What CD80 inhibitors are in clinical trials currently?

11 March 2025
Introduction to CD80 and Its Role
CD80 is a cell‐surface glycoprotein belonging to the B7 family of costimulatory molecules that is expressed primarily on antigen‐presenting cells such as dendritic cells, B lymphocytes, and monocytes. Its pivotal role in modulating the immune response lies in its ability to interact with receptors on T cells—most notably CD28 and CTLA‐4—to either promote or attenuate T-cell activation. Through these interactions, CD80 not only aids in the initiation of immune responses against pathogens and tumors but also contributes to maintaining immune tolerance to prevent autoimmunity. CD80’s ability to regulate the balance between immune activation and inhibition makes it an attractive target for therapeutic intervention in a range of diseases, including autoimmune disorders, transplantation rejection, and various forms of cancer.

Biological Function of CD80
At the molecular level, CD80 provides the “second signal” required for full T-cell activation. Following antigen recognition via the T-cell receptor, the binding of CD80 to CD28 on T cells reinforces the activation signal, promoting clonal expansion, cytokine production, and effector function. Conversely, engagement of CD80 with CTLA‑4, a receptor with much higher affinity for CD80 than CD28, leads to an inhibitory signal that dampens T-cell responses. This bidirectional role in costimulation and coinhibition validates CD80’s status as a checkpoint molecule in the immune system and highlights its importance in fine-tuning the immune balance to avoid uncontrolled inflammation and autoimmunity.

CD80 in Immune Response and Disease
Dysregulation of CD80 expression or function can drastically alter immunity. In autoimmune diseases, for example, an imbalance in CD80 expression on antigen-presenting cells may contribute to aberrant activation of autoreactive T cells, thereby exacerbating tissue damage. In the context of cancer, many tumors exploit the immune checkpoint network, including the pathways governed by CD80, to create a tolerogenic microenvironment that facilitates immune escape. In these instances, tumor cells may indirectly manipulate CD80-mediated signals or express altered levels of costimulatory molecules to suppress effective anti-tumor T-cell responses. Therefore, the modulation of CD80 signaling is seen as a promising strategy to restore immunosurveillance in cancer as well as to rein in pathological autoimmunity.

Overview of CD80 Inhibitors
Given CD80’s central role in modulating T-cell responses, a number of therapeutic strategies have emerged to inhibit its function. These inhibitors are designed to interfere with the interaction between CD80 and its T-cell receptors, thereby altering downstream signals that are critical for the initiation and regulation of immune responses.

Mechanism of Action
CD80 inhibitors work by competitively blocking the binding interface of CD80, thus preventing its engagement with costimulatory molecules such as CD28 and inhibitory receptors such as CTLA‑4. There are different modalities by which this inhibition is achieved:

• Monoclonal antibodies that specifically recognize and bind CD80 have been developed to block its interaction with T-cell receptors. For instance, clinical studies have investigated the use of anti-CD80 monoclonal antibodies in hematologic malignancies such as follicular lymphoma, where they have been shown to retard tumor cell proliferation by modulating pro- and anti-apoptotic signals.

• Competitive antagonist peptides are small molecule-based or peptide-based agents designed to mimic the binding regions involved in the CD80–CD28/CTLA‑4 interaction. These peptides adopt a polyproline type II helical conformation that structurally mimics the natural binding interface, thereby blocking receptor recognition and attenuating T-cell costimulation.

• Soluble forms of CD80 can function as decoy receptors that intercept ligands or inhibitory partners before they bind to the cell surface–expressed CD80. By sequestering these molecules—in some contexts, PD-L1 for instance—sCD80 can both neutralize immunosuppressive signals and provide a costimulatory boost to T cells.

• Small molecule inhibitors, often based on novel heterocyclic scaffolds, have also been patented as CD80 antagonists. These compounds are engineered to interfere with the CD80–CD28 interaction through binding to the shallow hydrophobic pockets on CD80. Although many of these are still in preclinical development, patents indicate that chemical compounds capable of robustly antagonizing CD80 are under exploration for therapeutic use.

Together, these approaches aim to interrupt the pathological signaling pathways driven by CD80 dysregulation while preserving or fine-tuning the delicate balance required for normal immune function.

Therapeutic Applications
Therapeutically, CD80 inhibitors are being investigated in several disease areas. In autoimmune disorders, where an overactive immune response may be driven by excessive costimulation, CD80 inhibitors could lead to reduced autoreactive T-cell activation and consequent amelioration of disease symptoms. Experimental models of multiple sclerosis have provided proof-of-principle that CD80 blockade can enhance the expression of protective molecules such as glucocorticoid-induced leucine zipper and prevent disease relapse.

In organ transplantation, modulation of CD80-mediated costimulation may help in inducing tolerance and reducing graft rejection. Agents such as belatacept—a CTLA‑4 fusion protein that binds to and blocks CD80/CD86—have shown utility in kidney transplantation by reducing the reliance on calcineurin inhibitors; although belatacept is not exclusively a CD80 inhibitor, it exemplifies a strategy of costimulation blockade that indirectly targets CD80 function.

In oncology, tumors that exploit the immune checkpoint system by dysregulating CD80 expression contribute to immune evasion. By inhibiting CD80’s interaction with its receptors, novel agents such as soluble CD80 can potentially restore anti-tumor T-cell responses. This is underlined by preclinical studies in animal models where sCD80 has been reported to “awaken” natural immune surveillance against tumor cells, paving the way for its development as an immunotherapeutic agent.

Current Clinical Trials of CD80 Inhibitors
Evaluating CD80 inhibitors in clinical trials is a relatively emerging field. The clinical landscape includes both early-phase studies of novel biologics and peptide-based inhibitors as well as later-phase evaluations of related costimulation blockade agents. Although the repertoire remains limited compared to more established checkpoints like PD‑1 or CTLA‑4, current clinical trials are yielding promising results and insights into the therapeutic modulation of CD80.

Ongoing Trials and Phases
One of the most notable clinical endeavors involves the use of anti-CD80 monoclonal antibodies in the treatment of hematological malignancies. Specifically, a Phase I/II study of a CD80-specific antibody has been conducted on patients with relapsed or refractory follicular lymphoma. In this study, signaling through CD80 was exploited to induce apoptosis and retard B-cell lymphoma proliferation. Although the study does not provide a commercial name for the antibody, its efficacy in modulating disease outcomes in a patient population that had exhausted other treatment options highlights the therapeutic potential of directly targeting CD80.

Additionally, competitive antagonist peptides have shown preclinical efficacy in experimental autoimmune encephalomyelitis—a murine model of multiple sclerosis—by enhancing the expression of anti-inflammatory molecules and reducing disease severity. Translational efforts are now being directed at moving these peptide-based inhibitors into clinical evaluation. The rapid pace of innovation in peptide engineering, coupled with advances in delivery methods, suggests that Phase I clinical trials evaluating these agents in conditions such as multiple sclerosis or other autoimmune disorders could be on the horizon.

Another developing candidate is soluble CD80. BriaCell Therapeutics, for instance, has secured an exclusive license from the University of Maryland, Baltimore County to develop sCD80 as a cancer therapeutic. The technology behind soluble CD80 leverages its dual ability to both block inhibitory signals and simultaneously provide a costimulatory boost to T cells. While details of clinical trial phases for sCD80 are still emerging, the licensing agreement and preclinical data suggest that early-phase clinical trials are either underway or in the advanced planning stages for its use in multiple tumor types.

It is important to note that while belatacept is a well-known agent that binds to CD80 and CD86, its role is that of a fusion protein to induce immune tolerance in transplant settings rather than a selective CD80 inhibitor. Nonetheless, belatacept’s success in clinical trials for kidney transplantation sets a precedent for the efficacy of CD80/CD86 blockade and reinforces the potential value of more selectively targeted CD80 inhibitors.

Furthermore, there are patents covering novel heterocyclic compounds that act as CD80 antagonists. Although these particular small-molecule inhibitors have not yet reached the clinical trial stage, their existence in the intellectual property space underlines active research in this area and anticipates future clinical development. Companies and academic laboratories are exploring these compounds with the aim of achieving robust CD80 blockade using oral or parenteral small molecules, which would add another modality to the CD80 inhibitor portfolio once first-in-human trials commence.

Collectively, the current clinical development of CD80 inhibitors remains in its nascent phases. Agents such as the anti-CD80 monoclonal antibody for lymphoma, peptide-based competitive inhibitors demonstrating promising preclinical data, and soluble CD80 for cancer immunotherapy represent the forefront of clinical investigations. Each candidate is at a different stage of clinical evaluation—from early phase studies to more advanced translational investigations—and targets distinct disease indications ranging from malignancies and autoimmune conditions to transplantation tolerance.

Key Players and Sponsors
Several academic institutions and biopharmaceutical companies are actively engaged in the development and clinical testing of CD80 inhibitors. For instance, the licensing deal between UMBC and BriaCell Therapeutics for the development of soluble CD80 indicates strong academic–industry collaboration and investment in the cancer immunotherapy space. BriaCell’s commitment to advancing sCD80 into clinical trials reflects the company’s strategic focus on novel biologics that modulate immune checkpoints.

In the hematologic space, clinical trials testing CD80-specific antibodies in relapsed or refractory follicular lymphoma have likely been supported by pharmaceutical sponsors with a history of developing monoclonal antibody therapies for B-cell malignancies. Such companies often have extensive experience in antibody engineering, clinical trial execution, and regulatory strategy. Although specific sponsors are not always named explicitly in the published literature, industry sources and collaborative research networks underscore the key role played by major biopharma entities in advancing CD80-targeted therapies.

Additionally, academic research groups specializing in peptide drug design are actively exploring CD80 competitive antagonist peptides. The translation of these peptides from bench to bedside is a collaborative effort involving academic laboratories known for innovative immunotherapy research and biopharmaceutical partners that provide clinical expertise, formulation development, and trial management support. Given the promising preclinical outcomes observed in models of multiple sclerosis and other immune-mediated conditions, these collaborations are expected to lead to early-phase clinical trials sponsored by either biotechnology startups or larger pharmaceutical companies seeking to expand their pipeline of immunomodulatory compounds.

Moreover, patents covering novel small molecule CD80 antagonists signal the potential entry of chemical inhibitor candidates into clinical testing. Though the companies owning these patents may be in early development stages, their continued R&D investments and licensing activities suggest that industry interest in small molecule CD80 inhibitors is growing. Key players in this arena will likely emerge as these compounds undergo clinical trial readiness evaluations and safety/efficacy studies in controlled settings.

Challenges and Future Directions
Despite the potential of CD80 inhibitors, several challenges and future research avenues remain to be addressed to fully translate these agents into effective therapies.

Current Challenges in Development
One significant challenge in the development of CD80 inhibitors is achieving the desired specificity. CD80 interacts with multiple receptors, and nonselective inhibition may blunt both stimulatory and inhibitory immune signals. This could lead to unintended consequences such as excessive immunosuppression or paradoxical immune activation, both of which can be associated with adverse clinical outcomes. For instance, while belatacept successfully blocks CD80/CD86 interactions in transplant patients, it exemplifies the delicate balance required in modulating costimulatory signals without compromising overall immune competence.

Another challenge is the identification of robust biomarkers. Reliable biomarkers are needed to predict responsiveness, monitor treatment effects, and guide dosing adjustments during clinical trials of CD80 inhibitors. The complexity of the immune system and the variability in CD80 expression among patients with different pathological conditions make it difficult to establish universally applicable markers. As observed in studies exploring CD80 expression in autoimmune disease and cancer, the dynamic regulation of this protein means that treatment effects must be monitored over time and in the context of the surrounding immune landscape.

There are also formulation and delivery issues to contend with, particularly for peptide-based inhibitors and novel biologics such as soluble CD80. Ensuring that these agents maintain stability, achieve adequate bioavailability, and reach designated target tissues without eliciting immunogenicity is vital. Additionally, the risk of off-target effects must be minimized to prevent complications, especially in chronic therapies where long-term immune modulation is involved. These challenges underscore the importance of rigorous preclinical studies before entering clinical phases.

From a regulatory standpoint, the relatively new approach of targeting costimulatory molecules like CD80 requires the development of novel clinical trial endpoints. Traditional efficacy metrics employed in oncology or autoimmunity trials may not fully capture the effects of modulating immune costimulation. As such, trial designs may need to integrate immune monitoring, functional assays, and clinical endpoints that reflect both efficacy and safety in the context of complex immunomodulatory pathways.

Future Research and Development Directions
Looking ahead, future research on CD80 inhibitors is expected to focus on a multipronged strategy that includes refining the specificity of inhibitors, expanding the breadth of indications, and developing combination strategies. One promising direction is the combination of CD80 inhibitors with other immunotherapies. For example, combining CD80 blockade with inhibitors of PD‑1/PD‑L1 could synergistically enhance anti-tumor immune responses—the concept being that while PD‑1 inhibitors relieve T-cell exhaustion, CD80 inhibitors can further modulate costimulation to optimize T-cell activation. Such dual or triple combination therapies may overcome the limitations of monotherapy and address primary or acquired resistance observed in immune checkpoint inhibitor treatments.

Additionally, further development of peptide-based inhibitors will likely be bolstered by advances in peptide synthesis, extended half-life strategies, and improved delivery systems. These advancements could enable a more targeted delivery to inflamed tissues or the tumor microenvironment while minimizing systemic exposure and adverse events.

The future of small molecule development against CD80 also holds promise. As indicated by patents covering novel heterocyclic compounds, there is ongoing research aimed at discovering orally bioavailable CD80 inhibitors that exhibit high affinity and selectivity. Success in this area could offer significant advantages in terms of ease of administration, patient compliance, and scalability of production. Early-phase clinical trials would then need to focus on defining pharmacodynamic profiles and establishing safe dosing regimens.

Furthermore, the integration of advanced biomarker discovery with clinical trial design will be crucial. Future research should focus on identifying molecular signatures that correlate with CD80 expression and function in diverse disease states. Such biomarkers could serve not only as predictors of response but also as early indicators of potential adverse events, thereby enabling more personalized and adaptive treatment strategies.

From a translational perspective, the lessons learned from approved agents such as belatacept can provide valuable insights. Although belatacept targets both CD80 and CD86, its successful application in kidney transplantation offers proof-of-concept that modulating costimulatory signals can improve clinical outcomes. Future CD80 inhibitors—whether they are monoclonal antibodies, peptides, or small molecules—must build on these results by demonstrating similar or improved safety profiles while offering greater specificity for CD80. This might involve innovative engineering approaches that fine-tune the binding kinetics and downstream signaling effects of the inhibitor.

Finally, collaboration between academic institutions, biotechnology startups, and large pharmaceutical companies will be instrumental in advancing the clinical development of CD80 inhibitors. Multidisciplinary collaborations that integrate immunology, medicinal chemistry, advanced drug delivery technologies, and robust clinical trial design are likely to accelerate the progress of these compounds from preclinical models into late-phase clinical trials and, eventually, clinical practice.

Conclusion
In summary, the current landscape for CD80 inhibitors in clinical trials is characterized by a blend of innovative biologics, peptide-based molecules, and emerging small molecule candidates. CD80, as a key costimulatory molecule in the immune system, holds significant therapeutic promise for the treatment of autoimmune diseases, cancer, and transplantation rejection. Its dual role in facilitating both stimulatory and inhibitory signals necessitates that therapeutic interventions be precisely tailored to modulate these pathways without causing broad immunosuppression or excessive immune activation.

Recent clinical studies have demonstrated the potential of anti-CD80 monoclonal antibodies in treating relapsed and refractory follicular lymphoma, where early-phase trials have yielded promising efficacy data. At the same time, competitive antagonist peptides designed to inhibit CD80 function have shown encouraging preclinical results in models of multiple sclerosis, with translational efforts underway to bring these molecules into first-in-human studies. The licensing of soluble CD80 by BriaCell Therapeutics for cancer immunotherapy further highlights the diverse therapeutic applications of CD80 inhibition, with early-phase clinical trials expected to assess its safety and efficacy in restoring anti-tumor immunity.

While the clinical pipeline for selective CD80 inhibitors remains in the early stages relative to more established immune checkpoint therapies, the innovative approaches being pursued—from monoclonal antibodies and peptides to small molecules—demonstrate a vibrant and expanding field. However, challenges remain; these include achieving the necessary specificity to modulate the CD80–CD28/CTLA‑4 axis without off-target effects, developing reliable biomarkers for monitoring clinical response, and designing trials that can adequately capture the complex immunomodulatory effects of these agents.

Future research will likely focus on combination therapies, which may pair CD80 inhibitors with other immunomodulatory agents such as PD‑1/PD‑L1 inhibitors to amplify therapeutic benefits while minimizing resistance. Advances in drug formulation, delivery systems, and molecular engineering are anticipated to refine these agents further, paving the way for personalized immunotherapy regimens. Continued collaboration across academic, biotech, and pharmaceutical sectors will be essential to overcome current hurdles and to establish CD80 inhibitors as a clinically validated therapeutic modality.

Overall, from the evidence provided in the synapse-sourced literature, it is clear that while CD80 inhibitors are still at an early phase of clinical evaluation, they represent a promising new frontier in the targeted modulation of immune responses. Their development – both as standalone therapies and in combination with other immunomodulators – holds great potential to address unmet clinical needs in autoimmune diseases, cancer, and transplantation. The emerging data and ongoing clinical trials will further inform the optimal use of these agents, ultimately guiding the next generation of personalized immunotherapy approaches in the clinic.

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