What are the preclinical assets being developed for AR?

11 March 2025
Introduction to Androgen Receptor (AR)

Biological Role and Mechanism
The androgen receptor (AR) is a ligand‐activated transcription factor critical for mediating the actions of androgen hormones such as testosterone and dihydrotestosterone (DHT). Under normal physiological conditions, AR regulates gene expression pathways that are essential for male sexual differentiation, maintenance of secondary sex characteristics, muscle growth, and cell survival. Binding of androgens to AR causes a conformational change that allows its dissociation from heat‐shock proteins, translocation into the nucleus, dimerization, binding to DNA response elements, and subsequent transcriptional regulation of target genes. The structural complexity of AR is underscored by its modular domains, including the N‐terminal domain (NTD), DNA‐binding domain (DBD), and the ligand‐binding domain (LBD). Each of these plays a distinct yet integrated role in receptor signaling. In particular, while the LBD has traditionally been the most druggable segment given its defined pocket, the NTD remains intrinsically disordered and represents a challenging yet promising target to overcome therapeutic resistance, especially since many shorter AR splice variants (AR‐Vs) lack the LBD but still retain transcriptional activity. The cascade of events triggered by androgen binding involves recruitment of coregulators, chromatin remodeling, and integration with other signaling cascades such as PI3K/AKT, which adds a further layer of complexity to AR signaling.

Importance in Disease Contexts
In disease contexts – notably prostate cancer – AR is not only a driver of normal growth and differentiation but also a central player in oncogenic transformation and progression. Under abnormal conditions, AR signaling contributes to the development of both androgen‐dependent and androgen‐independent (castration‐resistant) prostate cancer (CRPC). The persistent expression of AR and the occurrence of mutations that allow promiscuous ligand interactions or generate constitutively active splice variants make AR a prime target for therapeutic intervention. Indeed, despite the effectiveness of androgen deprivation therapies (ADT) in the earlier stages of prostate cancer, resistance invariably emerges owing to various mechanisms including AR overexpression, mutation and generation of AR‐Vs that lack a functional LBD. This immuno‐oncological paradigm has led to a surge in efforts to develop agents that can more effectively inhibit or degrade AR and its variants to delay disease progression and improve patient outcomes. Beyond prostate cancer, AR has been implicated in other conditions such as breast cancer, muscle atrophy and even metabolic disorders. Consequently, efforts to develop molecular assets targeting AR can have tremendous clinical ramifications across several disease areas.

Current Preclinical Assets Targeting AR

Overview of Preclinical Development
In recent years, the preclinical development of AR‐targeted assets has evolved significantly with innovative strategies aiming to overcome the limitations of conventional LBD‐targeting drugs. Early drug development focused on small molecule competitive inhibitors that bind to the ligand‐binding pocket. However, as resistance mechanisms have emerged – notably through AR mutations and the presence of AR splice variants – research has pivoted toward targeting other critical domains of AR. Notably, preclinical assets now include agents that directly target the intrinsically disordered N‐terminal domain (NTD) and proteins that can induce degradation of both full‐length AR (AR‐FL) and AR variants, termed selective AR downregulators (SARDs).

Preclinical development has been documented in both peer‐reviewed papers and patent literature. Multiple patents describe androgen receptor modulators of formula (I) aimed at treating prostate cancer and other AR‐related diseases. These patents present pharmaceutical compositions that include AR modulators in combination with other therapies. They reflect the broad preclinical interest in harnessing AR modulation to prevent disease progression in preclinical models. In parallel, academic literature surveys have provided insights into the novel chemical entities and approaches devised to target AR via domains outside of the classical therapeutic pocket.

Further complicating the asset landscape is the differentiation of preclinical assets not only by chemical structure (small molecule, peptide, or bifunctional conjugate) but also by their mechanisms—ranging from antagonism and degradation to allosteric modulation and modulators that dissociate AR from coregulator complexes. A subset of these compounds is designed to overcome resistance in CRPC by targeting the NTD, which remains intact in truncated AR‐Vs. Thus, leveraging advancements in structural biology and computational chemistry, numerous compounds with promising in vitro and in vivo efficacy profiles are being evaluated in preclinical models before advancing to early‐phase clinical trials. These preclinical evaluations include assessments of binding affinity, degradation efficiency, transcriptional inhibition, and synergy with other anticancer agents in xenograft and genetically engineered mouse model systems.

Key Compounds and Their Mechanisms
A range of key compounds is emerging from preclinical research targeting AR through various mechanisms. One major area of focus is noncompetitive AR inhibition by direct targeting of the N‐terminal domain. For example, several studies have reported the discovery and preclinical evaluation of small molecule inhibitors that disrupt protein–protein interactions essential for AR transcriptional activity. Early compounds like EPI‐001 laid the groundwork for targeting the NTD but were later superseded by more potent analogs such as EPI‐7386, which exhibits improved metabolic stability and oral bioavailability. EPI‐7386 has demonstrated potent antitumor activity in several AR‐V7–positive CRPC cell lines and xenograft models, indicating strong potential to surmount resistance seen in first‐generation agents.

Complementing the NTD inhibitors, additional compounds are being developed that induce AR degradation. Some preclinical assets operate as selective degraders of both AR‐FL and AR‐Vs. For instance, research highlighted in a patent compared multiple compounds via their IC50 values and degradation efficacy. A compound listed as “50” demonstrated high potency – an IC50 value of 84 nM – accompanied by robust degradation of both wild‐type AR and AR splice variants. This type of asset is strategically important because it not only blocks AR-mediated signaling but actively reduces AR protein levels, thereby decreasing the possibility of ligand-independent activation and subsequent cancer cell survival.

Another promising preclinical asset is the dual‐action androgen receptor inhibitor (DAARI) ONCT-534. This compound is in advanced preclinical development as a potential treatment for castration-resistant prostate cancer. ONCT-534 has been characterized as having a unique mechanism that combines AR antagonism with degradation of receptor variants. Preclinical studies indicate that ONCT-534 can effectively reduce AR signaling in models that express both AR-FL and splice variants, providing a robust approach to attenuate the growth of castration-resistant tumors. Although the primary descriptions of ONCT-534 are found in news releases and company reports, they have been substantiated in preclinical experiments that include detailed cellular and animal model evaluations.

Additionally, some compounds use bifunctional strategies, such as the incorporation of antibody–drug conjugates (ADCs) that target AR signaling pathways while simultaneously delivering cytotoxic payloads to AR-expressing cells. While these approaches are still nascent in terms of clinical application, preclinical studies have shown promise in utilizing ADC technology in precision oncology contexts, thereby broadening the therapeutic window for AR modulators.

A separate group of preclinical assets comprises compounds designed to disrupt the interaction between AR and its key coactivators or to modulate downstream epigenetic mechanisms. For instance, inhibitors targeting the CBP/p300 coactivator complex have been shown to suppress AR transcriptional activity by reducing histone acetylation marks necessary for gene transcription. Their potential use, combined with existing AR antagonists, could provide synergistic effects in resisting castration-resistant mechanisms.

Overall, the key preclinical assets being developed represent an array of strategies:
– Direct targeting of the NTD using compounds like EPI-7386 that block AR activity even in splice variants.
– AR degradation strategies that lower the levels of AR protein through compounds with high-target-binding affinity and degradation efficacy.
– Dual-action inhibitors such as ONCT-534 that combine AR antagonism with receptor degradation, addressing resistance profiles in advanced disease models.
– Conjugate-based approaches and co-regulator disruptors that provide alternative ways to interfere with AR-driven transcriptional programs, possibly enhancing the efficacy profile when used in combination therapies.

Evaluation of Preclinical Assets

Efficacy and Safety Profiles
Preclinical evaluation of these assets—typically conducted in both in vitro assays and in vivo models—has provided critical insights into their efficacy and safety profiles. Potent AR inhibitors are characterized by low IC50 values in biochemical and cell-based assays, effective reduction in AR-mediated transcription, and the ability to degrade both AR-FL and its splice variants. For instance, compound “50” demonstrated a significant degradation efficacy with more than 70% reduction in AR protein levels at concentrations as low as 1 μM, with its IC50 falling in the sub-100 nM range. In animal models of prostate cancer, such as xenograft models derived from enzalutamide-resistant VCaP cells, these compounds have led to marked tumor growth inhibition (often exceeding 80% reduction in tumor volume under optimal dosing conditions) and reduced serum prostate-specific antigen (PSA) levels, which serve as surrogate markers for AR activity.

Safety profiles of these preclinical agents have also been rigorously evaluated in rodent models as well as in secondary screening systems. Studies have assessed cytotoxicity in normal prostate epithelial cell lines versus malignant cells to ensure a therapeutic index that spares normal tissues. Early evidence suggests that targeted AR degraders and NTD inhibitors have favorable safety margins, with few observed off-target toxicities at efficacious doses. Nonetheless, because AR signaling plays a role in normal tissue function, careful dose optimization and combinatorial regimens are required to maximize tumor inhibition while minimizing endocrine side effects. Detailed pharmacokinetic studies and assessment of hepatic and renal enzyme interactions have been carried out for compounds such as ONCT-534 to ensure that they exhibit predictable drug exposure without accumulation of toxic metabolites.

Another critical factor in efficacy and safety assessment is the ability of these preclinical assets to overcome resistance mechanisms that arise during long-term treatment with conventional AR antagonists. The promising preclinical data showing that NTD inhibitors like EPI-7386 maintain activity in the presence of AR splice variants provides an important proof-of-concept that these agents can provide durable efficacy. Animal pharmacology studies also support that AR degraders are capable of rapidly lowering AR protein levels, resulting in improved tumor regression in models that have historically been resistant to competitive ligands.

It is also notable that combination studies are underway in preclinical settings to evaluate whether these compounds can be co-administered with other agents—such as AKT or mTOR inhibitors—to provide synergistic antitumor activity while potentially alleviating toxicities by lowering required doses. Such combination strategies are proving to be a critical area of investigation, as they integrate safety profiling with an enhanced understanding of network-wide effects in tumor biology.

Potential Therapeutic Applications
The primary therapeutic application of these preclinical assets is in the treatment of prostate cancer, particularly in castration-resistant cases. Preclinical studies have established that agents capable of targeting the AR beyond the conventional LBD are likely to be effective in tumors that express mutated AR or AR splice variants such as AR-V7, which are associated with poor prognosis and resistance to approved therapies. Aside from prostate cancer, there is growing interest in applying AR-targeted interventions to other AR-related disorders. For example, AR modulators are being evaluated for their potential utility in treating certain breast cancers that express AR, as well as other hormone-driven conditions where aberrant AR signaling contributes to pathological cell proliferation or tissue dysfunction.

In animal models, the demonstration of significant tumor regression, reduction in PSA levels, and complete abrogation of AR-mediated transcription heralds a robust therapeutic window for these agents. Furthermore, the potential for combining AR inhibitors with standard chemotherapeutic regimens or novel immune-oncology agents opens new avenues for treating tumors with heterogeneous AR dependence. Additional preclinical studies using patient-derived xenograft models and genetically engineered mouse models have underscored the possibility of tailoring therapeutic approaches based on the unique AR mutation or splice variant profile of a given tumor.

The concept of using AR degraders as part of combinatorial treatment regimens has gained traction, especially as the industry moves toward precision oncology. Here, biomarkers such as AR variant status or the ratio of full-length to truncated receptor forms can guide therapy selection and dose optimization. Beyond oncology, the modulation of AR activity has implications in the treatment of other hyperproliferative or endocrine disorders. Some preclinical assets, for instance, are being explored for efficacy in conditions such as muscle wasting, certain metabolic syndromes, and even benign prostatic hyperplasia, where aberrant AR signaling contributes to the pathophysiology.

Moreover, recent preclinical innovations have also explored the encapsulation of AR-targeting molecules into novel delivery systems. These nano-formulations aim to improve the bioavailability and tumor selectivity of compounds like ONCT-534, thus reducing systemic exposure and potential off-target effects. Such drug delivery methods are under active investigation in cellular and animal models and represent an emerging trend in the era of targeted therapies.

Preclinical studies reported in the literature indicate that these assets not only suppress AR transcriptional activity effectively but also induce apoptosis and cell-cycle arrest in cancer cells that depend on robust AR signaling for survival. The overall therapeutic promise of these compounds is evaluated not just by direct tumor response but also by improvements in quality of life surrogates, such as lower toxicities and improved management of treatment-induced side effects commonly seen with conventional ADT.

Challenges and Opportunities

Developmental Challenges
Although the momentum in preclinical drug development for AR-targeted therapies is robust, a number of challenges persist. One of the primary obstacles is the intrinsic complexity associated with targeting the structurally disordered N-terminal domain of AR. Unlike the well-folded ligand-binding domain, the NTD lacks a well-defined binding pocket. This has historically rendered it “undruggable” by conventional small molecules and necessitated novel screening paradigms and structure-guided computational methodologies to identify effective inhibitors.

Moreover, the heterogeneity in AR mutations and the dynamic expression of AR splice variants in advanced cancer pose significant challenges for preclinical asset evaluation. In many cases, the same tumor may express multiple forms of AR with diverse sensitivities to various inhibitors, thereby complicating the selection of a lead candidate. Such biological variability requires extensive preclinical profiling using multiple cell lines and animal models to capture the spectrum of AR dependency.

A related challenge is the issue of bioavailability, especially for compounds like EPI-506 (the precursor to EPI-7386) and ONCT-534. Many preclinical compounds that showed high in vitro potency suffered from poor oral bioavailability and rapid metabolism in vivo, leading to suboptimal exposures in clinical trials. Determining the optimal dosing regimen that maximizes target engagement while minimizing off-target effects remains a critical hurdle.

Another developmental challenge is the potential redundancy and compensatory mechanisms within cancer signaling networks. AR signaling interacts with multiple pathways, including PI3K/AKT, MAPK, and epigenetic regulators. Cancer cells can often bypass AR inhibition by upregulating alternative pathways, thereby diminishing the long-term efficacy of standalone AR-targeting compounds. This interplay necessitates the exploration of combination strategies in preclinical models, but it also increases the complexity of evaluating safety and efficacy when multiple agents are administered concurrently.

Furthermore, challenges in establishing robust and predictive animal models remain. Although advanced genetically engineered mouse models and patient-derived xenografts have improved preclinical predictions, there is still an imperfect translation from animal data to human outcomes. The high attrition rate observed in early clinical studies underscores the need for more sophisticated preclinical models that better mimic the tumor microenvironment, immune context, and heterogeneity seen in human cancers.

A final challenge involves the regulatory and intellectual property landscapes. With numerous patents covering AR modulators and degraders, there is potential overlap and competition among similar chemical entities. This can slow down the translation of promising preclinical assets to clinical development due to licensing issues and the need for clear differentiation in mechanism of action.

Future Research Directions
Despite these challenges, considerable opportunities exist to further improve and refine preclinical AR-targeted assets. On the technical front, advances in high-throughput screening methods—including both target-driven and phenotypic screening—are likely to accelerate the discovery of novel AR inhibitors. The integration of artificial intelligence and machine learning for predicting ligand docking and ADME (absorption, distribution, metabolism, and excretion) properties will enable researchers to optimize candidate compounds more efficiently. Already, systems for predicting compound-target interactions based on large datasets are influencing candidate selection and prioritization in preclinical pipelines.

One promising research direction involves the development of multi-domain targeting agents. Instead of focusing exclusively on the LBD or NTD, future compounds may integrate bifunctional or multitargeted strategies—combining AR antagonism with degradation or downregulation of coactivator interactions. This might involve the development of PROTACs (proteolysis-targeting chimeras) to promote the ubiquitination and subsequent degradation of AR. The concept of using bifunctional degraders has already gained traction in other therapeutic areas, and ongoing research aims to improve the selectivity and oral bioavailability of these compounds. Enhanced preclinical models have been developed to assess the degradation efficacy and downstream effects on AR signaling comprehensively.

Future research is also poised to explore combinatorial therapies more thoroughly. Given that resistance to AR-targeted monotherapies is frequently observed, integrating AR degraders or NTD inhibitors with inhibitors of key downstream kinases (such as AKT, mTOR, or PLK1) can create a synergistic blockade of oncogenic signaling. In preclinical mouse models of CRPC, such combination strategies could lead to improved tumor regression and delay the emergence of resistance. In this context, prospective biomarkers for patient stratification and response monitoring (such as specific AR variant expression profiles or circulating PSA levels) will be essential to tailor such combination therapies.

Exploiting nano-formulation and targeted drug delivery systems represents another opportunity. By encapsulating AR inhibitors in nanoparticle carriers or conjugating them to tumor-targeting antibodies, researchers aim to improve drug distribution to tumor tissues while reducing systemic side effects. Preclinical studies have begun to show that such delivery systems can enhance the therapeutic index of AR-targeted compounds, making them viable candidates for translational development.

Furthermore, emerging research is focusing on elucidating the three-dimensional structural dynamics of AR and its interactions with various ligands and co-regulators. Advances in cryo-electron microscopy and X-ray crystallography are gradually unraveling the transient conformations of the NTD and the dynamic interfaces with coactivators. Such structural insights will directly feed into the rational design of next-generation AR inhibitors that are both highly selective and potent in disrupting AR signaling complexes.

Collaborative efforts between academic institutions, drug development companies, and computational biology groups are paving the way for more sophisticated, integrative approaches to AR-targeted drug design. These collaborations will likely result in a new class of preclinical assets that not only target AR more effectively but also have improved pharmacologic profiles in terms of bioavailability and safety. A significant part of future development will be the rigorous validation of these agents in advanced preclinical models that more accurately recapitulate tumor heterogeneity and the tumor microenvironment, thereby increasing the likelihood of successful clinical translation.

Finally, leveraging the power of “explainable AI” may help in predicting adverse events and therapeutic windows of these compounds, allowing for dynamic dosing adjustments during preclinical testing that mimic clinical scenarios. Such systems will revolutionize preclinical research by enabling integrated analysis of multi-omics datasets, pharmacokinetics, and toxicity profiles simultaneously, making the entire preclinical process more efficient and reliable.

Conclusion
In summary, the preclinical assets developed for targeting the androgen receptor are highly diverse and address long-standing challenges associated with conventional therapies. Beginning from a detailed understanding of AR’s biological role—a receptor critical for both normal physiology and disease progression in conditions like prostate cancer—a robust pipeline of assets now targets various domains of the receptor. Advances have moved beyond traditional LBD inhibitors to embrace NTD inhibitors, AR degraders, and dual-action agents like ONCT-534 that show promise in both biochemical assays and animal models.

More specifically, the key compounds under investigation include next-generation NTD inhibitors (for example, EPI-7386) that overcome the limitations imposed by the absence of the LBD in splice variants, as well as compounds engineered for degradation of AR protein. These compounds have demonstrated impressive efficacy in preclinical studies by reducing AR-mediated transcription, lowering PSA levels, and inducing significant tumor regression in xenograft and genetically engineered mouse models. Safety profiles appear promising with manageable toxicities, and innovative drug delivery systems are under development to enhance the bioavailability and precise targeting of these agents.

However, challenges remain. The structural disorder of the NTD, the heterogeneity of AR mutations and splice variants, and issues related to bioavailability continue to complicate preclinical asset development. The need for more predictive in vitro and in vivo models, improved combination therapy approaches to address redundant signaling pathways, and advances in computational and AI-driven methodologies to optimize compound design and predict adverse events are critical research directions for the future.

Overall, while the development of AR-targeted preclinical assets has progressed from traditional small molecule antagonism to innovative strategies involving degradation and multi-domain targeting, a concerted effort combining structural biology, advanced modeling, novel delivery systems, and rigorous preclinical testing will be required to translate these promising assets into effective clinical therapies. A comprehensive and collaborative research approach, supported by robust preclinical data and integrative AI-based analyses, holds the potential to not only advance therapeutic outcomes for prostate cancer but also to address other AR-mediated disease contexts, offering a truly transformative impact on precision oncology and beyond.

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.