What are the new molecules for CD112R antagonists?

11 March 2025
Introduction to CD112R
CD112R is a recently identified immune checkpoint receptor that has garnered considerable attention because of its unique role in regulating the activation of T cells and natural killer (NK) cells. Structurally, CD112R is a single-pass transmembrane protein containing a single extracellular immunoglobulin variable domain, a transmembrane segment, and a lengthy intracellular domain equipped with tyrosine residues—including motifs reminiscent of immunoreceptor tyrosine-based inhibitory motifs that mediate downstream inhibitory signaling. This structure positions CD112R to function as a key modulator within the intricate network of immune co-stimulation and co-inhibition. Its extracellular domain mediates high-affinity interactions with one of the poliovirus receptor family ligands, CD112, distinguishing it from other related molecules such as TIGIT and CD226 based on the differences in binding affinities observed via sensitive techniques like Biacore.

CD112R plays a central role in immune regulation by counterbalancing stimulatory signals on T cells. When CD112 binds to CD112R, it transmits inhibitory signals that help temper T cell activation—an important mechanism that safeguards against the damaging effects of autoimmunity while also, unfortunately, often being exploited by tumor cells to foster an immunosuppressive tumor microenvironment. Recently, studies have demonstrated that the expression of CD112R is dynamically regulated, being upregulated upon T cell activation as well as in various NK cell subsets, further underpinning its importance in orchestrating immune responses in both inflammatory and oncologic settings. This balance between co-stimulatory and coinhibitory signals is essential; CD112R’s interaction with its ligand serves as an “immune brake” that can be manipulated to achieve therapeutic benefits, especially in the context of cancer immunotherapy.

CD112R Structure and Function
At the molecular level, CD112R is defined by its single extracellular IgV domain that is critical for ligand recognition. The difference in binding preferences between CD112R and other PVR family members—such as TIGIT or CD226—stems from subtle changes in the amino acid sequence and the spatial presentation of the receptor’s surface. The intracellular tail of CD112R harbors tyrosine residues that become phosphorylated upon ligand engagement, triggering recruitment of inhibitory adapter proteins that dampen downstream signaling cascades such as those involving the NFAT pathway. This modulation of cellular activation is crucial for maintaining immunological tolerance and preventing excessive tissue damage during immune responses.

Functionally, CD112R is involved in “fine-tuning” the immune response. By engaging CD112 with high affinity, CD112R outcompetes the low-affinity interactions that other receptors might have with CD112, thereby shifting the overall cellular response toward inhibition. This mechanism has been observed not only in T cells but also in NK cells, where it shapes the cytotoxic potential against stressed or transformed cells. In the tumor microenvironment, high CD112 expression on tumor cells can engage CD112R on infiltrating lymphocytes, leading to reduced production of key cytokines such as interferon-γ and ultimately contributing to an immunosuppressive milieu that protects the tumor from immune attack.

Role of CD112R in Immune Regulation
The inhibitory effects mediated by CD112R are particularly significant in the context of cancer. Tumors often exploit checkpoint pathways by upregulating ligands like CD112 to bind inhibitory receptors, thereby reducing T cell effector functions and impairing anti-tumor immunity. Experimental studies have shown that blockade of the CD112R–CD112 interaction results in enhanced T cell activity and increased production of pro-inflammatory cytokines, leading to tumor regression in various preclinical models. Moreover, the co-expression of CD112R with other checkpoint receptors, such as TIGIT and PD-1, on tumor-infiltrating lymphocytes in several solid tumors, reinforces the notion that CD112R is an important contributor to immune exhaustion in the tumor microenvironment.

The multifaceted role of CD112R in immune regulation extends beyond direct suppression of T cell activation. It appears to be involved in the broader orchestration of immune cell interactions by modulating dendritic cell functions and influencing the recruitment or retention of lymphocytes within the tumor. This regulatory capacity makes CD112R a highly attractive target for immunotherapeutic intervention, as its blockade may release multiple inhibitory constraints on the immune system simultaneously.

Development of CD112R Antagonists
Recent years have witnessed rapid progress in the development of therapeutic agents aimed at modulating checkpoint pathways. Specifically, CD112R has emerged as a novel checkpoint receptor with several research groups and pharmaceutical companies pursuing antagonists that can block its interactions with CD112. The rationale behind developing CD112R antagonists is to disrupt this inhibitory signaling, thereby enhancing the activation and expansion of effector T cells and NK cells capable of mediating tumor cell killing.

Current Status of CD112R Antagonists
At present, the therapeutic landscape in the CD112R field is defined largely by monoclonal antibodies designed to interfere with the receptor–ligand binding. One of the earliest and most promising molecules in this category is COM701, a humanized, hinge-stabilized IgG4 antibody developed to bind selectively to human CD112R. Clinical investigations involving COM701 have demonstrated that its administration can effectively block the CD112–CD112R interaction, thereby enhancing T cell effector functions and potentiating the anti-tumor effects of other immunotherapeutic agents when used in combination with anti-PD-1 and anti-TIGIT antibodies.

In parallel, Junshi Biosciences has developed another anti-CD112R monoclonal antibody, TAB009/JS009, which has received Investigational New Drug approval from the U.S. FDA for the treatment of advanced solid tumors. This recombinant humanized IgG4 antibody similarly functions by binding to CD112R with high affinity, thus preventing its interaction with CD112 and facilitating an enhanced immune response against tumors. Early-phase clinical trials and preclinical studies for these molecules have reported promising signs of both safety and preliminary efficacy. For instance, COM701 has been shown not only to increase cytokine production by T cells but also to reverse immune suppression in the tumor microenvironment, providing a mechanistic rationale for its use as a monotherapy or in combination with other checkpoint inhibitors.

Patent literature also supports the advancement of CD112R antagonists in the therapeutic pipeline. One notable patent describes anti-CD112R antibody compositions that block the binding interaction between human CD112 and CD112R without cross-reacting with the mouse counterpart, addressing an important consideration for translational preclinical studies. Such patents outline the structural and functional attributes that are essential for effective CD112R antagonism and offer insights into optimizing these molecules for enhanced pharmacological profiles.

Novel Molecules and Their Mechanisms
The strides made in the development of CD112R antagonists have not been limited to reiteration of known mAb formats but have also led to the design of novel molecular entities with unique mechanisms of action.

One of the foremost new molecules for CD112R antagonism is COM701. The design of COM701 leverages detailed structural insights into the CD112R receptor, which facilitated extensive optimization of the antibody’s binding affinity and specificity. COM701 specifically blocks the CD112R–CD112 interaction, thereby reactivating T cell activity and fostering a more robust immune response against tumor cells.

Another significant candidate in this arena is TAB009/JS009, developed by Junshi Biosciences. TAB009/JS009 has been engineered as a recombinant humanized IgG4 antibody that binds to CD112R with high affinity, thereby effectively preventing its engagement with CD112 on tumor and immune cells. The high-affinity binding kinetics of TAB009/JS009 allow for a comprehensive blockade of CD112R-mediated inhibitory signals. This design also includes considerations to minimize immune-related adverse events, such as unwanted cross-reactivity or the triggering of Fc-mediated effector functions that could complicate its clinical profile.

Beyond these two leading molecules, the broader pipeline includes additional antibody candidates that are currently under development or in earlier stages of research. Some of these advanced agents are engineered with modifications in their Fc regions to enhance stability and reduce clearance, thereby improving their pharmacokinetic properties. Additionally, bispecific formats that simultaneously target CD112R and other inhibitory receptors appear promising. Such molecules are being designed to achieve synergistic blockade of multiple checkpoints simultaneously—for example, targeting both CD112R and TIGIT—but their specific clinical candidates are in the early preclinical stage.

Moreover, patents related to anti-CD112R compositions highlight strategies involving different antibody isotypes and formats, such as fully human versus humanized antibodies. The modifications detailed in these patents emphasize the importance of fine-tuning the interactions at the molecular level so that the resulting antagonists maintain effective blockade while avoiding undesired cross-reactivity with other receptors or species homologs. This is especially critical since the degree of sequence conservation between human and mouse CD112R can affect the interpretation of preclinical models.

While the above examples primarily refer to antibody-based modalities, there is also increasing interest in other molecular formats, such as engineered variants derived from recombinant proteins or even aptamer-based molecules. Indeed, the approach of developing DNA aptamers that agonize or antagonize receptor function has been demonstrated in other immune checkpoint systems. Although such aptamers are not yet prominently featured in the CD112R space, the success in analogous systems provides reason to believe that similar strategies may be adapted for CD112R antagonism in the future.

At a mechanistic level, these novel molecules function by sterically hindering the interaction between CD112R and its ligand CD112. By occupying the ligand-binding region of CD112R, these antibodies prevent CD112 from engaging with the receptor, thereby mitigating the receptor’s capacity to transmit inhibitory signals upon ligand binding. This blockade assists in restoring the balance in favor of immune activation. It promotes T cell proliferation, cytokine secretion, and the reengagement of cytotoxic functions essential for effective immunosurveillance and tumor cell killing.

The detailed biochemical characterization of these novel antagonists, including high-resolution binding studies and surface plasmon resonance analyses, reveals that the critical determinants for their activity are precise molecular interactions within the CD112R binding pocket. Such studies demonstrate that the novel molecules have markedly higher binding affinities compared to the native interactions between CD112R and CD112, ensuring that the receptor is effectively sequestered in an inactive state even in the presence of elevated ligand concentrations in the tumor microenvironment.

Research and Development Methodologies
The discovery and development of novel molecules for CD112R antagonism have benefitted greatly from advances in drug discovery and screening techniques. A multifaceted approach combining structural biology, high-throughput screening, computational modeling, and preclinical pharmacology has accelerated progress in this domain.

Drug Discovery and Screening Techniques
Initial identification of CD112R function and its interaction with CD112 relied heavily on genomic searches and surface protein profiling, using methods such as high-throughput RNA sequencing and flow cytometry to determine expression patterns on T cells and NK cells. Once CD112R was established as a viable target, biophysical techniques such as SPR and Biacore experiments were employed to characterize the binding affinities between CD112R and its ligands, revealing the striking differences in kinetics that underpin its inhibitory function.

With the target validation in hand, antibody discovery efforts typically involved techniques such as phage display, hybridoma generation, and more recently, next-generation sequencing-assisted screening of antibody libraries. These approaches have been instrumental in isolating mAbs like COM701 and TAB009/JS009, which exhibit the necessary specificity and potency to serve as clinical candidates. Computational methods, including structure-based design and molecular docking, have further refined the lead candidates by guiding the optimization of binding interfaces, thereby enhancing the antagonistic activity while reducing potential off-target effects.

In addition, fragment-based drug discovery has increasingly been integrated into modern antibody engineering pipelines. Using fragment-based drug discovery, chemists and biologists are able to dissect the structural details of the receptor–ligand interface into smaller, manageable binding hotspots. These insights confront the design of molecules that specifically target these key regions on CD112R, emulating or even surpassing the natural inhibitory conformations that the receptor adopts upon ligand binding.

Preclinical and Clinical Development Strategies
Once promising candidates such as COM701 and TAB009/JS009 were identified through rigorous screening, their preclinical evaluation involved a series of in vitro and in vivo assays. In vitro studies have shown that blockade of CD112R by these antagonists significantly enhances T cell activation markers and cytokine production. For instance, human T cells co-cultured with tumor cell lines exhibit increased interferon-γ production upon treatment with these antibodies, correlating well with the proposed mechanism of action.

Animal models have provided further evidence of the therapeutic potential of these molecules. Murine models genetically engineered to express human CD112R or bearing human tumor xenografts have been used to evaluate the efficacy and toxicity profiles of the new antagonists. These studies confirmed that treatment with COM701 or TAB009/JS009 can delay tumor growth and reprogram the tumor microenvironment, providing proof-of-concept for their use in subsequent clinical trials.

On the clinical development front, early-phase trials are designed to assess safety, tolerability, pharmacokinetics, and preliminary efficacy. COM701, for example, has entered phase I trials where dose-escalation studies aim to determine the maximum tolerated dose while monitoring biomarker changes in T cell activity and cytokine levels. Similarly, TAB009/JS009 has progressed to an IND-approved stage, with phase I/II clinical trial designs that include both monotherapy arms and combination regimens with established checkpoint inhibitors such as anti-PD-1 agents.

Regulatory documentation and patent filings, further validate that these approaches are built upon early knowledge from preclinical screening methods and robust in vitro signaling assays. By coupling these experimental findings with advanced computational models, developers have been able to refine candidate molecules more quickly and with higher precision, ultimately shortening the developmental timeline from target identification to clinical evaluation.

Implications and Future Directions
The therapeutic potential of CD112R antagonists extends well beyond their immediate impact on T cell activation. Their development symbolizes a broader shift in oncology from treatments that directly target tumor cells to those that modulate the patient’s immune system. The novel molecules designed for CD112R antagonism are thus envisioned to be part of next-generation combination therapies intended to synergize with established checkpoint inhibitors, such as PD-1/PD-L1 and TIGIT blockades.

Potential Therapeutic Applications
The primary therapeutic application for novel CD112R antagonists lies in the treatment of advanced solid tumors where the tumor microenvironment is characterized by high levels of immunosuppression. By blocking the CD112R–CD112 interaction, molecules like COM701 and TAB009/JS009 can potentially reverse immune evasion, leading to enhanced T cell function and more effective tumor cell killing. Early-stage clinical data suggest that patients with tumors expressing high levels of CD112 or those whose tumor-infiltrating lymphocytes co-express CD112R with other inhibitory receptors, such as TIGIT or PD-1, may derive particular benefit from such treatments.

Furthermore, beyond oncology, the modulation of CD112R signaling might hold promise in diseases where immune suppression or dysregulation plays a critical role. For instance, in chronic infections or in settings where enhanced immune activation is desired, the blockade of CD112R could potentiate the cellular response without causing overt autoimmunity. This notion is supported by mechanistic studies that show how the interruption of CD112R signaling relieves inhibitory constraints on effector cells.

Challenges and Opportunities in CD112R Antagonist Development
Even as novel CD112R antagonists show promise, several challenges remain. One key issue is ensuring that the blockade of CD112R does not lead to uncontrolled immune responses that might precipitate autoimmunity or other inflammatory sequelae. The balance between efficacy and safety is delicate, especially in combinational regimens where multiple checkpoints are simultaneously targeted. In the context of antibodies like COM701 and TAB009/JS009, ongoing clinical studies are meticulously monitoring for immune-related adverse events and other toxicities.

Another challenge is linked to the pharmacokinetic and pharmacodynamic properties of these large molecules. Optimizing their half-life, bio-distribution, and tissue penetration requires iterative engineering of the Fc regions or altering the antibody format. Some current development strategies, as indicated by recently published patents, involve modifications in antibody structure to achieve more favorable pharmacokinetic profiles without compromising their antagonistic activity.

Opportunities in this field are considerable. Continued advances in high-throughput screening technologies, computational modeling, and antibody engineering are poised to further enhance the design of CD112R antagonists. For example, multiplexed screening techniques coupled with deep learning algorithms could yield entirely new classes of antagonists by predicting optimal binding conformations and guiding mutational modifications for improved selectivity and potency. Additionally, the rising trend in developing bispecific or multispecific antibodies may allow for simultaneous targeting of CD112R alongside other immune checkpoints, potentially leading to synergistic therapeutic outcomes that overcome resistance mechanisms in refractory tumors.

Moreover, the emerging field of aptamer-based therapeutics, which has already shown promise for other checkpoint receptors like CD200R1, suggests that alternative molecular formats could also be explored for CD112R. While monoclonal antibodies currently dominate the space, advances in aptamer technology could eventually give rise to smaller, more easily manufactured agents that have comparable efficacy with potentially fewer side effects.

The future direction of CD112R antagonist development is also likely to be guided by a deeper understanding of the receptor’s biology. Future studies may uncover additional intracellular partners and signaling cascades associated with CD112R, highlighting new intervention points that could be exploited by small molecules or novel biologics. The use of CRISPR-based genetic screens and proteomics to map the signaling network downstream of CD112R will provide invaluable insights into how best to manipulate this pathway for therapeutic gain.

Conclusion
In summary, the new molecules for CD112R antagonists represent a significant advancement in the field of cancer immunotherapy by targeting a previously underexplored immune checkpoint. Novel agents such as COM701 and TAB009/JS009 have been developed using state-of-the-art screening and engineering techniques, and they function by effectively blocking the high-affinity interaction between CD112R and its ligand CD112. These antagonists relieve the inhibitory signals limiting T cell and NK cell activity, thereby enhancing anti-tumor immunity.

The development process has been supported by a robust body of preclinical work—including high-throughput screening, detailed biophysical characterization, and advanced in vivo models—as well as early-phase clinical trials that have demonstrated promising safety and activity profiles. Additional innovation is seen in the refinement of antibody formats and the exploration of alternative molecular modalities, such as engineered aptamers, that may offer complementary advantages in terms of stability, cost, and ease of manufacturing.

Looking forward, the integration of CD112R antagonists into combination treatment regimens, particularly with other checkpoint inhibitors like PD-1 and TIGIT blockers, holds enormous promise for treating advanced solid tumors and potentially other diseases marked by immune suppression. The challenges related to optimal dosing, balancing efficacy and safety, and favorable pharmacokinetic profiles are being actively addressed through both empirical and computational approaches. The continuous refinement of these molecules, together with a deeper understanding of CD112R’s role in immune modulation, will ensure that their future applications are both broad and impactful.

Overall, the discovery of these new molecules not only broadens the arsenal of immunotherapeutic strategies available to clinicians but also provides an impetus for further research into novel immune checkpoints. The journey from target validation to clinical approval for CD112R antagonists is emblematic of the rapid progress being made in modern drug discovery, indicating a bright future for the development of more effective and safe cancer treatments.

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.