What CD39 inhibitors are in clinical trials currently?

11 March 2025
Introduction to CD39
CD39 is an ectonucleotidase that plays a fundamental role in converting extracellular adenosine triphosphate (ATP) and adenosine diphosphate (ADP) into adenosine monophosphate (AMP). Understanding CD39 is essential because it serves as a key “brake” on the pro-inflammatory signals released by ATP, thus shifting the balance toward an immunosuppressive microenvironment when AMP is further converted into adenosine by CD73. This enzyme is expressed on a range of cells—including immune cells, endothelial cells, and even tumor cells—and is central to controlling the levels of extracellular ATP and adenosine.

Role of CD39 in the Body
In normal physiology, CD39 is integral to the regulation of vascular thrombo-regulation and immunomodulation. Extracellular ATP, which is considered a damage-associated molecular pattern (DAMP), is released by stressed or dying cells, and CD39 is responsible for its rapid hydrolysis. This activity is crucial for preventing excessive inflammation and tissue damage because high ATP concentrations can lead to robust recruitment and activation of immune cells. By promptly converting ATP into AMP—and subsequently to adenosine via CD73—the activity of CD39 helps maintain immune homeostasis. Such regulation is pivotal in settings as varied as wound healing and preventing autoimmunity.

Importance of CD39 in Disease Pathology
In disease pathology, particularly in the realm of oncology, the CD39/CD73 adenosinergic pathway becomes problematic when overexpressed. Many tumors and the associated stromal elements upregulate CD39, which shifts the extracellular milieu to one enriched in adenosine. Adenosine binds to receptors on immune cells, resulting in the suppression of anti-tumor immunity. This mechanism of immune escape permits tumor cells to avoid cytotoxic T-cell responses and other immune effector mechanisms. Beyond cancer, aberrant CD39 activity has been implicated in inflammatory and thrombotic disorders, demonstrating the enzyme’s broad impact on pathophysiology.

CD39 Inhibitors
CD39 inhibitors have emerged as a promising class of immunomodulatory agents intended to block the enzyme’s activity, thereby maintaining higher levels of extracellular ATP and reducing the production of adenosine. These inhibitors come in various forms, most notably as monoclonal antibodies that bind specifically to CD39, allosterically inhibit its enzymatic activity, or induce CD39 downregulation on the cell surface. By halting or diminishing CD39 activity, these agents aim to reverse the immunosuppressive tone of the tumor microenvironment and foster more robust anti-tumor immune responses.

Mechanism of Action
The primary mechanism of action of CD39 inhibitors is to prevent the catalysis of ATP and ADP into AMP, and subsequently, the formation of adenosine. This mechanism works on two levels. First, by blocking CD39, the inhibitors help sustain high levels of ATP which is known to serve as a “danger signal” that recruits and activates immune cells—such as dendritic cells, CD8+ T cells, and natural killer (NK) cells—to the tumor site. Second, by lowering adenosine production, they reduce the activation of adenosine receptors (e.g., A2A and A2B) on immune cells, which are typically responsible for dampening immune function and promoting regulatory phenotypes in the tumor microenvironment. Most of the clinical strategies have focused on using monoclonal antibodies that can inhibit CD39 allosterically, meaning they bind to epitopes on the CD39 protein in a manner that changes its conformation and blocks its catalytic function without competing directly with high concentrations of ATP.

Potential Therapeutic Applications
Therapeutically, CD39 inhibitors have a dual rationale. In oncology, numerous studies have demonstrated that tumors engage the CD39/CD73 adenosinergic pathway to suppress anti-tumor immune responses. Blocking this pathway can therefore restore the activity and proliferation of cytotoxic T lymphocytes and NK cells, enhance dendritic cell maturation, and promote inflammatory cytokine production—all crucial for robust anti-tumor immunity. Beyond cancer, targeting CD39 is also being explored as a means to modulate immune responses in autoimmune disorders and in conditions where excessive adenosine generation plays a detrimental role. In preclinical studies, CD39 inhibition has been associated with improved outcomes in models of metastasis prevention and tumor regression, establishing the basis for advancing these inhibitors into early-phase clinical trials.

Clinical Trials of CD39 Inhibitors
Recent years have witnessed rapid advances in the clinical exploration of CD39 inhibitors. Several molecules have entered clinical trials, and these studies are predominantly focused on assessing the safety, tolerability, pharmacokinetics, and preliminary efficacy of these agents in cancer patients. A closer look at these trials is essential to understand which CD39 inhibitors are currently under clinical investigation and what the early results imply for future immuno-oncology therapies.

Current Trials and Phases
Multiple ongoing clinical trials are evaluating various CD39 inhibitors using different study designs. Among these, the recombinant fully human anti-CD39 monoclonal antibodies have received significant attention. For instance, the antibody JS019 is currently being evaluated in several Phase I clinical studies. One study assesses its activity in patients with advanced solid tumors or lymphomas, while another trial focuses exclusively on advanced solid tumors. A similar Phase I clinical study was reported investigating the safety, tolerability, and pharmacokinetic profile of JS019 monotherapy in patients with advanced solid tumors. In addition, JS019 is also being studied in a trial for patients manifesting advanced solid tumors or lymphomas, indicating its potential versatility across different tumor types. Another trial is investigating JS019 in subjects with hypereosinophilia in a randomized, double-blind, placebo-controlled Phase I/II design.

Beyond JS019, other CD39 inhibitors are being actively developed and assessed in clinical trials. One key agent is TTX-030, a human IgG4 CD39-targeted antibody. TTX-030 binds human CD39 and blocks its enzymatic activity allosterically, thereby modulating the tumor microenvironment by preserving extracellular ATP and curtailing adenosine production. It is in the early stages of clinical evaluation with early-phase studies designed to determine optimal dosing and to evaluate safety in patients with advanced tumors.

Another notable CD39 inhibitor is SRF617, an anti-CD39 monoclonal antibody developed by Surface Oncology. SRF617 is being evaluated in a Phase II clinical trial in combination with other agents, such as AB928 (an adenosine receptor antagonist) and AB122 (another immunomodulatory antibody), specifically in patients with metastatic castration-resistant prostate cancer. The design of these trials is to assess the synergistic effects of targeting multiple components of the adenosinergic pathway.

IPH5201 also represents one of the anti-CD39 agents that has entered first-in-human clinical evaluation. Like the aforementioned inhibitors, IPH5201 is designed to inhibit CD39 enzymatic activity and to enhance the local accumulation of immunostimulatory ATP in the tumor microenvironment. Although direct clinical trial details such as trial registration numbers and specific outcomes for IPH5201 are less detailed in the available references, it is widely recognized as part of the emerging landscape of CD39-targeted immunotherapies.

Supporting evidence from clinical trial databases further underscores these developments. For example, trial registration numbers indicate that JS019 is being tested in a Phase I setting, while others confirm the active clinical investigation of this antibody in various patient populations. Additionally, the trial shows JS019’s evaluation in an advanced disease setting, and another brings attention to its potential use in immunologically mediated disorders like hypereosinophilia. Collectively, these studies paint a picture of robust and multifaceted clinical investigation into the use of CD39 inhibitors, particularly in oncology settings, and in combination with standard chemotherapy and immune checkpoint inhibitors where immune suppression via adenosine has been implicated in treatment resistance.

Key Molecules and Developers
A detailed examination of the key molecules in this space reveals a diverse portfolio pursued by multiple developers:

• JS019 is a recombinant fully human anti-CD39 monoclonal antibody under clinical investigation. It is currently assessed in multiple Phase I trials involving advanced solid tumors and lymphomas. The design of these trials, including dose-escalation and pharmacodynamic endpoints, reflects the necessity to establish safety while evaluating preliminary efficacy. Although the specific corporate developer of JS019 is not always explicitly mentioned, the trial identifiers and comprehensive data underscore its promise as a therapeutic candidate.

• TTX-030 is developed by Tizona Therapeutics. As a human IgG4 CD39-targeted antibody, TTX-030 is engineered to bind to CD39 and allosterically inhibit its enzymatic function. Given its molecular design, TTX-030 aims to maintain higher extracellular ATP levels to stimulate an anti-tumor immune response. It is presently in early phase clinical trials, which are designed to assess its safety profile and potential to reverse immune suppression in a clinical setting.

• SRF617, developed by Surface Oncology, is another leading anti-CD39 antibody that has advanced into a Phase II clinical trial. SRF617 is being tested in combination with other targeted agents—specifically, in a study that combines it with AB928 (an adenosine receptor antagonist) and AB122 (an immunomodulatory antibody)—in patients with metastatic castration-resistant prostate cancer. The combination strategy is based on the rationale that simultaneous inhibition of CD39 and adenosine receptor signaling can synergize to produce a more potent anti-tumor effect than monotherapy.

• IPH5201 is cited as one of the antibodies entering first-in-human clinical studies. Although less detailed than JS019, IPH5201 is recognized as a promising CD39 inhibitor that works by blocking both membrane-bound and soluble forms of CD39, potentially enhancing dendritic cell and T-cell responses by preserving immunostimulatory ATP levels. Collaborations involving IPH5201 suggest joint developmental efforts, possibly with large pharmaceutical companies, to ensure robust evaluation in early clinical studies.

These candidates collectively represent the forefront of clinical investigation against CD39. Their development reflects the consolidated effort to utilize immunotherapy paradigms that target not only immune checkpoints like PD-1/PD-L1, but also components of the purinergic signaling pathway that foster tumor immune evasion. Each agent has its unique molecular properties, pharmacokinetic characteristics, and dosing regimens—all of which are being meticulously evaluated in the context of early-phase clinical trials designed to determine safety, tolerability, and preliminary efficacy.

Challenges and Future Directions
While the clinical potential of CD39 inhibitors is promising, several challenges must be addressed to ensure successful translation into clinical practice. As research in the field intensifies, researchers and developers are encountering various hurdles that span from molecular specificity to designing appropriate combination regimens.

Current Challenges in Development
One of the foremost challenges is achieving the optimal balance between therapeutic efficacy and safety. Since CD39 is involved in multiple physiological processes such as thromboregulation, indiscriminate inhibition could lead to unexpected adverse effects. Although monoclonal antibodies like JS019, TTX-030, SRF617, and IPH5201 are engineered for specificity, there remains a need to thoroughly evaluate the potential for off-target impacts or immune-related adverse events in clinical settings.

Another challenge is the determination of the appropriate dose and schedule. Many of the current trials are in Phase I, where the primary objective is to determine maximum tolerated doses and pharmacokinetic profiles. The pharmacodynamics of CD39 inhibition—namely, how long ATP levels remain elevated and adenosine levels suppressed—are critical parameters that affect therapeutic outcomes, yet they can vary considerably among patients due to differences in tumor biology and the microenvironment. This also ties into the broader challenge of inter-patient variability in clinical responses and the need for predictive biomarkers that can stratify patients likely to benefit from CD39 inhibition.

Combination strategies pose additional challenges. Many early-phase trials combine CD39 inhibitors with other agents such as PD-1/PD-L1 checkpoint inhibitors, chemotherapy, or other modulators of the adenosinergic pathway. While these combinations are scientifically appealing, they complicate the assessment of the individual contributions of each agent, and adverse event profiles may be additive or synergistic. Moreover, compensatory mechanisms in the purinergic signaling cascade—such as upregulation of CD73 or increased expression of adenosine receptors—may dampen the efficacy of single-agent CD39 inhibition unless appropriately managed by combination treatments.

Manufacturing and scaling of antibody-based therapies is another issue. Ensuring consistent quality and stability of recombinant antibodies across batches is crucial, especially when such products are intended for widespread clinical use. Furthermore, the cost of these biologics remains high, which could limit access if their development does not yield clear benefits over existing therapies.

Future Research Directions
Future research must focus on several fronts to overcome the hurdles identified above. One key area is the refinement of molecular design to enhance specificity. Advances in protein engineering and development of bispecific antibodies or antibody-drug conjugates may provide improved efficacy with lower toxicity, especially when these agents are combined with established immune checkpoint inhibitors. Preclinical data supporting the advantage of dual targeting (for instance, blocking both CD39 and CD73) suggest that future trials may incorporate more complex combination regimens that are aimed at mitigating compensatory feedback mechanisms.

There is also a critical need for the identification and validation of biomarkers that can predict which patients are most likely to respond to CD39 inhibition. Expression levels of CD39 on tumor cells and tumor-infiltrating immune cells, alongside measurements of adenosine concentration and ATP turnover in the tumor microenvironment, could serve as potential predictive markers. Collaborative efforts between academic centers and industry will be important to develop and standardize assays for these biomarkers, ultimately guiding personalized treatment strategies.

On the clinical trials front, adaptive trial designs that allow for modifications based on emerging pharmacodynamic or efficacy data could play an important role. Such designs would enable researchers to fine-tune dosing regimens and combination strategies as more real-time data become available. Additionally, longer follow-up periods and robust correlative studies are necessary to understand the durability of responses and to ensure that any potential immune-related toxicities are identified early in the course of treatment.

Research should also explore the use of CD39 inhibitors beyond oncology. Given the pivotal role of CD39 in regulating inflammation, future studies may assess their utility in autoimmune or chronic inflammatory diseases where adenosine-mediated immunosuppression either exacerbates or protects against pathology. Translational models of diseases such as rheumatoid arthritis, multiple sclerosis, or even sepsis could be valuable in expanding the therapeutic indications of these inhibitors.

Finally, an integrated approach to drug development will be required. This includes not only optimizing the molecular design but also ensuring that manufacturing processes are robust and economically viable. Strategic partnerships between biotech companies, academic institutions, and pharmaceutical giants will be essential to drive these agents from early-phase trials to later-phase studies and eventual regulatory approval. Future research should focus on head-to-head trials comparing CD39 inhibitors to other immunotherapeutic agents, as well as studies assessing their use in neoadjuvant or adjuvant settings to maximize their clinical benefit.

Conclusion
In summary, current clinical efforts are actively investigating several CD39 inhibitors with the aim of countering adenosine-mediated immunosuppression and enhancing anti-tumor immunity. A number of recombinant fully human anti-CD39 monoclonal antibodies, notably JS019, are under Phase I clinical investigation in patients with advanced solid tumors, lymphomas, and even hypereosinophilia. Additional agents such as TTX-030, developed by Tizona Therapeutics, and SRF617, developed by Surface Oncology, are being evaluated in early-phase and Phase II clinical trials, respectively. Furthermore, IPH5201 adds to the growing portfolio of first-in-human CD39 inhibitors that aim to restore immune activation within the tumor microenvironment by preserving extracellular ATP levels.

From a general standpoint, these developments are fueled by a growing understanding of CD39’s critical role in both normal physiology and pathological contexts, particularly in the immunosuppressive landscape of cancer – where increased CD39 activity contributes to treatment resistance by dampening anti-tumor immune responses. More specifically, the mechanism of action of these inhibitors revolves around blocking the enzymatic conversion of ATP to immunosuppressive nucleosides, thereby promoting an environment that is conducive to robust immune activation. The potential therapeutic applications are broad and include not only oncology indications but also potential applications in autoimmune and inflammatory diseases.

However, challenges remain. Clinical development must balance the benefits of CD39 inhibition against the risks of interfering with physiological processes, particularly given CD39’s involvement in thromboregulation and normal immune responses. Determining optimal dosing regimens, managing combination therapy complexities, validating predictive biomarkers, and ensuring manufacturing consistency are all critical areas that require further research and refinement.

Future directions include further molecular refinements to enhance inhibitor specificity, adaptive clinical trial designs that allow for real-time adjustments based on emerging data, and exploration of combination strategies that integrate CD39 inhibitors with other immunotherapeutic agents and conventional treatments. Ongoing and future studies will also need to elucidate the role of CD39 inhibition in non-oncology settings, potentially paving the way for broader therapeutic applications.

In conclusion, while several CD39 inhibitors such as JS019, TTX-030, SRF617, and IPH5201 are already in clinical trials, their journey from early-phase clinical testing to eventual regulatory approval will depend on continued research and a multidisciplinary approach to overcome current challenges. These agents represent a cutting-edge initiative in immuno-oncology with the promise of transforming treatment paradigms by reversing tumor-induced immune suppression. The evolving landscape of adenosinergic pathway inhibitors offers hope for improved clinical outcomes and potentially broader applications in the near future.

For an experience with the large-scale biopharmaceutical model Hiro-LS, please click here for a quick and free trial of its features

图形用户界面, 图示

描述已自动生成