What are the new molecules for CD39 inhibitors?

11 March 2025
Introduction to CD39 and Its Role
CD39, also known as ecto-nucleoside triphosphate diphosphohydrolase‐1 (ENTPD1), is a membrane-bound enzyme that plays a central role in the metabolism of extracellular nucleotides. It catalyzes the hydrolysis of adenosine triphosphate (ATP) and adenosine diphosphate (ADP) into adenosine monophosphate (AMP), which is subsequently converted into adenosine by CD73. This enzymatic cascade is crucial in modulating the concentration of immunomodulatory mediators in the extracellular space, thereby regulating inflammatory and immune responses. Research from the synapse source has consistently highlighted the importance of CD39 in shaping the immune microenvironment, particularly in diseases where the balance between ATP (a pro-inflammatory danger signal) and adenosine (a key immunosuppressive molecule) is disturbed.

Biological Function of CD39
At the molecular level, CD39 is expressed on a broad range of cells, including immune cells (such as T regulatory cells, dendritic cells, and natural killer cells), endothelial cells, and even tumor cells. CD39’s primary function is to act as the rate-limiting enzyme in the extracellular degradation of ATP, a process that has significant biological implications. ATP is released from cells upon activation, stress, injury, or cell death and serves as a potent inflammatory signal by engaging purinergic receptors, such as P2X and P2Y receptors. The conversion by CD39 diminishes the levels of ATP and favors the production of AMP, which is later converted by CD73 into adenosine. Adenosine, in turn, binds to adenosine receptors (primarily A2A and A2B) on various immune cells and contributes to the suppression of inflammatory responses and maintenance of immune homeostasis.

In addition to its enzymatic activity, CD39 has been implicated in cell adhesion and the modulation of thrombotic responses, evidenced by its role in inhibiting ADP-dependent platelet aggregation. Its broad tissue expression and its coupling to key immunomodulatory and prothrombotic pathways underline its importance in both physiological and pathological processes.

CD39 in Disease Pathogenesis
CD39 plays a dual role in many diseases. On one hand, its enzymatic activity is essential for dampening excessive inflammation, but on the other hand, the elevated expression of CD39 in the tumor microenvironment has been associated with the development of immune tolerance. In many solid tumors, high levels of CD39 expression are observed on tumor-infiltrating regulatory T cells (Tregs) as well as on the tumor cells themselves. This high level of CD39-mediated ATP hydrolysis contributes to the accumulation of adenosine, creating a highly immunosuppressive microenvironment that supports tumor immune escape and progression. Moreover, in the context of chronic infections, autoimmune diseases, and inflammatory conditions, CD39-mediated modulation of nucleotide levels significantly influences disease progression. The dysregulation of the ATP-adenosine balance can exacerbate tissue fibrosis, autoimmune responses, and chronic inflammatory states.

Development of CD39 Inhibitors
The therapeutic potential of targeting CD39 resides in its ability to modulate the extracellular purine environment. By inhibiting CD39, it is possible to preserve higher levels of ATP which acts as an immune-stimulatory signal, while limiting the production of immunosuppressive adenosine. This has driven significant research into the development of both small-molecule inhibitors and biologics that target CD39 with the aim of restoring or enhancing antitumor immunity and modulating other pathological processes.

Current CD39 Inhibitors
Historically, several inhibitors of CD39 have been identified and used primarily in preclinical studies. Compounds such as ARL67156 and POM-1 are among the earliest described CD39 inhibitors. ARL67156 is a nucleotide analog that competitively inhibits CD39’s enzymatic activity, though its inhibitory potency is modest and its selectivity is limited given that it can also intersect with the activities of related enzymes. POM-1, on the other hand, is a polyoxometalate that blocks CD39 activity in vitro. However, studies have shown that POM-1 lacks selectivity, as it also inhibits several other kinases and ATPases, which poses a challenge for its potential clinical application.

In addition to these small-molecule inhibitors, therapeutic efforts have led to the development of monoclonal antibodies (mAbs) that target CD39. One prominent example is IPH5201, a human monoclonal antibody being evaluated in clinical research for its ability to block the hydrolysis of extracellular ATP by CD39, thereby alleviating tumor-mediated immunosuppression. While these current inhibitors have provided proof-of-concept for the therapeutic targeting of CD39, their non-selective mechanisms and suboptimal pharmacokinetic properties have spurred further research into new molecular entities.

Recent Advances in Molecule Development
Efforts to develop new molecules for CD39 inhibition have diversified in recent years. Researchers have focused not only on improving selectivity and potency but also on discovering novel chemical scaffolds that possess favorable drug-like properties. Some of the major recent advances include:

1. Triazinoindole-Based Compounds:
Recent studies have used homology modeling combined with virtual screening and in vitro enzymatic assays to identify novel scaffolds for CD39 inhibition. In one study, researchers reported the identification of triazinoindole-based compounds that could effectively prevent CD39’s enzymatic activity. One compound was reported to inhibit CD39 with an IC50 value of 27.42 ± 5.52 μM, while an analogue of this molecule exhibited an IC50 of 79.24 ± 12.21 μM. These compounds not only demonstrated potent inhibition of CD39 in enzymatic assays but also significantly reduced adenosine monophosphate (AMP) production in colorectal cancer cell lines such as HT29 and MC38. Molecular docking studies supported the binding specificity, indicating that residues like R85 in CD39 played a crucial role in interacting with these inhibitors. This virtual screening approach and subsequent hit-to-lead identification represent a promising shift toward molecule development for selective CD39 inhibition.

2. Repurposing of Approved Drugs – Ceritinib:
Another exciting avenue in the development of CD39 inhibitors comes from repurposing strategies. The tyrosine kinase inhibitor ceritinib—originally approved for the treatment of anaplastic lymphoma kinase (ALK)-positive non-small cell lung cancer—has been recently discovered to strongly inhibit CD39 activity. Ceritinib was found to exert a non-competitive, allosteric inhibition on CD39, displaying low micromolar potency. Its mechanism does not rely on competition with the natural substrate ATP, which provides an advantage in terms of kinetic properties and potential efficacy. Ceritinib’s identification as a CD39 inhibitor was further validated in immune and cancer cell assays, showing that it could increase extracellular ATP and block adenosine generation. This repurposing of an already approved drug represents a strategic shortcut toward clinical translation while also opening the door for dual-target inhibitors that might combine ALK and CD39 inhibition.

3. Ticlopidine Derivatives and Benzotetrahydropyridines:
Another novel class of compounds under investigation involves derivatives of ticlopidine. Ticlopidine, an antithrombotic agent, in its intact, non-metabolized form has been reported to inhibit CD39. Researchers have synthesized extensive libraries of ticlopidine derivatives and analogs, carefully examining the structure-activity relationships (SAR). Among these, benzotetrahydropyridine derivatives—where the labile thiophene motif is replaced by a benzene ring—have emerged as a new class of allosteric CD39 inhibitors. These molecules offer improved metabolic stability and reduced off-target effects as compared with the parent compound. This approach is particularly exciting because it offers a rational design strategy to modulate CD39 enzymatic activity through allosteric mechanisms, potentially achieving higher selectivity.

4. Nucleotide Analogs Based on ARL67156:
While ARL67156 itself has been widely used as a lead compound, researchers have further optimized its chemical structure to generate analogs with enhanced potency and dual inhibitory effects on both CD39 and CD73. These nucleotide analogs have undergone extensive medicinal chemistry efforts involving modifications at the N6- and C8-positions of the adenine core, as well as alterations to the triphosphate chain. Some derivatives have demonstrated Ki values in the low micromolar range and act as dual-target inhibitors that also inhibit CD73 activity. Although these compounds are still in the early stages of development, they serve as valuable tools for elucidating the mechanistic roles of ectonucleotidases in the adenosinergic pathway and may evolve into clinically useful agents.

5. Allosteric Inhibitors with Improved Selectivity:
Novel strategies also include designing allosteric inhibitors that target sites on CD39 distinct from the substrate-binding pocket. By focusing on these alternative binding sites, researchers hope to minimize competitive interactions and reduce cross-reactivity with other ATP-binding enzymes. The discovery of ticlopidine derivatives acting as allosteric inhibitors is a prime example of this strategy. The allosteric binding offers the possibility of modulating enzyme activity without completely abolishing substrate processing, which might provide a more balanced restoration of immune function.

6. Antibody-Based and Nanobody Approaches:
Although the question focuses on small molecule inhibitors, it is important to note that antibody-based approaches have also advanced. In addition to the already mentioned IPH5201, which is currently under clinical investigation, other research groups have developed nanobodies or single-domain antibodies targeting CD39. These molecules, due to their small size and unique binding capabilities, may offer improved tissue penetration and specific blocking of CD39-mediated ATP hydrolysis. Such biologics not only block the enzymatic function of CD39 but may also induce receptor internalization, serving as a complementary strategy to small molecules.

7. Dual-Targeted Inhibitors and Combination Strategies:
In the evolving landscape of CD39 inhibition, there is growing interest in compounds that can simultaneously target multiple components of the adenosine pathway. For example, dual inhibitors that block both CD39 and CD73 could offer superior antitumor effects by simultaneously preserving pro-inflammatory extracellular ATP and diminishing immunosuppressive adenosine production. The development of such dual inhibitors is in its early exploratory phase but represents a significant trend in the rational design of novel immunomodulatory agents.

Therapeutic Potential and Applications
The new molecules for CD39 inhibition exhibit considerable promise both as standalone therapies and in combination with other treatment modalities. The modulation of extracellular ATP and adenosine levels is crucial not only in cancer therapy but also in other immune-mediated conditions.

Applications in Cancer Therapy
In the realm of oncology, the inhibition of CD39 is emerging as an attractive strategy because tumors often overexpress CD39 either on the tumor cells directly or on infiltrating regulatory immune cells. This overexpression contributes to the creation of an adenosine-rich microenvironment that significantly attenuates antitumor T-cell responses. The new generation molecules—such as the triazinoindole-based compounds, ceritinib, and ticlopidine derivatives—have shown strong preclinical evidence of increasing extracellular ATP levels while reducing adenosine production. This shift is associated with enhanced dendritic cell maturation, restored cytotoxic T lymphocyte and natural killer cell activity, and improved responses to immune checkpoint inhibitors.

For instance, by repurposing ceritinib as a CD39 inhibitor, researchers have demonstrated that its administration translates into a significant blockade of CD39 enzymatic activity in vitro and in cell-based models, thus offering a tangible method to overcome the immunosuppressive barrier in the tumor microenvironment. Additionally, the allosteric ticlopidine derivatives provide an alternative route to inhibit CD39 without triggering complete substrate competition, potentially reducing adverse off-target effects and permitting a more refined modulation of immune responses.

These novel agents are being considered for combination therapies that integrate CD39 inhibition with standard chemotherapy, immune checkpoint blockade (such as PD-1/PD-L1 inhibitors), and radiation therapy. When combined, these therapeutic strategies may convert “cold” tumors—those lacking significant immune infiltration—into “hot” tumors that are responsive to immunotherapy, offering renewed hope in the treatment of resistant cancer types.

Applications in Autoimmune Diseases
Beyond cancer therapy, CD39 inhibitors hold potential in the treatment of autoimmune disorders. In diseases where immune activation is dysregulated, the enhanced clearance of ATP may contribute to chronic inflammation and tissue damage. By inhibiting CD39, the retention of extracellular ATP could potentiate controlled immune activation while simultaneously counteracting the adverse effects of excessive adenosine production. While most of the current clinical emphasis has been on immuno-oncology, there is growing interest in exploring these novel CD39 inhibitors in autoimmune contexts such as multiple sclerosis, rheumatoid arthritis, and inflammatory bowel disease, where the regulation of immune cell activation is critical.

In autoimmune conditions, the restoration of ATP levels might help recalibrate aberrant inflammatory pathways that frequently lead to tissue damage, while selective CD39 blockage may assist in preserving certain beneficial aspects of immune tolerance. As these molecules enter more advanced stages of preclinical development, researchers are carefully examining their effects on diverse immune cell populations, cytokine production profiles, and overall tissue homeostasis.

Challenges and Future Directions
Despite the significant progress in discovering and optimizing new molecules for CD39 inhibition, several challenges remain. Balancing potency, selectivity, pharmacokinetics, and safety is a complex task that involves iterative cycles of medicinal chemistry, in vitro and in vivo testing, and translational research.

Current Challenges in Development
One of the principal challenges in the development of CD39 inhibitors is the issue of selectivity. Early inhibitors like POM-1, while effective at inhibiting CD39 activity, also interact with other ATP-dependent enzymes (e.g., kinases and ATPases), leading to unintended side effects. This off-target activity complicates the interpretation of biological effects and may limit clinical application. Therefore, a major focus of current research is on designing molecules that are highly selective for CD39. For example, the development of allosteric inhibitors such as some ticlopidine derivatives offers an advantage by targeting alternative binding sites distinct from the highly conserved ATP binding pocket, thereby reducing the risk of cross-reactivity.

Another challenge is the inherent complexity of the tumor microenvironment and the multifaceted role of CD39 in both immune suppression and thrombotic regulation. Inhibitors must be carefully balanced to enhance antitumor immunity without inadvertently increasing the risk of thrombosis or exacerbating inflammatory conditions elsewhere in the body. The diversity of cell types that express CD39—from tumor and stromal cells to various immune cells—adds further complexity and necessitates thorough preclinical evaluation to understand differential responses.

Pharmacokinetic properties and bioavailability of these new molecules also need improvement. Many early lead compounds present challenges such as poor solubility or rapid metabolism, which can limit their in vivo efficacy. The repurposing of approved drugs like ceritinib demonstrates one approach to bypass some of these hurdles, but additional medicinal chemistry efforts are required for molecules like triazinoindole-based compounds and novel ticlopidine derivatives to optimize their drug-like properties.

Finally, translating preclinical successes to clinical outcomes is often a major hurdle. Animal models may not fully recapitulate human tumor immunology or autoimmune pathogenesis, making it crucial to validate these inhibitors in more predictive model systems. The continued integration of pharmacodynamic biomarkers and advanced imaging techniques in clinical studies will be essential for monitoring the real-time activity of these inhibitors and fine-tuning dosages to achieve the desired immunomodulatory outcomes.

Future Research Directions
Future directions in the field of CD39 inhibitor development are multifaceted. One promising avenue is the refinement of computational approaches combined with high-throughput screening (HTS) techniques to discover novel chemotypes. Advances in homology modeling, molecular dynamics simulations, and virtual screening not only allow for the identification of novel binding pockets on CD39 but also enable the systematic optimization of molecular interactions. For instance, further structure-based optimization of the triazinoindole scaffold could yield compounds with nanomolar potency and excellent selectivity profiles.

Simultaneously, novel methods in chemical synthesis such as de novo design of small molecule inhibitors and iterative refinement using artificial intelligence and machine learning are expected to accelerate the discovery of CD39 inhibitors. These cutting-edge techniques leverage large chemical libraries and structure–activity relationship (SAR) data to identify promising candidates with enhanced specificity.

Another future direction is the exploration of combination therapies that target multiple nodes of the adenosinergic pathway. Dual inhibition strategies—targeting both CD39 and CD73 concurrently—are currently under investigation and represent an appealing approach to achieve a synergistic blockade of adenosine-mediated immunosuppression. Such dual inhibitors or concomitant use of two selective inhibitors may significantly amplify therapeutic responses, particularly in refractory cancers and complex autoimmune disorders.

Furthermore, antibody engineering and nanobody technology represent additional avenues for future research. Advances in bispecific antibodies and chimeric antigen receptor (CAR) T cells engineered to secrete anti-CD39 nanobodies have shown promising preclinical activity. These biologics can be designed to combine tumor-targeting specificity with the blockade of CD39-mediated adenosine production, offering an integrated approach to overcome immune resistance mechanisms in the tumor microenvironment.

It is equally important to further elucidate the molecular mechanisms underlying CD39 regulation in various cellular contexts. Ongoing research should focus on identifying key regulatory elements controlling CD39 expression and activity, potentially revealing new targets for indirect inhibition. Such studies can inform the development of combination treatment strategies that modulate upstream signaling pathways in conjunction with direct CD39 inhibition, ultimately leading to more effective and durable therapeutic outcomes.

In autoimmune disease applications, future research should investigate the optimal modulation of CD39—to strike a balance between immune activation and suppression. Given that both excessive and insufficient CD39 activity can be detrimental, fine-tuning its inhibition in diseases like rheumatoid arthritis or inflammatory bowel disease will require precise dosing and perhaps localized delivery systems to achieve therapeutic benefits while minimizing systemic side effects.

Finally, with the increasing trend for personalized medicine, it is likely that future clinical studies will focus on patient stratification based on biomarkers such as CD39 expression levels, tumor immune infiltration patterns, and genetic predispositions. This approach will facilitate the identification of patients most likely to benefit from CD39 inhibitors, thereby enhancing the clinical impact of these novel therapeutic agents.

Conclusion
In summary, the landscape of CD39 inhibition has evolved significantly over the past few years. Initially, inhibitors such as ARL67156 and POM-1 served as important proof-of-concept tools to demonstrate that targeting CD39 can modulate the extracellular ATP/adenosine balance. However, due to issues with selectivity and pharmacokinetic limitations, new molecules have emerged that promise improved therapeutic profiles. These include:

• Triazinoindole-based compounds – identified using a combination of homology modeling and high-throughput virtual screening, showing promising IC50 values and binding specificity with key residues such as R85.
• Repurposed molecules like ceritinib – originally a potent tyrosine kinase inhibitor, it has been repurposed to act as a non-competitive, allosteric CD39 inhibitor with low micromolar potency, demonstrating the feasibility of dual-target strategies.
• Ticlopidine derivatives and benzotetrahydropyridines – representing a new class of allosteric inhibitors designed through extensive structure-activity relationship studies, these molecules are optimized for selectivity and metabolic stability.
• Optimized nucleotide analogs derived from ARL67156 – these compounds are being refined to serve as dual inhibitors of CD39 and CD73, although selectivity remains an ongoing challenge.
• Advanced antibody-based modalities and nanobodies – while not small molecules per se, these biologics are attaining importance as they can block CD39 on multiple cell types and may serve as complementary or alternative approaches to small molecule inhibitors.

This systematic evolution in molecule development reflects a general-to-specific-to-general pattern where the initial broad inhibition strategy has been refined through medicinal chemistry, virtual screening, and repurposing approaches, and is now being integrated into a broader therapeutic framework. New molecules are being evaluated from various perspectives: on the basis of enzyme kinetics and structure, through computational and metabolic engineering for enhanced specificity, in cell-based assays showing favorable biological outcomes, and in preclinical models demonstrating synergistic effects with other therapies.

From a clinical standpoint, these novel CD39 inhibitors are poised to not only enhance antitumor immunity in resistant cancers by preserving pro-inflammatory ATP levels but also potentially rebalance immune responses in autoimmune diseases where dysregulated purinergic signaling contributes to pathology. Nonetheless, challenges remain—primarily regarding selectivity, pharmacokinetic properties, and translating preclinical efficacy into meaningful clinical outcomes.

In conclusion, the development of new molecules for CD39 inhibition is a rapidly advancing field that spans innovative chemical scaffolds like triazinoindole compounds, repurposed kinase inhibitors such as ceritinib, and the rational design of allosteric inhibitors including ticlopidine derivatives. These agents hold promise not only as monotherapies but also as part of combination strategies that target the full adenosinergic cascade. Future research focused on optimizing selectivity, elucidating molecular mechanisms, and integrating advanced computational tools will be essential to fully harness the therapeutic potential of these novel CD39 inhibitors. As our understanding of CD39’s role in the tumor microenvironment and immune regulation deepens, so too will our ability to design targeted therapeutics that can restore immune competence and improve patient outcomes in cancer and autoimmune diseases.

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.