of lupus, allowed for a rapid and efficient assessment of safety and activity before committing to a larger SLE trial.
Building on the successful proof-of-concept in CLE, this larger, ongoing trial is designed to definitively evaluate the efficacy and safety of Afimetoran in the broader and more complex SLE patient population.
Across the entire completed clinical program, encompassing studies in both healthy volunteers and patients with cutaneous lupus, Afimetoran has consistently demonstrated a favorable safety and tolerability profile.
Initial Phase 1 SAD/MAD studies in healthy volunteers established an acceptable safety profile, allowing for progression into patient populations.[12] The subsequent Phase 1b trial (NCT04493541) in CLE patients, which provides the most relevant data to date, met its primary safety and tolerability endpoints.[8] In this placebo-controlled setting, Afimetoran was well-tolerated, and no serious adverse events (SAEs) or concerning safety signals related to laboratory parameters, vital signs, or electrocardiograms were identified.[8] Similarly, in the drug-drug interaction study (NCT03950960) where Afimetoran was co-administered with the potent CYP3A4 inhibitor itraconazole, the combination was reported as safe and well-tolerated, with no SAEs, deaths, or discontinuations due to adverse events.[28]
A detailed examination of the adverse events (AEs) reported in the patient and DDI studies provides a clearer picture of Afimetoran's tolerability.
In the Phase 1b CLE study, AEs were generally characterized as mild or moderate in intensity and typically resolved without intervention.[8] Notably, the overall proportion of patients experiencing any AE was numerically lower in the Afimetoran group (5 of 8 patients, 62.5%) compared to the placebo group (4 of 5 patients, 80.0%).[8] The only discontinuation in the treatment arm was due to a symptomatic COVID-19 infection, an event not considered related to the drug's mechanism of action.[33]
The most common AEs reported in the DDI study with itraconazole were headache (30.4%), upper abdominal pain (17.4%), nausea (13.3%), and diarrhea (13.3%).[28] These represent common, generally manageable side effects often seen in early-phase clinical trials. The data from the CLE study, summarized in Table 3, further details the AE profile in a patient population.
Table 3: Summary of Treatment-Emergent Adverse Events from Phase 1b CLE Study (NCT04493541)
| Event Category / System Organ Class | Afimetoran (n=8) n (%) | Placebo (n=5) n (%) |
|---|---|---|
| Any Adverse Event (AE) | 5 (62.5) | 4 (80.0) |
| Serious AEs | 0 (0) | 0 (0) |
| AEs Leading to Discontinuation | 1 (12.5)¹ | 0 (0) |
| Drug-Related AEs | 2 (25.0) | 2 (40.0) |
| Infections and Infestations | 3 (37.5) | 1 (20.0) |
| Nervous System Disorders | 1 (12.5) | 2 (40.0) |
| Gastrointestinal Disorders | 1 (12.5) | 1 (20.0) |
| General and Administration Site Disorders | 2 (25.0) | 0 (0) |
| Data derived from source.41 | ||
| ¹Discontinuation was due to a symptomatic COVID-19 infection. |
The safety profile observed thus far is remarkably clean. For a novel immunomodulatory agent, a primary concern is the potential for increased susceptibility to infections or other mechanism-related toxicities. The available data, particularly from the placebo-controlled patient study, do not suggest such a liability. The fact that the overall AE rate was lower than placebo, and that the reported events are common and mild, supports the hypothesis that a targeted mechanism of action may translate to superior tolerability over broad immunosuppression. If this favorable safety profile is maintained in the larger, longer-term Phase 2 study, it would represent a significant competitive advantage for Afimetoran.
Afimetoran is an investigational drug and has not received marketing authorization from any major global regulatory agency, including the United States Food and Drug Administration (FDA), the European Medicines Agency (EMA), or the Australian Therapeutic Goods Administration (TGA).[42] It remains in clinical development.
The highest global development phase for Afimetoran is Phase 2, for the indication of Systemic Lupus Erythematosus, based on the ongoing NCT04895696 trial.[7] While one database notes that development for Cutaneous Lupus Erythematosus is "Discontinued," this likely refers to the completion of the specific Phase 1b trial (NCT04493541) rather than an abandonment of the indication, especially given that the trial produced positive safety and efficacy signals that support further investigation.[33]
Afimetoran is protected by intellectual property rights. Patents covering its chemical structure have been granted, with U.S. Patent 10,071,079 being specifically cited in public databases.[7] Further patent information is available through the World Intellectual Property Organization (WIPO), indicating a global patent strategy to protect the asset.[7] This patent protection is essential for securing market exclusivity and justifying the substantial investment required for late-stage clinical development and commercialization.
Consistent with its status as an investigational compound, Afimetoran is not available for therapeutic use outside of sanctioned clinical trials. However, the molecule is commercially available for purchase from various specialized chemical and life science suppliers, including GlpBio, MedKoo, MedChemExpress, TargetMol, and InvivoGen.[6] These suppliers provide Afimetoran strictly for "research use only" (RUO). Their terms of sale explicitly prohibit use in humans or animals for therapeutic or diagnostic purposes and clarify that the product is not sold to patients, ensuring its use is confined to preclinical and laboratory research settings.[6]
The development program for Afimetoran to date has built a compelling case for its potential as a novel therapy for lupus. The accumulated evidence reveals significant strengths while also highlighting key questions that must be addressed in ongoing and future studies.
Strengths:
Unanswered Questions:
Afimetoran is positioned to be a potential first-in-class oral therapy with a highly targeted mechanism of action. If the ongoing Phase 2 trial is successful, it would possess several key competitive advantages. Its oral route of administration offers a significant convenience benefit over injectable biologics. Its targeted immunomodulatory mechanism, which has so far yielded a favorable safety profile, could offer a significant advantage over broader immunosuppressants associated with greater toxicity.
However, its most powerful differentiator may be its demonstrated steroid-sparing potential. A therapy that can not only control lupus disease activity but also enable a significant reduction or even elimination of chronic corticosteroid use would be a paradigm-shifting advance. Such an agent would likely be used as an add-on to standard-of-care therapies (e.g., antimalarials) to improve overall disease control while addressing one of the most significant sources of long-term morbidity for lupus patients.
The entire future trajectory of Afimetoran hinges on the successful outcome of the ongoing Phase 2 SLE trial (NCT04895696). Positive data from this study, demonstrating a statistically significant and clinically meaningful benefit on the SRI-4 primary endpoint, would provide the necessary validation to proceed to a pivotal Phase 3 program. The data on secondary endpoints, particularly the ability to facilitate corticosteroid tapering, will be crucial for defining its ultimate value proposition.
Given the strength of the Phase 1b results in CLE, a dedicated pivotal development program for this indication should be strongly considered, as it may represent a more rapid path to market for a distinct patient population with a high unmet need. The long-term extension phase of the current SLE study will be invaluable for gathering the long-term safety and durability data required for regulatory submission. Overall, the outlook for Afimetoran is highly promising, backed by a strong scientific foundation and a well-executed early-stage clinical program. Confirmation of its efficacy in the larger SLE population is the final, critical step needed to unlock its potential as a transformative new treatment for lupus.
Published at: October 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.