What are the preclinical assets being developed for GPRC5D?

11 March 2025
Introduction to GPRC5D

Definition and Biological Role

GPRC5D is a member of the G protein-coupled receptor (GPCR) family and is classified as a Class C orphan receptor. Although its endogenous ligand and complete signaling mechanisms remain largely elusive, GPRC5D has been recognized for its unique expression pattern. Unlike many GPCRs that are broadly expressed in various tissues, GPRC5D shows limited expression in normal tissues such as skin (particularly in hair follicles and eccrine glands) and testes (in seminiferous tubules) while being overexpressed on malignant plasma cells, particularly in multiple myeloma (MM) patients. This distinctive biological role not only defines the basic receptor function but also makes GPRC5D an attractive target for therapeutic intervention. Its high expression on malignant cells, with a membranous distribution, positions it as a potential marker for tumor specificity, thereby reducing the possibility of on-target/off-tumor toxicities that are often encountered with targets expressed in both healthy and diseased tissues.

Importance in Drug Development

The receptor’s confined expression profile has sparked considerable interest in translational research and drug development. Given that conventional therapeutic targets such as B-cell maturation antigen (BCMA) may eventually suffer from antigen escape in multiple myeloma, GPRC5D emerges as an attractive alternative target. The therapeutic rationale behind targeting GPRC5D builds on its tumor-restricted expression: its modulation is hypothesized to result in potent anti-tumor responses while limiting the exposure of normal tissues to active agents. This concept is supported by evidence demonstrating that even low-level expression in normal tissues does not translate into significant toxicity, thus broadening the therapeutic window. In addition, the receptor’s involvement in various signaling pathways, even though not yet fully elucidated, opens up multiple avenues for pharmacological modulation with antibody-based agents, bispecific and trispecific T-cell engagers, and cell therapy modalities such as chimeric antigen receptor (CAR)-T or CAR-NK cell therapies. Increasingly, organizations are investing in preclinical programs that focus on leveraging these characteristics to generate novel assets that can overcome the limitations of current therapies in hematologic malignancies and potentially extend to solid tumors where aberrant GPRC5D expression may be present.

Preclinical Assets Targeting GPRC5D

Current Developments

In the past few years, the preclinical and early translational landscape for GPRC5D has seen a surge in the development of numerous innovative assets. Several companies have embarked upon early-stage development programs that focus on distinct modalities to target GPRC5D. Among these are projects reported in the Synapse database that outline the advancement of candidates from major pharmaceutical companies and specialized biopharmaceutical firms. For instance, Bristol Myers Squibb, Sanofi, Sanofi-Aventis U.S. LLC, and others have reported preclinical activities with documented phase times spanning from late 2023 to late 2024, reflecting an industry-wide momentum in exploring GPRC5D as a drug target. In parallel, companies such as Genor Biopharma Co., Ltd., Antengene Corp. Ltd., Integral Molecular, Inc., and Shanghai Symray Biopharma Co., Ltd. are also actively engaged in early development, with phase times indicating significant preclinical investment and strategic risk-taking to enable future clinical translation.

Preclinical development efforts are being demonstrated in several formats. More than one translational medicine abstract presented at leading conferences such as AACR 2024 highlights the engineering and preclinical validation of assets that combine potent anti-tumor activity with favorable safety profiles. For example, one abstract titled “Abstract 2727: Pre-clinical development of a novel anti-GPRC5D inducing potent anti-tumor effect through enhanced antibody-dependent cell-mediated cytotoxicity (ADCC) for multiple myeloma” outlines a candidate that harnesses enhanced ADCC, which is a promising approach for targeting cancer cells while engaging the host immune system. Similarly, another abstract—“Abstract LB128: A novel tri-specific T cell engager targeting BCMA and GPRC5D for treatment of multiple myeloma”—shows that preclinical efforts are not limited merely to traditional monoclonal antibodies. These emerging assets are designed to simultaneously engage multiple targets, thereby offering an innovative means to overcome antigen escape and resistance mechanisms observed in current clinical treatments.

In addition to these antibody and bispecific formats, there is substantial work ongoing in the arena of cellular therapies. Novel immunotherapeutic platforms, such as CAR-T and CAR-NK cells targeting multiple antigens including GPRC5D, are under preclinical investigation. One prominent example is the CAR-NK candidate, CYTO NK 301, developed by Cytoimmune Therapeutics. This asset is engineered to target a combination of antigens (BCMA, GPRC5D, and NKG2D), designed to produce a robust immune response against cancer cells while minimizing off-target effects. Such cellular therapies are being designed with a comprehensive understanding of tumor immunology and are supported by preclinical in vitro and in vivo models assessing cytotoxicity, cytokine production, and overall therapeutic efficacy.

Another integrative approach under preclinical evaluation involves the generation of multispecific antibodies, including anti-GPRC5D/CD3 bispecific constructs, which redirect T cells towards malignant cells expressing GPRC5D. These bispecific antibody platforms have evolved considerably in recent years, and several patent filings have been observed that detail methods to produce antibodies or antigen-binding fragments with high specificity for GPRC5D. Collectively, these preclinical assets underscore a robust and diverse pipeline that is striving to optimize therapeutic interventions for diseases where GPRC5D plays a central role.

Lead Compounds and Molecules

The lead molecules emerging from preclinical investigations present a finely tuned array of modalities geared toward modulating GPRC5D activity. One significant class of agents includes antibody-based molecules that have been refined for high binding affinity and potent effector functions. For example, multiple research outputs and patents detail anti-GPRC5D antibodies that have been engineered to mediate ADCC, offering a targeted method to harness the immune system in order to eliminate cancer cells. These antibody constructs are not only intended for passive immune engagement but are also developed to function in combination with other immune cells through bispecific formats that engage CD3 on T cells. The dual or even tri-specific engagement strategy—exemplified by the tri-specific T cell engager targeting BCMA, CD3, and GPRC5D—represents one of the front-line innovations in preclinical design, providing a modular approach that could potentially reduce relapse and therapeutic resistance in multiple myeloma.

Another promising lead asset is the CAR-NK cell therapy candidate CYTO NK 301. Unlike CAR-T cells, the CAR-NK approach offers advantages such as a reduced risk of cytokine release syndrome and immune effector cell-associated neurotoxicity while showing high cytotoxic potency against cancer cells. CYTO NK 301 has been engineered to target a combination of tumor-associated antigens, including GPRC5D, ensuring a broadened attack on the malignant plasma cells and addressing issues related to antigen heterogeneity.

In addition, there is a growing focus on developing novel bispecific antibodies that feature one arm targeting GPRC5D and the other binding to CD3. These molecules are designed to bridge T cells and tumor cells, thereby triggering immune-mediated cytotoxicity directly within the tumor microenvironment. The platform leverages innovations in antibody engineering to improve parameters such as half-life, tissue penetration, and immunogenicity profile. Patent data suggest that these bispecific formats have been optimized through iterative improvements in binding domains and have been tested in preclinical models demonstrating significant anti-tumor activity.

Furthermore, an array of small molecule and peptide-based modalities are being evaluated in the preclinical realm to provide alternative ways of modulating the GPCR. Although traditional small molecules often face challenges with GPCR targets due to the lack of well-defined agonist sites, emerging technologies in structural biology and computational modeling are expected to yield candidates that can stabilize the receptor in a desired conformation or act as antagonists. Even if most of the early phase work is centered on antibody and cellular therapies, the cumulative knowledge from these approaches is eventually expected to inform the development of novel small molecule inhibitors or modulators targeting GPRC5D directly.

Research and Development Methodologies

Preclinical Testing Approaches

The preclinical work on GPRC5D assets employs a multitude of methodologies that together form a comprehensive testing strategy. At the cellular level, in vitro assays are designed to measure binding affinity and specificity, cytotoxic activity, and the efficient engagement of immune effector functions. For instance, ADCC assays are crucial in evaluating the ability of engineered antibodies to mediate immune cell-driven tumor cell lysis. In vitro cytotoxicity assays using multiple myeloma cell lines help to determine not only the potency but also the dose–response characteristics of these assets.

In vivo animal models remain an integral part of the preclinical evaluation process. Xenograft studies in immunocompromised mice are frequently used to assess the therapeutic efficacy, toxicity, and pharmacodynamics of CAR-T and CAR-NK therapies, as well as bispecific antibody constructs. These models provide critical data regarding tumor regression, survival outcomes, and potential adverse effects that need to be minimized before clinical translation. Moreover, grey-box in vivo studies that simulate the human immune system (humanized mouse models) allow for a more refined analysis of immune-oncology strategies involving GPRC5D targeting.

In addition to traditional biological assays, advanced computational methods are being leveraged to support preclinical asset discovery. Structure-based drug design (SBDD) and molecular dynamics simulations help in modeling the conformational states of GPRC5D and predicting the binding behavior of candidate therapeutics. Fragment-based drug discovery (FBDD) techniques, in which low molecular weight compounds are screened for binding efficacy, are also gaining traction due to their potential to guide the design of small molecule modulators. Such computational biology approaches have been integrated with high-throughput screening setups to identify and optimize lead candidates, refining both antibody and non-antibody-based therapies. This integration of in silico and in vitro methodologies facilitates a robust and iterative design process that accelerates progression through the preclinical pipeline.

The use of biophysical techniques—for example, surface plasmon resonance (SPR) and biolayer interferometry (BLI)—allows developers to monitor interactions between GPRC5D and the candidate molecules in real time. These methods are critical for determining binding kinetics and affinity, which ultimately influence the functional potency of these assets. Coupled with structural genomics initiatives aimed at resolving high-resolution structures of GPCR targets, these methodologies provide a multipronged lens to optimize preclinical assets before they move into clinical phases.

Challenges in Targeting GPRC5D

Despite the promising therapeutic rationale, there are several challenges inherent in targeting GPRC5D that are being addressed throughout the preclinical phase. One of the foremost difficulties lies in the receptor’s orphan status—without a defined endogenous ligand, it becomes complex to model receptor activation and signal transduction precisely. This uncertainty forces researchers to rely on surrogate binding studies and innovative screening methodologies to approximate the receptor’s physiological conformations.

Another significant hurdle is the inherent flexibility and dynamic conformations of GPCRs. The receptor’s propensity to adopt multiple active and inactive states complicates efforts to design molecules that can stably bind and modulate GPRC5D without leading to unintended signaling cascades. Moreover, while the restricted expression profile of GPRC5D reduces the possibility of off-target effects, even low-level expression in normal tissues such as hair cells presents the risk of on-target/off-tumor toxicity, especially for potent modalities like CAR-T or bispecific antibodies. Achieving the right balance between robust anti-tumor activity and minimal collateral damage remains a central challenge.

From the manufacturing perspective, the production of complex biologics, such as multispecific antibodies and engineered cell therapies, is fraught with technical hurdles. Maintaining reproducibility, ensuring batch-to-batch consistency, and scaling up production to meet clinical demands are significant obstacles that the field is actively addressing. Advances in protein engineering have led to the development of more consistent antibody constructs (e.g., humanized IgG4 formats and engineered bispecific formats) and improved cell culture techniques for CAR therapies; however, these processes require continued innovation and validation through rigorous preclinical testing.

Finally, regulatory and developmental uncertainties also pose challenges. Since GPRC5D-targeting therapies are relatively new, the lack of long-term clinical data makes it difficult to predict potential adverse events or resistance mechanisms that could emerge post-treatment. This uncertainty drives a need for more comprehensive preclinical safety data and carefully designed dose-escalation studies that will inform future clinical trials.

Potential Therapeutic Applications

Disease Areas of Interest

The primary disease area of interest for preclinical assets targeting GPRC5D is multiple myeloma—a malignancy characterized by the clonal proliferation of plasma cells. Multiple myeloma represents a significant clinical challenge because of the eventual emergence of resistance to current therapies, including BCMA-targeted treatments. GPRC5D is being actively investigated as an alternative target to deal with antigen escape phenomena. Preclinical data indicate that by engaging GPRC5D, therapeutic interventions may overcome some of the limitations associated with BCMA and other antigens.

Beyond multiple myeloma, research suggests that the expression of GPRC5D in certain subsets of hematologic malignancies opens the prospect for broader clinical applications. With ongoing studies evaluating the receptor’s expression pattern in various tumor types, there is potential for expanding GPRC5D-targeted therapies to other cancers that may exhibit aberrant GPRC5D expression. The selective expression also hints at the possibility of using these agents to treat cancers with a high tumor burden or in patients who have relapsed after standard treatments. These disease areas are of paramount interest to clinical investigators who are seeking to improve patient outcomes by introducing new modalities that offer more precise targeting with reduced systemic toxicity.

Future Prospects and Innovations

Looking ahead, the innovation landscape for GPRC5D-targeted preclinical assets is vibrant and multifaceted. The future promises further refinements in antibody engineering techniques, where next-generation bispecific and trispecific formats are expected to deliver improved efficacy, reduced immunogenicity, and adaptive pharmacokinetics tailored to patient needs. Innovations such as the tri-specific T cell engager—which not only targets GPRC5D but also incorporates BCMA and CD3—are at the forefront of this translational shift. Such assets exemplify how modular design can address multiple therapeutic challenges simultaneously, including antigen heterogeneity and reduced efficacy due to antigen escape.

In cellular therapies, the progress in CAR-based approaches is particularly promising. CAR-NK therapies, in contrast to CAR-T cells, offer the allure of a more manageable safety profile while still delivering potent anti-tumor activity. The development process for these therapies continues to benefit from innovations in gene editing, cell culture, and manufacturing technologies that aim to increase the yield, purity, and functional persistence of the modified cells. Future developments could include fully off-the-shelf cellular therapies, which have the potential to dramatically reduce time to treatment and manufacturing costs, further expanding treatment options for patients suffering from advanced or refractory cancers.

Another promising avenue is the integration of artificial intelligence (AI) and machine learning algorithms into the preclinical research workflow. These tools are being used to optimize virtual screening processes, refine structure-based drug design, and predict potential off-target effects or toxicities before assets reach the in vivo testing stage. By harnessing large datasets and multi-parameter analyses, AI-driven platforms may accelerate the identification of novel small molecule modulators or confirmatory peptides that can effectively inhibit GPRC5D with high selectivity. This computational approach, when combined with empirical validation, is likely to streamline the discovery process and optimize the lead candidates that emerge from initial preclinical screens.

Intellectual property filings and patents related to anti-GPRC5D antibodies and bispecific constructs are continuously being updated, suggesting an environment of dynamic innovation. Patents such as those describing anti-GPRC5D antibodies for bispecific T-cell engagement not only protect these novel molecules but also indicate the rapid diversification of technological approaches in this arena. Such patents underscore the competitive nature of the field and foreshadow a surge in clinical candidates that may enter the market in the coming years. These innovations are expected to be supported by strategic partnerships and collaborations among biotech companies, which facilitate the pooling of advanced technologies, funding, and clinical expertise.

Future prospective studies will also explore combination therapies where GPRC5D assets are used in tandem with other immunomodulatory agents or standard chemotherapy regimens. The rationale for these combination strategies is to build on the additive or even synergistic effects that may result from targeting multiple pathways simultaneously—a particularly important consideration in cancers like multiple myeloma, where heterogeneous tumor subpopulations can rapidly develop resistance against monotherapies. This combination approach is likely to be a major field of investigation in subsequent preclinical and early clinical trials and may eventually lead to personalized therapeutic regimens that are closely tailored to individual patient profiles.

Moreover, innovations in nanotechnology and drug delivery systems are expected to refine the pharmacokinetics and biodistribution of these assets. For instance, nanoparticle formulations or lipid-based carriers that deliver antibody fragments or small molecules directly to tumor sites can further mitigate systemic toxicity while maximizing local efficacy. Enhancements in these delivery technologies not only contribute to the overall safety profile but also enhance the therapeutic potency of the preclinical candidates. As these technology platforms mature, they will likely integrate with GPRC5D-targeted therapies to form a next-generation suite of cancer treatments.

Detailed and Explicit Conclusion

In summary, the preclinical assets being developed for GPRC5D represent a diverse and innovative portfolio aimed at addressing unmet clinical needs, particularly in the field of multiple myeloma and potentially in other malignancies where GPRC5D is aberrantly expressed. Starting from the fundamental understanding of GPRC5D as an orphan GPCR with a restricted expression profile, drug development strategies have targeted its biological unique attributes to forge therapeutic assets with enhanced tumor specificity and reduced toxicity. Key preclinical asset classes include engineered antibody-based therapies—such as anti-GPRC5D monoclonal antibodies and bispecific or tri-specific T-cell engagers—which are designed to harness and redirect immune cell activity toward malignant targets. Additionally, innovative cellular therapies like CAR-NK products, exemplified by CYTO NK 301, are emerging as potent alternatives to traditional CAR-T therapies, offering improved safety profiles and the capacity to target multiple antigens simultaneously.

The advancement of these assets has been supported by robust research and development methodologies. Preclinical testing encompasses a wide array of in vitro binding and cytotoxicity assays, in vivo xenograft studies, and state-of-the art biophysical and computational techniques that refine structural models and predict binding kinetics. This comprehensive approach helps in ensuring that candidate molecules not only demonstrate high potency and specificity but also maintain acceptable safety margins before advancing to early clinical stages. Despite these advances, the field faces notable challenges, including receptor conformational flexibility, difficulty in stabilizing transient active states, and potential off-target ramifications. Such challenges are being addressed through iterative antibody engineering, integration of novel computational modeling, and refinements in cellular assay systems.

The potential therapeutic applications of GPRC5D-targeted preclinical assets primarily underscore their utility in multiple myeloma, especially in the setting of antigen escape from existing therapies. However, the strategic versatility of these assets has also paved the way for future applications in other hematologic malignancies and possibly certain solid tumors where GPRC5D may serve as a biomarker or a functional target. Looking forward, innovations that blend multispecific antibody formats, next-generation CAR-based therapies, and AI-assisted discovery techniques are expected to further shape the pipeline, ensuring improved efficacy, reduced toxicity, and better patient outcomes.

Overall, the current landscape indicates a concerted and multidisciplinary effort that integrates molecular biology, immuno-engineering, computational science, and advanced screening methodologies. These converging technologies are poised to overcome the challenges of targeting a complex and dynamic receptor like GPRC5D. As preclinical candidates continue to mature through rigorous evaluation and optimization processes, they hold the promise of transforming therapeutic paradigms in oncology, particularly for patients with refractory or relapsed multiple myeloma. The robust preclinical data and diverse approaches documented in Synapse-funded research and patent filings illustrate a future where GPRC5D-targeted assets may combine with existing modalities, ultimately forming the backbone of next-generation anticancer therapeutics. Future innovations, fortified by strategic collaborations and technological breakthroughs, are expected to further extend the scope and impact of these therapies, driving forward the field of precision oncology towards a more effective and personalized treatment landscape.

In conclusion, the preclinical assets being developed for GPRC5D span a wide range of modalities—including antibody-based agents (monoclonal, bispecific, and tri-specific constructs), CAR-NK and other cellular therapies, and emerging small molecule or peptide inhibitors—all serving to exploit the unique tumor-selective expression of GPRC5D. These innovations, backed by state-of-the-art testing methodologies and computational support, are charting a path toward more effective and safe therapeutic options for multiple myeloma and possibly beyond. The convergence of these comprehensive research efforts is expected to lead to transformative clinical outcomes, ultimately providing new hope for patients with hard-to-treat cancers.

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.