What CD40L inhibitors are in clinical trials currently?

11 March 2025
Introduction to CD40L and Its Role

CD40 ligand (CD40L), also known as CD154, is a critical costimulatory molecule that plays a central role in immune system activation. It is primarily expressed on activated T lymphocytes and, upon engagement with its receptor CD40—found on antigen‐presenting cells (APCs) such as B cells, dendritic cells, and macrophages—triggers a cascade of intracellular events that ultimately lead to adaptive immune responses. This interaction not only bolsters antibody production and B-cell differentiation but also orchestrates the maturation and cytokine secretion of dendritic cells, thereby influencing both humoral and cellular immunity.

Biological Function of CD40L

Biologically, CD40L is essential for regulating the interplay between T cells and B cells by providing the necessary “second signal” during immune activation. When T cells become activated, they express CD40L on their surface. The interaction of CD40L with the CD40 receptor on APCs results in the induction of costimulatory molecules and proinflammatory cytokines. These signals are crucial for initiating and sustaining B-cell proliferation, immunoglobulin isotype switching, and the formation of germinal centers. Moreover, CD40L can exist in both membrane-bound and soluble forms, with the soluble form (sCD40L) also being capable of triggering biological responses such as platelet activation and modulation of endothelial cell function.

CD40L in Disease Pathogenesis

In pathological conditions, dysregulation of CD40L expression or signaling has been implicated in the development and progression of several autoimmune and inflammatory diseases. Elevated levels of sCD40L are frequently observed in disorders such as systemic lupus erythematosus (SLE), rheumatoid arthritis, multiple sclerosis, and even in certain cardiovascular diseases, where it contributes to thromboembolic events by activating platelets. The pivotal role of the CD40–CD40L interaction has made it an attractive therapeutic target for modulating aberrant immune responses. However, the early clinical experience with CD40L-targeted agents has been complicated by safety concerns, particularly thrombotic complications thought to result from Fc-mediated activation of platelets.

Overview of CD40L Inhibitors

Therapeutic inhibition of CD40L aims to disrupt abnormal CD40–CD40L interactions that contribute to disease pathogenesis while preserving essential immune functions. This approach is generally achieved through the use of antibody-based agents that neutralize CD40L function, thereby decreasing proinflammatory and prothrombotic signaling.

Mechanism of Action

CD40L inhibitors are designed to block the binding of CD40L to its receptor, CD40. By preventing this interaction, these inhibitors can:
- Mitigate T-cell driven B-cell activation and subsequent autoantibody production.
- Reduce the maturation and activation of dendritic cells and macrophages, leading to a decrease in proinflammatory cytokine release.
- Potentially minimize the thromboembolic risks associated with the interaction of CD40L with platelet receptors (e.g., integrins) that contribute to platelet aggregation.

The majority of these inhibitors are monoclonal antibodies or antibody fragments engineered to neutralize CD40L. Notably, modifications such as removing or altering the Fc region have been implemented in newer agents to reduce the risk of Fcγ receptor-mediated platelet activation—a key factor implicated in the thromboembolic events observed with earlier CD40L-targeted therapies.

Types of CD40L Inhibitors

There are several types of CD40L inhibitors under investigation, primarily falling into antibody-based classes. The two most notable approaches include:

1. Full Monoclonal Antibodies
Early CD40L monoclonal antibodies (e.g., hu5c8) showed promising immunomodulatory effects; however, their development was halted due to safety issues related to thromboembolism. These agents bound to CD40L and interfered with its binding to CD40 but maintained an intact Fc region that could engage Fc receptors on platelets, leading to adverse events.

2. Fc-Modified Antibody Fragments
To overcome the limitations of conventional antibodies, newer agents have been engineered to lack a functional Fc domain. This design strategy diminishes the risk of platelet activation and subsequent thromboembolic complications. Among these, dapirolizumab pegol (formerly known as CDP7657) has emerged as a leading candidate. Similarly, humanized antibodies such as Toralizumab (also known as IDEC-131) have been revisited in clinical trials with improved safety profiles by either modifying the Fc region or employing dosing strategies that mitigate thrombotic risk.

Additionally, there are experimental immunotoxin constructs and bispecific molecules in development; however, in terms of clinical trials, the current focus remains primarily on Fc‐modified monoclonal antibodies.

Current Clinical Trials of CD40L Inhibitors

At present, two principal CD40L inhibitors have advanced into clinical trials following promising preclinical optimization and early-phase human studies. These include dapirolizumab pegol and Toralizumab/IDEC-131. Their development reflects the lessons learned from the initial setbacks with earlier anti-CD40L antibodies.

Ongoing Trials and Phases

Dapirolizumab Pegol (CDP7657)
Dapirolizumab pegol represents a significant evolution in CD40L inhibitor design. By engineering the antibody fragment to lack a functional Fc region, researchers aimed to eliminate the platelet activation mechanism that was responsible for the thromboembolic events observed with previous molecules such as hu5c8. Clinical studies with dapirolizumab pegol have demonstrated a favorable safety profile and promising immunomodulatory effects in patients with autoimmune diseases. Specifically, Phase III clinical trials are currently underway to evaluate its efficacy in systemic lupus erythematosus (SLE).
- Clinical Focus: The trial focuses on patients with SLE, a disease where heightened CD40–CD40L signaling contributes to autoantibody production, tissue inflammation, and end-organ damage.
- Study Design: The Phase III trial is multicenter, randomized, and double-blind, with endpoints that include suppression of T-cell-dependent antibody responses, reduction of disease activity, and careful monitoring for thromboembolic events.
- Interim Insights: Early-phase clinical data suggest that the drug effectively blocks CD40L without triggering the adverse thrombotic events noted in earlier agents. The modification preventing Fc-mediated interactions has been a pivotal development, and initial safety data indicate a prolonged target engagement with a favorable pharmacodynamic profile.

Toralizumab/IDEC-131
Another CD40L inhibitor that has been re-explored in clinical settings is Toralizumab, also known as IDEC-131. This humanized monoclonal antibody targets CD40L and has been evaluated in Phase I clinical trials, particularly in the context of multiple sclerosis (MS) and other autoimmune conditions.
- Clinical Focus: Toralizumab is being investigated for its potential to modulate the aberrant immune responses in MS conditions. By inhibiting CD40L, the drug seeks to dampen the inflammatory cascade that contributes to neural damage.
- Study Design: Phase I studies involve a dose-escalation design aimed at establishing safety parameters, assessing pharmacokinetics, and determining the optimal dosing regimen to achieve effective blockade of CD40L signaling without inducing systemic adverse events.
- Interim Insights: Preliminary findings from Phase I indicate that Toralizumab/IDEC-131 is well tolerated, with no significant thromboembolic events reported. These encouraging results provide a basis for further exploration in larger trials.

It should be noted that the overall clinical development of CD40L inhibitors has been shaped by earlier challenges. Initial clinical trials of anti-CD40L antibodies were marred by significant thromboembolic events, which prompted a reassessment of the molecular design and prompted the development of Fc-modified compounds such as dapirolizumab pegol. The reintroduction of CD40L inhibition strategies with improved safety profiles marks a critical step forward in harnessing the therapeutic potential of modulating this pathway.

Key Findings and Interim Results

The clinical data available thus far suggest that the modified approach to inhibiting CD40L is bearing fruit. Several key points have emerged from early-phase trials and ongoing studies:

1. Enhanced Safety Profile:
The removal or modification of the Fc region in dapirolizumab pegol has significantly reduced the risk of platelet activation and subsequent thromboembolic events. This design change differentiates it from earlier prototypes such as hu5c8, which were discontinued due to safety concerns.

2. Effective Target Engagement:
Biomarker studies from preliminary trials have demonstrated that dapirolizumab pegol achieves full receptor occupancy and is capable of suppressing T-cell-dependent antibody responses for an extended duration. These findings underscore the potential of the drug to exert its immunomodulatory effects effectively in clinical settings.

3. Promising Immunomodulatory Effects:
By blocking the CD40–CD40L interaction, these inhibitors are anticipated to reduce downstream proinflammatory cytokine production and mitigate the autoantibody-driven pathology observed in diseases such as SLE and MS. Early efficacy signals in terms of reduced disease activity and improved immunologic profiles have been noted, although comprehensive efficacy results from comprehensive Phase III studies are still pending.

4. Dose-Optimization Achievements:
The phase I trials of Toralizumab/IDEC-131 have been designed to carefully optimize the dosing regimen, thereby balancing the need for effective immunomodulation while minimizing adverse events. Early results in this cohort have been favorable, with no significant infusion reactions or thrombotic complications reported, which is critical given the historical challenges in this therapeutic area.

Taken together, these key findings highlight a renewed optimism in the clinical community regarding CD40L inhibitors. The success of these agents hinges on their ability to neutralize pathogenic CD40L-mediated signaling while preserving the necessary aspects of immune function.

Challenges and Future Directions

Despite the improvements achieved with the current generation of CD40L inhibitors, several challenges and opportunities remain as the field moves forward.

Safety and Efficacy Challenges

One of the central challenges in developing CD40L inhibitors has been balancing efficacy with safety. Early clinical trials highlighted the risk of thromboembolic events due to Fc-mediated platelet activation—a risk that necessitated a complete reevaluation of antibody design. Although modifications such as the removal of the Fc domain—as seen in dapirolizumab pegol—have mitigated these issues, long-term safety data remain crucial. Other challenges include:

- Monitoring for Subtle Adverse Effects:
Even with improved safety profiles, CD40L inhibitors must be monitored for subtle adverse effects related to immune suppression. Since the CD40–CD40L axis also plays a role in normal immune surveillance and inflammation, there is a risk of increased susceptibility to infections or impaired responses to vaccines.

- Defining Optimal Dosing Regimens:
Determining the optimal dosing regimen to achieve maximal therapeutic benefit without side effects is critical. Phase I trials for Toralizumab/IDEC-131 are currently focused on defining the minimal effective dose that ensures complete receptor blockade while limiting adverse events.

- Heterogeneity in Patient Populations:
Autoimmune diseases, such as SLE and MS, present with diverse clinical manifestations and underlying immunopathology, leading to variability in patient responses. Biomarker-driven patient stratification and personalized treatment regimens will likely play an increasingly significant role in future clinical trials, ensuring that patient subgroups most likely to benefit from CD40L inhibition are accurately identified.

- Combination Therapy Considerations:
Given the complexity of autoimmune pathogenesis, CD40L inhibitors might need to be used in conjunction with other immunomodulatory agents (e.g., PD-1/PD-L1 inhibitors or conventional immunosuppressants) to achieve optimal disease control. Each combination therapy introduces additional layers of risk, such as compounded immunosuppression, which demands careful clinical evaluation.

- Regulatory and Translational Hurdles:
The translation from promising Phase I/II results to Phase III efficacy, and eventually regulatory approval, is fraught with financial, logistical, and scientific challenges. Regulatory agencies will require robust data not only on the efficacy and safety of these agents but also on their long-term impact on patients’ quality of life and disease progression.

Potential Future Developments

Looking forward, there are several avenues for development that could further refine the therapeutic index of CD40L inhibitors:

1. Generation of Next-Generation Antibody Formats:
Future innovations may involve the development of bispecific antibodies that simultaneously target CD40L and another relevant molecule to enhance specificity and reduce off-target effects. Novel scaffolds such as single-chain variable fragments (scFv) or nanobodies may offer additional design flexibility to improve tissue penetration and half-life.

2. Enhanced Biomarker-Guided Therapeutics:
As our understanding of the CD40–CD40L signaling cascade deepens, a refined set of biomarkers could be established to predict therapeutic responses and guide patient selection. In addition to standard immunologic and clinical markers, genomic, proteomic, and cellular biomarkers may contribute to personalized therapeutic strategies.

3. Exploration in Combination Regimens:
There is significant potential for combining CD40L inhibitors with other immunotherapies to achieve synergistic effects. For example, pairing dapirolizumab pegol with immune checkpoint inhibitors may amplify antitumor immune responses in solid cancers that exhibit autoimmune features. Similarly, combination with standard-of-care treatments in autoimmune diseases could result in additive or synergistic benefits while allowing lower doses of each agent to minimize toxicity.

4. Expansion to Other Indications:
Although current trials focus primarily on autoimmune diseases such as SLE and MS, the role of CD40–CD40L signaling in other conditions—such as transplant rejection, atherosclerosis, and even certain malignancies—provides a rationale for broadening the scope of clinical trials. Future studies might assess the efficacy of CD40L inhibitors in these contexts, provided that safety concerns are adequately addressed.

5. Advanced Drug Delivery Systems:
Novel delivery systems, including nanoparticle encapsulation or targeted delivery vehicles (e.g., conjugation with tumor-targeting ligands), could improve drug distribution and reduce systemic exposure. Such strategies may further enhance the safety profile of CD40L inhibitors by localizing their activity to diseased tissues and minimizing off-target effects.

6. Addressing Immunogenicity and Tolerance:
As with many biologic therapies, the risk of developing anti-drug antibodies remains a concern. Ongoing research into protein engineering and glycosylation patterns may help to reduce immunogenicity, ensuring prolonged efficacy and reducing the risk of adverse immune responses.

Conclusion

In summary, the development of CD40L inhibitors is an evolving field driven by the urgent need to modulate deleterious immune responses in a variety of autoimmune and inflammatory diseases. In the introduction, we outlined the biological function of CD40L and its role in disease pathogenesis, emphasizing its importance in normal immune function as well as in the development of conditions such as SLE and MS. In the overview section, we discussed the mechanisms of action and types of CD40L inhibitors, focusing on how neutralizing antibodies and specially engineered antibody fragments can block the CD40–CD40L interaction effectively.

The current clinical trial landscape reveals two key agents:

Dapirolizumab pegol (CDP7657):
A next-generation CD40L inhibitor that has been engineered to lack the Fc region to avoid the thromboembolic complications observed with earlier antibodies. It is presently in a Phase III clinical trial investigating its safety and efficacy in patients with systemic lupus erythematosus (SLE). Early data suggest that dapirolizumab pegol achieves full receptor occupancy, effectively suppresses T-cell-dependent antibody responses, and has a favorable safety profile.

Toralizumab/IDEC-131:
A humanized anti-CD40L monoclonal antibody currently undergoing Phase I trials, particularly in the context of multiple sclerosis. This agent has also been optimized to reduce Fc-mediated unwanted events and has shown promising safety results with no significant thromboembolic events reported to date.

Both of these agents reflect the current focus on improving the therapeutic index of CD40L inhibitors through molecular modifications that mitigate previous safety issues while preserving—or even enhancing—the desired immunomodulatory effects.

Looking ahead, the challenges remain significant. Dosing, long-term safety, patient heterogeneity, and combination therapy strategies represent key hurdles that must be overcome to bring CD40L inhibitors to broader clinical use. Advances in antibody engineering, biomarker development, and drug delivery systems, along with rigorous clinical testing, promise to further refine these therapies. The future of CD40L inhibition may well depend on personalized treatment approaches that tailor therapy to individual patient profiles, thereby maximizing efficacy while minimizing risk.

In conclusion, as we transition from earlier, less refined CD40L inhibitors to these newer, Fc-modified agents like dapirolizumab pegol and Toralizumab/IDEC-131, the field is entering an exciting phase. Early clinical data affirm the potential of these agents to provide effective immunomodulation without the previously encountered safety liabilities. Continued research and carefully designed clinical trials will be pivotal in confirming these early successes and determining the long-term benefits for patients with autoimmune and inflammatory diseases. The current pipeline, as evidenced primarily by these two agents, highlights the promise of CD40L-targeted therapies in offering new treatment avenues—a promise that may well extend beyond autoimmunity into other areas of immune-mediated pathology once challenges are systematically addressed.

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.