What RORG antagonists are in clinical trials currently?

11 March 2025
Introduction to RORG

Definition and Function
Retinoic acid receptor-related orphan receptor gamma (RORG) is a member of the nuclear receptor superfamily that functions as a transcription factor. RORG—including its immune cell-specific isoform RORγt—is essential in regulating gene expression related to immune cell differentiation and inflammatory responses. In particular, RORγt plays a key role in inducing the differentiation of T helper 17 (Th17) cells and in initiating the production of interleukin 17 (IL-17), a cytokine that contributes to protective immunity against pathogens as well as to the development of autoimmune and inflammatory diseases. This receptor works by binding to specific DNA response elements in the regulatory regions of target genes and recruiting coactivators necessary for transcription. Its activity is influenced by endogenous sterols and synthetic small molecules, each of which can drive conformational changes that determine whether the receptor is in an active state or, conversely, inhibited.

Role in Disease Mechanisms
Dysregulation of RORG has been implicated in several immune-mediated and inflammatory conditions. Overactivity of RORγt, in particular, leads to an increase in IL-17 production that has been implicated in the pathogenesis of psoriasis, rheumatoid arthritis, multiple sclerosis, and other autoimmune disorders. In psoriasis, aberrant RORγt signaling contributes to the development of chronic plaque lesions by promoting proinflammatory cytokine cascades and skin immune cell infiltration. Beyond autoimmunity, emerging evidence has revealed roles for RORG in metabolic regulation and even in cancer biology—for instance, influencing tumor cholesterol metabolism as well as modulating the activity of immune cells within the tumor microenvironment. The involvement of RORG in these diverse biological pathways makes it an attractive target for therapeutic modulation: both its agonism and antagonism may have utility depending on the disease context. For the purpose of controlling inflammatory responses, the inhibition of RORG activity has become a strategic focus in drug development.

RORG Antagonists

Mechanism of Action
RORγ antagonists are designed to suppress the transcriptional activity of RORG by interfering with its ligand-binding domain (LBD). Under normal circumstances, binding of endogenous agonists to the LBD facilitates the establishment of a stable “agonist lock” formed by critical residue interactions (such as the His479–Tyr502–Phe506 network) that promote coactivator recruitment and drive gene transcription. RORG antagonists work through several potential mechanisms: they may directly compete with endogenous agonists at the orthosteric site, thereby disrupting the agonist lock; alternatively, they may bind allosterically and induce conformational changes that either displace or prevent the proper orientation of helix 12 (H12), resulting in impaired recruitment of the transcriptional coactivators. This disruption steers the receptor toward an inactive conformation and ultimately leads to downregulation of the inflammatory gene program managed by Th17 cells. Importantly, many of these antagonists display tissue- and gene-context selectivity. In other words, while they effectively block RORG activity in pathogenic conditions, their activities may vary in different cell types or tissues, depending on the local chromatin context and the repertoire of coactivators or corepressors present. The ability to fine-tune this signal modulation is critical, especially for drugs aimed at controlling chronic autoimmune responses without causing broad immunosuppression.

Potential Therapeutic Applications
Given the pivotal role of RORG, and particularly RORγt, in the regulation of IL-17 production and Th17 cell differentiation, its antagonists are being actively pursued as therapeutic agents for autoimmune and inflammatory conditions. The most advanced clinical development efforts are focusing on skin-related inflammatory diseases such as plaque psoriasis. By blocking the overactive signaling of RORG, these agents aim to reduce the production of proinflammatory cytokines, thereby leading to clinical improvements in the inflammatory skin lesions and reducing scales, redness, and infiltration.
In addition to psoriasis, RORG antagonists are of interest for other Th17-driven autoimmune diseases such as rheumatoid arthritis and multiple sclerosis. Moreover, because recent pathological insights have linked aberrant cholesterol biosynthesis and proinflammatory signaling in advanced prostate cancer and other cancers to RORG activity, there is also an emerging rationale for exploring these compounds in oncology to mitigate tumor-promoting inflammatory pathways. However, to date, the most mature indications in the clinical trial arena remain those related to autoimmune skin conditions, where the visible clinical benefits can be more readily evaluated alongside biochemical markers such as reductions in IL-17 levels. Overall, the potential therapeutic applications of RORG antagonists are expansive, ranging from enhancing the safety profile of immunomodulatory regimens to potentially combining with other targeted therapies for synergistic effects.

Current Clinical Trials

Overview of Ongoing Trials
Several RORG antagonists have entered clinical development, particularly with psoriasis as the key indication. Among the leading agents currently under investigation are:

• PF-06763809: A topical steroid-like formulation being tested in subjects with mild to moderate chronic plaque psoriasis. According to a CTGOV-registered clinical trial, this compound is being evaluated in a randomized, double-blind, vehicle and active comparator-controlled study. The investigational agent is designed to assess safety, tolerability, and its impact on psoriatic skin parameters such as infiltrate thickness.

• AZD0284: This candidate has undergone studies in both healthy volunteers and patient populations. An initial Phase I single-blind, placebo-controlled study in healthy subjects has been conducted to evaluate safety, tolerability, pharmacokinetics, and pharmacodynamics. Subsequently, a Phase 1b study was initiated to assess the efficacy and safety of a single oral dose regimen in patients with moderate to severe plaque psoriasis over a four-week period. An additional Phase I study assessing its bioavailability and intravenous microtracer kinetics further supports its development.

• JTE-451: This compound has been the focus of multiple clinical investigations. Early-phase clinical trials (Phase I) have evaluated the safety, tolerability, pharmacokinetics, and pharmacodynamics of JTE-451 in subjects with active plaque psoriasis. In parallel, another trial has analyzed the potential of JTE-451 formulated as an ointment for single and multiple dosing, examining both safety profiles and pharmacodynamic outcomes in Phase I studies. Moreover, a multicenter, randomized, double-blind, placebo-controlled, parallel-group study—referred to as the IMPACT-PS trial—has been initiated as a Phase II evaluation focused on efficacy and safety in patients with moderate to severe plaque psoriasis.

Each of these clinical trials demonstrates the growing effort to bring RORG antagonists from early preclinical research into clinical practice. The shared theme among these agents is their targeting of the ligand-binding domain of RORG to antagonize aberrant activation in autoimmune pathologies while maintaining a tolerable safety profile, a concept well supported by preclinical mechanistic studies.

Phases and Objectives
The clinical development programs for these antagonists are structured in sequential phases to address key questions regarding safety, pharmacokinetics, pharmacodynamics, and ultimately the efficacy of the molecules:

• For PF-06763809, the trial is designed as a Phase I study where the primary objectives are to establish safety and tolerability upon topical administration. Secondary objectives include evaluating the impact on psoriatic plaque thickness using quantitative imaging and histological assessments. The design is randomized and double-blind, ensuring a robust control of bias. The early initiation of such a trial underscores the interest in addressing local inflammation driven by RORG activity in a well-defined patient population.

• AZD0284 has been evaluated in a study involving healthy volunteers in Phase I, where single and multiple ascending doses are administered to assess its pharmacokinetic (PK) profile and pharmacodynamic (PD) effects. In a subsequent Phase 1b trial, its clinical utility is being specifically examined in patients with moderate to severe plaque psoriasis. Here, the compound’s efficacy is being measured in terms of clinical endpoints such as improvements in skin lesion scores as well as the downregulation of inflammatory markers like IL-17. An additional trial focusing on the absolute bioavailability of oral versus intravenous administration further refines our understanding of the compound’s behavior in vivo.

• JTE-451’s clinical development has spanned multiple study phases. An initial Phase I trial evaluated the systemic safety and PK/PD profiles of the compound when administered for four weeks in individuals with active plaque psoriasis. A parallel investigation into a topical ointment formulation assessed whether localized treatment could provide similar anti-inflammatory benefits with potentially reduced systemic exposure. Most notably, the IMPACT-PS trial is a Phase II multicenter study that aims to robustly assess the efficacy and safety of JTE-451 over a 16-week treatment period in patients with moderate to severe plaque psoriasis. Objectives in this study include both clinical outcomes and biomarker endpoints to confirm the mechanistic action of RORG antagonism in mitigating pathological inflammation.

Each phase is designed not only to build upon safety and tolerability data obtained in healthy subjects but also to confirm the pharmacodynamic effects that would ideally lead to clinical improvements in psoriasis and potentially other autoimmune conditions. The diversity in trial design (topical versus oral formulations) also highlights the strategic variations in delivery that might cater to different patient needs or disease severities.

Outcomes and Future Directions

Preliminary Results and Efficacy
Although the full datasets from these clinical trials are still emerging, early indications suggest that RORG antagonists are showing promising profiles:

• PF-06763809, in its Phase I trial, has demonstrated acceptable local tolerability when applied topically, along with encouraging signals in reducing psoriatic skin infiltrate thickness in affected subjects. Given that plaque thickness and skin inflammation are reliable surrogate markers in psoriasis, these early improvements suggest a beneficial pharmacodynamic impact that is consistent with the expected mechanism of action of RORG antagonism.

• AZD0284 has progressed through initial Phase I studies in healthy volunteers with a favorable safety and pharmacokinetic profile. The mild-to-moderate adverse events reported have allowed escalation of doses to levels that are predicted to achieve sufficient receptor engagement without systemic toxicity. In its Phase 1b trial in psoriasis patients, preliminary efficacy endpoints such as changes in lesion severity and patient-reported outcome measures are being monitored closely. The initial results indicate that AZD0284 is well tolerated, and early observations hint at measurable improvements in psoriatic symptoms, reinforcing the potential of RORG antagonism to reduce pathogenic inflammation.

• JTE-451 has been studied in several trial formats. In the Phase I evaluation in active plaque psoriasis subjects, systemic administration of JTE-451 showed a manageable safety profile while eliciting significant pharmacodynamic changes—most notably, a reduction in inflammatory biomarkers, including those linked to Th17 cell activity. The IMPACT-PS trial, with its extended 16-week treatment duration, is expected to provide more definitive evidence of the compound’s efficacy in improving clinical endpoints such as lesion size, scaling, and overall skin clearance. Additional studies on a topical ointment formulation have offered insights into the feasibility of localized treatment approaches that could bypass some of the challenges of systemic immunomodulation. Collectively, these studies support the hypothesis that inhibiting RORG activity can directly modulate the inflammatory cascade that underpins psoriasis pathology.

From multiple perspectives, the early signals—ranging from histological improvements to the downregulation of inflammatory mediators (e.g., IL-17)—strongly validate the concept that RORG antagonists can effectively modulate key disease pathways. While it is still early to conclusively comment on long-term efficacy, the combination of favorable safety profiles, positive PK/PD data, and the promising early clinical outcomes provide a robust justification for advancing these compounds into later phase trials where larger patient populations and longer treatment durations will allow a more comprehensive assessment of therapeutic potential.

Challenges and Future Research
Despite these encouraging findings, a number of challenges remain as RORG antagonists transition from early-phase clinical trials to broader therapeutic applications. One of the foremost challenges is the establishment of the optimal dosing regimen. The dose–response relationship for these compounds appears to be complex, largely due to the tissue- and gene-context selectivity inherent to nuclear receptor modulation. Identifying the dose that achieves maximal target inhibition without compromising safety is critical, especially when considering that chronic administration may be necessary in autoimmune diseases such as psoriasis.

Another challenge lies in patient selection and stratification. Given that autoimmune diseases are heterogeneous and that the contribution of RORG-driven inflammation may vary among individuals, future trials may need to incorporate biomarker-based inclusion criteria. For example, baseline measurements of Th17 cell counts or IL-17 levels could help identify patients who are most likely to benefit from RORG antagonism. Such approaches would not only improve the efficacy outcomes in clinical trials but also reduce the risk of adverse effects in patients less likely to respond.

The long-term impact on the immune system is another critical aspect under investigation. As RORγ plays roles in both innate and adaptive immune responses, chronic inhibition might pose risks, including unintended immunosuppression or alterations in the normal circadian regulation of immune functions. Ongoing pharmacovigilance and extended follow-up periods will be necessary to fully document the safety profiles of these compounds over prolonged treatment durations.

Furthermore, the molecular diversity among RORG antagonists may translate into differences in clinical efficacy and tolerability. For instance, while PF-06763809 is administered topically, AZD0284 and JTE-451 have been formulated for both systemic and localized delivery. Each formulation comes with its own set of pharmacokinetic challenges, such as absorption, distribution, metabolism, and excretion (ADME) parameters that must be carefully balanced to achieve the desired therapeutic effect. Preclinical studies supported by in silico simulations and crystallographic data are continuously refining our understanding of the molecular dynamics underlying receptor–ligand interactions, and this knowledge will be pivotal in addressing these formulation challenges.

There is also an evolving interest in combination therapies. Given that RORG antagonism may only address part of the inflammatory process in autoimmune diseases, it is conceivable that future therapeutic regimens will pair RORG antagonists with other agents—such as conventional immunosuppressants or biologics—to achieve synergistic effects. Such strategies might also mitigate the risk of treatment failure seen in earlier candidates (for example, VTP-43742, which failed in Phase II despite promising early signals). Enhanced characterization of patient-specific molecular profiles could further augment the potential for successful combinatorial approaches by tailoring therapies to individual disease mechanisms.

Ongoing research into the receptor’s allosteric modulation further broadens the scope for future drug design. Structural studies have shown that small molecules binding at allosteric sites offer an alternative method to achieve specificity while potentially reducing competitive interference with endogenous ligands. Future research in this area may open the door to next-generation RORG antagonists with improved efficacy and less off-target toxicity.

The future directions in this field are therefore multi-pronged:
– Optimizing dosing strategies by integrating pharmacodynamic biomarkers with advanced modeling techniques.
– Stratifying patient populations using genomic, proteomic, and immune-profiling approaches to ensure that the right patient subgroups are selected for treatment.
– Exploring combination therapies that leverage the benefits of RORG antagonism while addressing the multifactorial nature of autoimmune diseases.
– Utilizing structural biology and allosteric modulation to design compounds with superior receptor selectivity and fewer side effects.

Each of these future research directions carries the promise of not only improving the clinical management of psoriasis and related autoimmune conditions but also expanding the therapeutic applications of RORG antagonists to other domains, such as oncology, where inflammatory signaling plays a crucial role in disease progression.

Conclusion
In summary, current clinical trials for RORG antagonists prominently feature compounds such as PF-06763809, AZD0284, and JTE-451. These agents have entered various phases of clinical testing largely in the context of treating plaque psoriasis—a model autoimmune disease driven by Th17/IL-17 dysregulation. PF-06763809 is being tested as a topical agent in a Phase I study, while AZD0284 is undergoing both Phase I trials in healthy subjects and Phase 1b studies in psoriasis patients, with its safety, pharmacokinetics, and pharmacodynamics carefully characterized. JTE-451 has the most extensive clinical portfolio, being evaluated in both systemic and topical formulations across Phase I and Phase II trials, with developments such as the IMPACT-PS trial aimed at assessing its long-term efficacy and safety in moderate to severe plaque psoriasis.

From a mechanistic perspective, RORG antagonists work by disrupting the receptor’s active conformation—primarily through interfering with the agonist lock formed by key residue interactions—thus reducing IL-17 production and modulating the inflammatory cascade. The targeted therapeutic applications are broad, with psoriasis being the focal point due to its well-characterized inflammatory pathology. However, there is also an emerging rationale for employing these antagonists in other autoimmune diseases and possibly even in oncology, underscoring the importance of RORG in diverse biological contexts.

Ongoing clinical trials are designed to answer crucial questions on dosing, long-term safety, and efficacy. Although early data indicate promising safety profiles and preliminary efficacy signals—such as improvements in skin lesion parameters and favorable pharmacodynamic outcomes—the challenges that remain include optimal dosing determination, patient stratification through biomarker identification, and managing long-term immunological consequences. Looking forward, future research is geared toward refining compound formulation, exploring combination therapies, and leveraging structural insights for next-generation drug design. This multifaceted clinical and research approach is essential to ensure that RORG antagonists can be effectively translated into safe and efficacious therapies for a range of immune-mediated conditions.

Thus, while the current clinical landscape shows that RORG antagonists such as PF-06763809, AZD0284, and JTE-451 are promising candidates in the treatment of psoriasis, ongoing and future studies are critical to overcoming the inherent challenges of dosing, long‐term safety, and patient variability. The ultimate goal is to develop therapeutics that not only ameliorate the signs and symptoms of autoimmune disorders but also address the underlying molecular dysregulation of RORG activity, thereby offering durable and comprehensive disease management options. This integrated clinical development strategy, underpinned by robust mechanistic insights and a commitment to personalized medicine, forms the cornerstone of the future success of RORG antagonists as a new class of immunomodulatory agents.

In conclusion, the clinical trials currently underway for RORG antagonists represent a significant step forward in the quest to modulate immune responses in autoimmune diseases. The promising early-phase data for PF-06763809, AZD0284, and JTE-451 provide a strong foundation for further research and development. While challenges such as optimal dosing and long-term safety remain, ongoing investigations—bolstered by advanced structural and pharmacodynamic studies—are expected to address these issues and ultimately unlock the full therapeutic potential of targeting RORG. Future research will undoubtedly expand the utility of these compounds beyond psoriasis, offering hope for more effective treatments for a range of diseases where immune dysregulation plays a critical role.

Discover Eureka LS: AI Agents Built for Biopharma Efficiency

Stop wasting time on biopharma busywork. Meet Eureka LS - your AI agent squad for drug discovery.

▶ See how 50+ research teams saved 300+ hours/month

From reducing screening time to simplifying Markush drafting, our AI Agents are ready to deliver immediate value. Explore Eureka LS today and unlock powerful capabilities that help you innovate with confidence.