What are the preclinical assets being developed for GR?

7 March 2025
Introduction to Glucocorticoid Receptor (GR)

Role and Function of GR
The glucocorticoid receptor (GR) is a ligand‐activated transcription factor belonging to the nuclear receptor superfamily. It plays a central role in the regulation of gene transcription in response to circulating glucocorticoids such as cortisol. Upon binding of its ligand, the GR undergoes a conformational change, dissociates from its cytoplasmic chaperone complex, and translocates into the nucleus. There, it binds directly to glucocorticoid response elements (GREs) in the DNA or interacts indirectly with other transcription factors such as NF‑κB and AP‑1 to exert both transactivation and transrepression effects. This dual modality is critical because it allows the GR to promote anti‐inflammatory and immunosuppressive pathways (via transrepression of pro‐inflammatory genes) while also mediating metabolic, developmental, and stress‐related responses (through transactivation of specific genes). In recent years, advances in our understanding of GR structure – including the roles of its N-terminal transactivation domain, central zinc finger DNA-binding domain (DBD), and the C-terminal ligand-binding domain (LBD) – have provided a structural framework for designing compounds that can modulate its activity with improved selectivity. This type of highly selective modulation aims to uncouple the beneficial anti-inflammatory or specific therapeutic effects from the metabolic and other side effects often observed with conventional glucocorticoids.

GR in Disease Pathophysiology
GR signaling is implicated in many physiological and pathological processes. In normal physiology, GR plays a regulatory role in stress adaptation, metabolic homeostasis, and the immune response. Abnormal GR signaling is associated with a range of diseases, including inflammatory disorders such as rheumatoid arthritis, asthma, and chronic obstructive pulmonary disease (COPD), as well as immune dysregulation, mood disorders, and even certain metabolic syndromes. The challenges of chronic high‐dose glucocorticoid therapy include the emergence of side effects such as osteoporosis, hyperglycemia, and muscle wasting. Therefore, there is tremendous interest in developing preclinical assets that target GR in a way that improves the therapeutic index of glucocorticoid‐based interventions – that is, achieving the desired therapeutic benefit (for example, potent anti‑inflammatory activity) while simultaneously mitigating adverse systemic effects.
Moreover, GR dysfunction may contribute to resistance phenomena in acute and chronic inflammatory settings. The concept of GR “uncoupling” – where selective modulation biases receptor signaling toward transrepression (which mediates anti-inflammatory activity) rather than the transactivation pathways linked to many side effects – is a key driver in the development of next-generation modulators. This context has provided both a mechanistic rationale and a platform for the preclinical discovery and development of novel compounds aimed at fine-tuning GR activity for various therapeutic applications.

Current Preclinical Assets Targeting GR

Overview of Preclinical Development
The current landscape of preclinical assets targeting GR is characterized by a multifaceted approach that involves both small-molecule chemical entities and biologics. Advances in high-throughput screening, structure-based drug design, and cell-based reporter assays have enabled researchers to identify compounds with selective GR agonistic or modulatory profiles. One of the novel developments in this area is the use of dedicated screening assays – such as the SEDIGRAM (Selective Dimerizing Glucocorticoid Receptor Agonists and Modulators) assay – which has been designed specifically to identify compounds that promote GR dimerization in a way that favors transrepression over transactivation. Through such assays, compounds like Cortivazol and AZD2906 have been identified as promising candidates that might elicit potent anti-inflammatory activity while reducing the adverse metabolic profile typically linked with traditional glucocorticoids.

Another promising avenue involves the development of antibody-drug conjugates (ADCs) that incorporate GR modulatory mechanisms. For example, ABBV-154 is an innovative preclinical asset that represents a targeted immunology ADC; it combines an antibody moiety with a GR modulator payload and is designed to inhibit tumor necrosis factor (TNF) activity concomitantly with modulating GR signaling to achieve sustained anti-inflammatory effects. These conjugates are engineered to enhance stability – for example, through the use of bromoacetamide instead of maleimide for drug-linker attachment – and to allow for subcutaneous self-administration. Such an approach might eventually enable a more predictable exposure profile with reduced systemic toxicity.

In addition to these modalities, there is also interest in “dual-target” or “combination” approaches. One asset in the early preclinical stage is CLX-A731, a chemical drug developed by Cellix Bio Pvt Ltd., which targets both the GR and the H1 receptor. This asset is being explored as a novel chemical entity that can modulate GR activity while simultaneously impacting histaminergic signaling. The rationale behind this dual modulation is to harness potential synergistic effects in conditions like respiratory or otorhinolaryngologic diseases, where inflammation and allergic components converge. Collectively, these assets exemplify how diverse the strategies are for modulating GR function, with platforms spanning from selective small molecules to complex biologics.

Key Compounds and Their Mechanisms
Among the key compounds currently under preclinical development are:

1. SEDIGRAM-Derived Modulators (e.g., Cortivazol and AZD2906):
The SEDIGRAM screening assay was established to specifically identify GR ligands that induce receptor dimerization in a manner that biases gene regulation toward anti-inflammatory transrepression. Compounds identified using this platform have shown promising in vitro efficacy, achieving potent anti-inflammatory activity while avoiding excessive GR-driven transactivation that usually leads to adverse effects such as hyperglycemia and osteoporosis. Cortivazol has been highlighted in preliminary studies as a compound with a unique profile, displaying strong anti-inflammatory properties with reduced metabolic side effects. Similarly, AZD2906 has emerged as another candidate with comparable properties, positioning these molecules as critical examples of selective GR modulators whose mechanisms are rooted in allosterically rearranging the receptor conformations for beneficial outcomes.

2. ABBV-154 (Anti-TNF GR Modulator Immunology ADC):
ABBV-154 represents a paradigm shift in the development of GR-targeted therapies by leveraging the advantages of ADC technology. This asset couples a monoclonal antibody with a glucocorticoid receptor modulator payload and is specifically designed to concurrently target TNF-mediated pathways and GR signaling. By using advanced linker chemistry – specifically, stable bromoacetamide drug-linkers – ABBV-154 is engineered to closely control the release of the GR modulator once internalized, ensuring long-term stability and sustained pharmacologic activity. The concept behind ABBV-154 is to achieve robust anti-inflammatory and immunomodulatory effects, which may be particularly relevant in diseases where TNF plays a critical role, such as in certain autoimmune conditions or inflammatory cancers.

3. CLX-A731 (Dual GR and H1 Receptor Modulator):
Developed by Cellix Bio Pvt Ltd., CLX-A731 is at the preclinical stage and represents a chemical modality designed to engage both the GR and the H1 receptor. The dual targeting strategy is based on the notion that in specific disease contexts – especially those involving inflammatory components alongside allergic or histaminergic pathways – simultaneous modulation of these receptors can yield enhanced therapeutic benefits. The compound acts by modulating GR signaling in a way that can potentially enhance anti-inflammatory outcomes while concurrently dampening adverse histamine-driven responses. Preclinical data supporting its efficacy and safety in relevant models are currently being generated.

4. Other Emerging Entities and Combination Strategies:
Although the assets highlighted above are among the most prominent, the preclinical pipeline also includes several additional small molecules and biologics at various stages of discovery. Some groups are investigating the use of GR modulators in a combinatorial context with other therapeutic agents, including targeted kinase inhibitors or immunomodulators. These efforts often aim to exploit synergistic interactions that might lower the required dosage of each individual agent—thus reducing toxicity and resistance—while maximizing therapeutic efficacy. In some experimental platforms, researchers are also exploring novel drug delivery systems (such as nanoparticle formulations) to enhance targeting to tissues where GR modulation will be most beneficial. These strategies, while still in the early stages, have the potential to further expand the preclinical asset portfolio targeting GR.

Development Stages and Evaluation

Preclinical Testing and Models
Preclinical evaluation of GR-targeted assets involves a series of rigorous and sequential studies designed to validate their mechanism of action, assess potency, and ensure acceptable safety profiles before these assets can be advanced into clinical testing. The development process is inherently multi-stage and spans from high-throughput cellular assays to advanced in vivo models.

Initially, candidate compounds are typically screened in vitro using cell-based reporter assays. These assays are engineered to monitor the activation or repression of specific GRE-dependent gene transcription, thereby providing an early readout of the compound’s ability to modulate GR activity. For example, the SEDIGRAM assay does not only quantify receptor dimerization but also evaluates downstream transrepression effects, which are considered crucial for anti-inflammatory efficacy. Such in vitro models are augmented by pharmacodynamic studies that involve measuring the expression levels of GR target genes and assessing potential off-target effects.

Following in vitro assays, promising candidates are advanced to animal models. Murine models are the most common in this domain, as they allow for the validation of GR-mediated activity in the context of complex biological systems where factors such as tissue-specific receptor expression and systemic healing responses are at play. Animal models are used in two major contexts: efficacy models and safety/toxicology models. For efficacy, inflammation models (such as LPS-induced endotoxemia or models of autoimmune arthritis) are frequently employed to measure parameters like cytokine levels, inflammatory cell infiltration, and histopathological alterations in target tissues. For safety evaluation, standard toxicological assessments are combined with pharmacokinetic analyses to determine metabolic clearance, tissue distribution, and maximum tolerated doses.

Furthermore, some preclinical studies incorporate imaging modalities and biomarker assessments to establish a relationship between the drug exposure and its pharmacodynamic impact. For instance, the evaluation of ABBV-154 has been supported by detailed biodistribution and stability studies that utilize advanced imaging techniques to track the ADC distribution in vivo and correlate it with improvements in inflammatory biomarkers. Such multi-tiered testing ensures that only the assets with the most favorable profiles advance to further development stages.

In safety evaluation, assessment parameters are not limited to classical measures such as body weight and clinical chemistry but extend to investigating off-target gene modulation – particularly given the pleiotropic nature of GR signaling. Regulatory endpoints include markers of metabolic changes, bone density assessments, and evaluation of immunosuppressive indices. This comprehensive approach to preclinical evaluation guarantees that the assets have been scrutinized from various angles prior to entering human clinical trials.

Efficacy and Safety Assessments
The efficacy assessment of GR-targeted preclinical assets is centered on their ability to achieve potent transrepression in inflammatory models while minimizing activation of transactivation pathways that lead to adverse systemic effects. For assets like the SEDIGRAM-derived modulators, efficacy is determined by measuring the suppression of pro-inflammatory cytokine production as well as histological improvements in inflamed tissues. For example, in acute inflammation models, compounds such as Cortivazol have been tested for their effect on reducing inflammatory mediator levels and mitigating tissue damage without producing significant hyperglycemia or adverse metabolic profiles.

For ABBV-154, efficacy is evaluated using its ability to simultaneously inhibit TNF signaling and modulate GR activity. Preclinical studies in animal models demonstrate that such ADCs can achieve sustained reductions in inflammatory biomarkers over extended periods, a factor that is especially desirable in chronic conditions where long-term management is required. Studies also measure endpoints like the duration of receptor occupancy, the half-life of the conjugate in circulation, and the magnitude of TNF suppression in relevant tissues.

Safety assessments of these preclinical assets are equally comprehensive. Beyond standard toxicological endpoints, researchers evaluate the potential for metabolic dysregulation, immune suppression, or unintended gene expression changes. For instance, the dual-targeting asset CLX-A731 undergoes extensive testing to ensure that its H1 receptor modulation does not interfere adversely with GR-mediated metabolic processes. In depth toxicology studies include dose-escalation trials in rodents, non-rodent species, and eventual exploratory studies in larger animals. Evaluation parameters include serum chemistry, hematology, histopathological examination of key organs (e.g., liver, kidney, and bone), and behavioral assessments to detect possible central nervous system effects.

Other innovative assessment methodologies include the use of “omics” techniques – such as transcriptomics and proteomics – to capture the breadth of the compounds’ effects on cellular pathways. In the case of GR modulators, these techniques can help distinguish selective modulation from full-scale receptor activation or blockade, thus providing a molecular fingerprint that correlates with the therapeutic index of the candidate asset. Such approaches are being increasingly incorporated into preclinical programs to support the rational selection of candidates and to provide a foundation for the eventual translation into clinical biomarker studies.

Challenges and Future Prospects

Current Challenges in GR Targeting
Despite significant progress, several challenges remain in the development of preclinical assets targeting GR. One of the primary issues is achieving the desired degree of selectivity. Conventional glucocorticoids, while effective, produce a wide range of side effects by activating both beneficial and deleterious GR-mediated pathways. The challenge for next-generation modulators is to bias receptor pharmacology – promoting transrepression while minimizing transactivation. Although assays such as SEDIGRAM have advanced the identification of candidate molecules with a more favorable signaling profile, the intricate network of coregulator interactions and tissue-specific gene expression patterns continues to complicate the prediction of in vivo outcomes.

Another challenge is the translation of preclinical effects into clinically meaningful endpoints. Animal models, despite their utility, often fail to fully recapitulate the complex pathophysiological environment of human diseases. As a result, assets that demonstrate impressive efficacy in murine or in vitro systems may encounter unforeseen hurdles in human clinical trials. This “translational gap” is compounded by differences in GR expression levels, receptor isoform distribution, and metabolite processing between species. Consequently, the design of preclinical studies now increasingly incorporates humanized models and comprehensive multi-parameter assessments in order to better predict human outcomes.

Additionally, the safety profile of GR modulators remains a critical area of concern. Even highly selective compounds may trigger subtle alterations in gene expression that have long-term consequences, such as immune suppression or metabolic alterations. For ADCs like ABBV-154, issues of immunogenicity, off-target toxicity, and the stability of the linker-drug complex need to be addressed thoroughly. Similarly, dual-targeting approaches such as those employed by CLX-A731 must demonstrate that simultaneous modulation of two different receptor systems does not lead to unanticipated interactions that could compromise safety.

Pharmacokinetic challenges also persist, particularly for compounds that must maintain sustained receptor occupancy and appropriate tissue distribution without accumulating to toxic levels. The development of nanoformulations or advanced drug delivery systems might offer solutions, but these too require extensive validation. The dynamic regulation of GR, including its cyclic activation and auto-downregulation, further complicates dosing regimens and necessitates well-designed preclinical studies that can capture these nuances.

Future Directions and Innovations
Looking ahead, the future of preclinical GR asset development is bright with promising avenues for innovation. One major direction is the integration of advanced screening technologies, such as high-content imaging, high-throughput transcriptomics, and computational modeling, to guide the rational design of molecules with optimized GR selectivity. These methods not only enhance the identification of promising candidate compounds – as exemplified by the SEDIGRAM assay – but also allow for the customization of compounds based on structure–activity relationships that predict downstream pathway activation patterns.

Another emerging trend is the refinement of antibody-drug conjugate (ADC) technologies. For instance, the development of ABBV-154 has demonstrated that with the proper selection of linker chemistry and payload, it is possible to achieve a balanced modulation of GR activity with improved stability and favorable pharmacokinetics. Future innovations may include the use of bispecific antibodies or dual-drug conjugates that can target both GR and synergistic inflammatory or oncogenic pathways, thereby providing a multi-pronged approach to complex conditions such as autoimmune disorders or inflammation-associated cancers.

There is also an increasing focus on combination strategies. Preclinical research now often explores the concurrent modulation of GR signaling along with other targets – for example, pairing GR modulators with targeted kinase inhibitors or immune checkpoint blockers. Such combinations are designed to produce synergistic effects that not only reduce the required dose of the GR-targeted agent (and hence its side effects) but also enhance overall therapeutic efficacy. The integration of precise biomarker-based assessments into these combination studies is likely to accelerate the translation from bench to bedside.

On the drug delivery front, nanotechnology and advanced vehicle systems are likely to play an increasingly important role. Nanocarriers, liposomal formulations, and targeted delivery systems are being developed to ensure that GR-modulating compounds achieve optimal tissue localization. This is critical for both efficacy and safety, as it minimizes systemic exposure and concentrates the active agent in the diseased tissue. By fine-tuning the pharmacokinetic profiles of these agents, researchers aim to maximize receptor occupancy during critical periods of disease activity while avoiding sustained high systemic concentrations that could lead to adverse effects.

Finally, the future research landscape will benefit from the convergence of interdisciplinary fields. The integration of systems biology, epigenetics, and artificial intelligence into preclinical research is poised to provide a more holistic view of GR signaling networks. Such approaches can help identify novel regulatory nodes within the GR pathway that may serve as additional targets or that may act as predictive biomarkers for treatment response. For example, using “omics” technologies to map the global change in gene expression following selective GR modulation could uncover new therapeutic opportunities and guide the design of next-generation compounds with finely tuned action profiles.

Conclusion
In summary, the development of preclinical assets targeting the glucocorticoid receptor (GR) represents a rapidly evolving field with broad therapeutic potential. GR is a critical regulator of gene expression in response to glucocorticoids, influencing a vast array of physiological processes including inflammation, metabolism, and immune regulation. Because conventional glucocorticoid therapies are burdened by side effects resulting from non-selective activation of GR pathways, current preclinical efforts are focused on developing selective modulators that bias receptor signaling toward beneficial transrepression while avoiding deleterious transactivation.

Key preclinical assets in this arena include selective small molecules identified via advanced screening assays such as the SEDIGRAM platform, which has yielded promising candidates like Cortivazol and AZD2906. These compounds are designed to induce a receptor conformation that favors anti-inflammatory gene repression and minimize metabolic side effects. In parallel, biologics such as ABBV‑154—a novel antibody-drug conjugate engineered to couple GR modulation with TNF suppression—and dual-targeting small molecules like CLX‑A731 that engage both GR and the H1 receptor are under active development. Such assets are being rigorously evaluated in state-of-the-art in vitro and in vivo models to address key pharmacodynamic and pharmacokinetic questions, and to ascertain their efficacy and safety profiles in complex biological systems.

Despite these significant advances, challenges remain. The intricate regulation of GR activity and the need for precise modulation to avoid adverse effects require the development of robust preclinical models and innovative screening methodologies. Translational hurdles, including species differences in GR signaling and receptor isoform distribution, remain obstacles that must be surmounted through the deployment of humanized models and comprehensive “omics” studies. Additionally, the evolving nature of drug delivery systems and combination approaches demands a careful balance between efficacy and safety.

Looking into the future, the integration of cutting-edge technologies—including high-content imaging, systems biology, and nanotechnology—with classical preclinical testing paradigms promises to further refine the discovery and optimization of GR-targeted agents. The continued development of advanced ADCs and combination therapies appears particularly promising, as does the exploration of innovative biomarker-based strategies for the prediction of treatment response. These efforts, collectively, are paving the way for the next generation of GR modulators that offer enhanced therapeutic efficacy with a greatly improved safety profile.

In conclusion, preclinical assets targeting the GR are being developed through a broad spectrum of innovative strategies that include selective small molecule modulators, antibody–drug conjugates, and dual-receptor targeting agents. These assets are undergoing extensive evaluation across multiple preclinical models that mimic complex human disease states. While challenges such as ensuring selectivity, translating preclinical efficacy to human outcomes, and optimizing drug delivery remain, the ongoing advances offer tremendous promise. With an integrated general-to-specific-to-general approach that builds on robust screening, rigorous animal model evaluation, and advanced drug design, the future of GR-targeted therapies looks set to provide novel solutions for inflammatory, immune, and metabolic disorders while improving patient safety and therapeutic outcomes.

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.