What are the therapeutic candidates targeting OX40?

11 March 2025
Introduction to OX40
OX40, also known as CD134 or TNFRSF4, is a member of the tumor necrosis factor receptor (TNFR) superfamily that plays a critical role in regulating immune responses. It is expressed primarily on activated T cells and is transiently upregulated following antigen recognition. As an important co‐stimulatory molecule, OX40 is essential for amplifying T cell responses, sustaining effector functions, and promoting memory T cell survival. These characteristics have made it an attractive target for immunomodulatory therapies in both cancer and inflammatory diseases. Understanding the structure and function of OX40 is fundamental to appreciating the therapeutic candidates that have been designed to target this receptor.

OX40 Structure and Function
OX40 is a type I transmembrane glycoprotein characterized by an extracellular domain that interacts with its natural ligand OX40L (CD252), a transmembrane protein expressed on antigen‐presenting cells (APCs) such as dendritic cells, B cells, and activated endothelial cells. The binding of OX40L to OX40 initiates intracellular signaling cascades that result in the activation of nuclear factor kappa B (NF-κB) and other transcription factors. These events ultimately promote T cell proliferation, survival, and differentiation while also counteracting the suppressive function of regulatory T cells (Tregs). The receptor’s structure, with its cysteine-rich domains, allows for high-affinity interactions that can be modulated by engineered antibodies or fusion proteins, thereby tailoring the immune response.

Role of OX40 in Immune Response
OX40 signaling is critical in both the initiation and maintenance phases of T cell activation. Upon antigen recognition via T cell receptors (TCRs), the inducible expression of OX40 on T cells provides an additional “second signal” that ensures robust cytokine production, proliferation, and the survival of T cells during an immune response. This costimulatory pathway not only reinforces the expansion of effector T cells but also helps shape the development of immunological memory, which is particularly useful in long-term protection against pathogens and in antitumor immunity. Moreover, by modulating the function of Tregs, OX40 engagement can tip the balance toward a more pro-inflammatory and cytotoxic immune response, a property that is being harnessed in cancer immunotherapy.

Therapeutic Candidates Targeting OX40
The targeting of OX40 has led to the development of various therapeutic candidates that are designed to either enhance (agonists) or, in some contexts, inhibit (antagonists) the OX40 signaling pathway. These candidates are being explored in diverse indications, such as cancer—where the aim is to amplify antitumor T cell responses—and autoimmune diseases, where modulating excessive inflammation is desired. The strategies include monoclonal antibodies, fusion proteins, and other engineered molecules that can alter the receptor’s behavior.

Overview of Current Candidates
There is a broad spectrum of therapeutic candidates targeting OX40, and many are in different stages of development. Among the promising candidates, a number have emerged from both established and emerging biopharmaceutical companies with preclinical and clinical data supporting their further development. Key candidates include:

• Rocatinlimab (developed by Amgen, Inc.)
Rocatinlimab is an afucosylated, humanized anti-OX40 receptor antibody that acts as an agonist. In clinical studies, it has demonstrated significant improvements in clinical scores in conditions such as atopic dermatitis, as evidenced by outcomes in patient-reported measures (IGA and vIGA-AD reductions) and improvements in SCORAD and DLQI scores during Phase 2 trials. Its design aims to enhance T cell activation, driving potent antitumor and anti-inflammatory immune responses.

• MEDI6469
MEDI6469 is another humanized agonistic anti-OX40 antibody that has been studied in early-phase clinical trials. It was evaluated as a monotherapy as well as in combination with other immunotherapeutic agents, with evidence that preoperative administration increases T cell activation and proliferation both in blood and directly within tumor microenvironments.

• INCAGN01949
Developed by Agenus, Inc., INCAGN01949 is an agonistic anti-OX40 monoclonal antibody with promising characteristics to enhance CD4+ T cell responses while also mediating the selective depletion of suppressive Tregs in the tumor microenvironment. Preclinical studies and early clinical data support its potential to improve antitumor efficacy across various tumor models.

• IMG-007
IMG-007 is an OX40 monoclonal antibody candidate under investigation by Inmagene Biopharmaceuticals. Detailed in a study, IMG-007 has shown favorable pharmacokinetics and safety profiles in healthy adult subjects, with dose-proportional increases observed in extended half-life parameters and favorable efficacy signals as indicated by improvements in eczema severity scores.

• YH002
YH002 is a recombinant humanized agonistic anti-OX40 IgG1 monoclonal antibody that has entered early-phase clinical trials. It is being evaluated for its potential in the treatment of advanced solid tumors. Early clinical results indicate that YH002 is safe and tolerable, with dose-escalation studies revealing no dose-limiting toxicities, and a favorable immune activation profile as evidenced by preliminary efficacy data.

• AGX-501
AGX-501 is a fusion protein candidate developed by AgonOx LLC that targets OX40. Classified as a fusion protein, it represents a slightly different approach than conventional IgG monoclonal antibodies as it aims to simultaneously engage the receptor with high avidity and modulate the immune response in a controlled manner. Currently, AGX-501 is in Phase 2 development, and its mechanism exploits the high binding specificity to enhance T cell activation while minimizing adverse events.

• BGB-A445
From BeiGene Ltd., BGB-A445 is an agonistic candidate that has been studied in the context of oncology. Clinical data indicate that BGB-A445, either as a monotherapy or in combination with other immunomodulatory therapies (for instance, Tislelizumab), induces treatment-emergent adverse events (TEAEs) of grade ≥3 in a significant fraction of patients, which informs its dose selection and safety profile in early-phase studies.

Additionally, candidates under development by Incyte Biosciences International SARL have been noted in multiple entries in preclinical licensure filings. Although specific product names were not always provided, these entries reflect an ongoing interest in the OX40 pathway by established biopharmaceutical organizations, and they indicate that anti-OX40 pharmacotherapies are being pursued both as monotherapies and in combination paradigms.

Even though most of these agents are engineered to act as agonists to stimulate the OX40 receptor, there are also instances of therapeutics designed to inhibit OX40 signaling for diseases where excessive T cell activation is detrimental. However, the majority of candidates discussed in the available literature, particularly in the synapse-verified sources, are agonistic in nature and focus on enhancing immune responses for cancer immunotherapy and for ameliorating inflammatory conditions such as atopic dermatitis.

Mechanisms of Action
Candidate molecules targeting OX40 are engineered to exert their therapeutic effects primarily through the modulation of T cell co-stimulation. In general, the following mechanistic principles apply:

• Agonistic Signaling
Agonistic antibodies like rocatinlimab, MEDI6469, INCAGN01949, IMG-007, and YH002 bind to the OX40 receptor on activated T cells, mimicking the action of the natural ligand OX40L. This engagement triggers multiple downstream signaling cascades that boost cytokine secretion, proliferation, and survival. Such responses help to generate robust effector T cell populations and enhance the generation of immunological memory, thereby improving the capacity to mount a sustained antitumor response.

• Modulation of T Regulatory Cells
A unique aspect of OX40 targeting is its dual impact on conventional versus regulatory T cells. When OX40 is activated on Tregs, it can diminish their suppressive capacity, permitting a more vigorous effector T cell attack on tumor cells. Candidates like INCAGN01949 have been specifically designed to exploit this differential regulation, thereby reducing intratumoral immune suppression and favoring antitumor activity.

• Enhanced Immune Cell Activation
By providing an additional costimulatory signal, these agents not only drive the expansion and activation of CD4+ and CD8+ T cells but also potentiate the natural tumoricidal functions of natural killer (NK) cells. Enhanced production of cytokines, such as interleukin-2 (IL-2) and interferon-gamma (IFNγ), is a hallmark of effective OX40 engagement, ultimately resulting in a robust systemic antitumor immune response.

• Fusion Protein Strategies
Fusion proteins such as AGX-501 combine the targeting specificity of antibodies with engineered domains that may increase binding avidity or extend half-life. This approach can result in more sustained receptor engagement and prolonged activation of T cells compared to conventional antibodies. Such mechanisms are particularly valuable in situations where continuous stimulation is needed for optimal therapeutic efficacy.

• Combination with Other Therapeutic Modalities
Many OX40-targeting agents are being evaluated in combination with other immunomodulatory therapies, such as PD-1 or CTLA-4 inhibitors. The rationale behind these combinations is to simultaneously block inhibitory checkpoints while providing strong co-stimulatory signals through OX40, ultimately synergizing to overcome tumor-induced immune suppression and resistance mechanisms.

Clinical Trials and Efficacy
Clinical trials evaluating OX40-targeting therapeutics have been structured to assess both the immunostimulatory potential of these agents and their safety profiles. The trials span early phases—ensuring safety and establishing recommended dosing—and later phases that address clinical efficacy in terms of tumor shrinkage, durable responses, and improved patient-reported outcomes. Data emerging from these studies are promising, particularly in indications such as atopic dermatitis and various solid tumors.

Current Clinical Trials
Several notable clinical trials are ongoing, reflecting the wide interest in OX40-targeted therapies:

• Rocatinlimab has been studied in Phase II and is advancing into Phase III trials, particularly in atopic dermatitis. In these trials, endpoints such as the Investigator Global Assessment (IGA) and the Eczema Area and Severity Index (EASI) have been used to demonstrate statistically significant improvements compared to placebo. These trials have enrolled hundreds of patients across multiple clinical sites in different geographical regions, with trial designs that incorporate both on-treatment and off-treatment follow-up periods to ascertain the durability of responses.

• MEDI6469 has been evaluated in Phase 1b/2 studies, with early data indicating that preoperative administration not only is safe but also increases activation markers on both CD4+ and CD8+ T cells in the tumor microenvironment. These studies have provided valuable pharmacokinetic data and defined maximum tolerated doses (MTDs) in patients with advanced solid tumors and B-cell lymphomas.

• INCAGN01949 is in early-phase trials primarily focused on patients with advanced or metastatic tumors. Clinical data support its mechanism of action by demonstrating enhanced T cell proliferation and an increase in effector cytokine production, which correlate with observed clinical improvements in tumor control.

• IMG-007, in trials, is being evaluated in healthy subjects initially to determine safety and pharmacokinetic profiles. Following encouraging results, IMG-007 is expected to move into patient populations to assess its efficacy in reducing symptoms of immune-mediated diseases such as atopic dermatitis.

• YH002 has been tested in phase 1 dose-escalation studies in advanced solid tumors across multiple sites in the United States and China. Early reports indicate a favorable safety profile without significant dose-limiting toxicities, and preliminary efficacy data point to immune activation consistent with the proposed mechanism of action.

• AGX-501, as a candidate fusion protein, has entered Phase 2 trials, where its unique mechanism and prolonged receptor engagement are being closely monitored. While detailed clinical outcome data for AGX-501 are still emerging, its design is anticipated to provide superior sustained T cell stimulation compared with conventional antibody formats.

• BGB-A445, with its robust preclinical characterization and early-phase clinical data, is being assessed in the realm of oncology. Early integration studies, including combination regimens with checkpoint inhibitors such as Tislelizumab, have provided important insights into the balance between efficacy and adverse events, with noted treatment-emergent adverse events (TEAEs) informing ongoing dose adjustments.

Efficacy and Safety Data
The efficacy of these agents has been measured using a variety of endpoints, ranging from changes in clinical scores (such as EASI, SCORAD, and DLQI in atopic dermatitis) to objective measures of tumor response (tumor volume reduction, progression-free survival, and overall survival in oncology). Safety profiles have been of significant interest given that systemic immune activation can lead to cytokine release syndrome and other immune-related adverse events. Detailed summaries across different candidates include:

• Rocatinlimab has shown that in Phase II trials, its administration resulted in significant improvement in patient-reported outcomes and skin scores compared to placebo. For instance, improvements in IGA (with a ≥2-point reduction in 24-week assessments) demonstrated a clear clinical benefit, while safety data indicated that adverse events were manageable and primarily consisted of mild-to-moderate infusion reactions.

• MEDI6469’s early-phase trials have established that it is well tolerated, with no significant dose-limiting toxicities reported. Its impact on peripheral and intratumoral T cell activation serves as a biomarker for its activity, and the data support its potential to synergize with other immunotherapeutic agents in combination regimens.

• Clinical results for INCAGN01949 have emphasized its ability to enhance effector T cell proliferation and reduce the presence of immunosuppressive regulatory T cells. Early markers of antitumor activity, including increased cytokine secretion and tumor-infiltrating lymphocyte expansion, have been observed. Safety signals in phase 1/2 studies suggest that while the treatment is generally well tolerated, careful dosing is required to avoid excessive immune-mediated toxicities.

• IMG-007 has demonstrated a favorable pharmacokinetic profile when administered to healthy adults. Dose-escalation studies have shown dose-proportional pharmacokinetics, implying predictable behavior in vivo. Although further trials in patient populations are awaited, early safety data are encouraging, and improvements in EASI scores support its potential efficacy in indications like atopic dermatitis.

• YH002’s Phase 1 studies in advanced solid tumors have helped establish its safety profile. The absence of dose-limiting toxicities and preliminary evidence of tumor immune activation indicates that YH002 can be safely administered at doses that provoke the desired immunologic response. Additionally, early signals hint at potential antitumor efficacy, which positions the candidate for further efficacy studies in subsequent phases.

• For AGX-501, the clinical efficacy data are still emerging; however, its mechanism as a fusion protein is designed to deliver sustained OX40 receptor engagement, potentially leading to durable T cell activation. Early-phase trials will further elucidate its safety, but preclinical data indicate that its design may circumvent some of the limitations seen in conventional antibody therapies.

• BGB-A445, evaluated in trials for oncology indications, has shown a notable incidence of higher-grade treatment-related adverse events when used as a monotherapy (~41% for grade 3 events) and even higher when combined with PD-1 inhibitors (up to 53% in certain cohorts). This emphasizes the need for careful patient selection and dosing regimens to maximize efficacy while minimizing risks. Despite these challenges, the observed immune activation suggests that, once optimally dosed, BGB-A445 could become an important component of combination immunotherapy regimens.

Challenges and Future Directions
Despite the promising results in early-stage clinical trials and preclinical models, the development of OX40-targeted therapies has not been without challenges. As researchers continue to optimize these therapeutic candidates, several key areas of concern and opportunity remain.

Development Challenges
The primary challenges associated with the development of OX40-targeted therapies can be broadly categorized into issues of safety, dosing, and patient selection. Specific challenges include:

• Balancing Efficacy and Safety
Since OX40 agonists work by potentiating T cell activation, there is an inherent risk of overstimulation of the immune system, leading to cytokine release syndrome or other autoimmune side effects. Clinical data from candidates such as BGB-A445 underscore that treatment-emergent adverse events must be carefully managed through dose titration and optimized combination strategies.

• Determining Optimal Dosing Regimens
The kinetics of OX40 expression on T cells are transient and can be influenced by the timing of antigen exposure. Therefore, identifying the optimal dosing schedule that aligns with the peak expression of OX40 on T cells is of paramount importance. For instance, preoperative administration studies with MEDI6469 and IMG-007 have provided key insights into the timing of dosing relative to antigen presentation, but further refinement is needed in larger patient populations.

• Patient Selection and Biomarkers
Another significant challenge is the identification of biomarkers that can predict responsiveness to OX40-targeted therapies. Given the variability in immune system status among patients and differences in tumor microenvironments, predictive biomarkers (such as OX40 expression levels, tumor mutation burden, or T cell infiltrate density) will be pivotal to selecting patients who are most likely to benefit from these therapies.

• Combination Therapy Complexities
While combining OX40 agonists with checkpoint inhibitors (e.g., PD-1 or CTLA-4 blockers) offers a synergistic mechanism, the complexity of immune interactions in such regimens adds additional layers of risk. The timing, sequencing, and dosing of each agent must be carefully controlled to avoid counterproductive immune effects or excessive toxicity.

• Manufacturing and Product Stability
For fusion proteins such as AGX-501 and antibody-based therapeutics, maintaining product stability and ensuring scalable manufacturing processes can be challenging. These factors are critical not only for clinical development but also for eventual commercial success.

Future Research Directions
Looking forward, several avenues of research and development are expected to drive improvements in the field of OX40-targeted therapy:

• Optimization of Combination Therapies
Future studies will likely focus on integrating OX40 agonists with other immunotherapies such as checkpoint inhibitors and cancer vaccines. Combination regimens will be designed based on comprehensive pharmacodynamic and pharmacokinetic data to maximize efficacy while controlling toxicity. Innovative trial designs that adapt dosing based on real-time biomarker feedback are anticipated to improve patient outcomes.

• Refinement of Biomarker Strategies
The identification and validation of biomarkers for patient selection remains an unmet need. Ongoing and future research will aim to incorporate next-generation sequencing, transcriptomic profiling, and immune phenotyping to better characterize patient populations and predict responses to OX40-targeted interventions. This approach will help tailor therapies to individual patient profiles and enhance the likelihood of sustained clinical responses.

• Exploration of Novel Molecular Formats
In addition to conventional monoclonal antibodies, alternative molecular formats such as bispecific antibodies, antibody fragments, and engineered fusion proteins are expected to be explored further. These formats may offer improved tissue penetration, enhanced receptor clustering, and more controlled activation of T cell responses. AGX-501 is an early example of how fusion protein design can overcome some inherent limitations of standard antibodies.

• Understanding Resistance Mechanisms
As with many immunotherapies, resistance to OX40-targeted treatments may develop over time. Future research will likely delve into the mechanisms of primary and acquired resistance, including the modulation of OX40 expression and alterations in T cell signaling pathways. Addressing these challenges could lead to the development of next-generation agents or combination strategies that overcome resistance and improve long-term outcomes.

• Translational and Real-Time Clinical Studies
Real-time biomarker studies and translational research embedded in clinical trials will help in understanding the mechanistic underpinnings of OX40-targeted therapies. For instance, longitudinal analysis of T cell activation markers, cytokine profiles, and regulatory cell populations during treatment will provide important insights into the dynamics of the immune response. These data can then be used to refine treatment protocols and dosage regimens.

• Application Beyond Cancer
Although much of the current focus has been on oncologic indications, the role of OX40 in immune regulation suggests a potential for these therapies in autoimmune and inflammatory diseases. Exploring the differential impact of OX40 modulation on effector versus regulatory T cells could pave the way for therapeutic interventions in conditions such as atopic dermatitis or rheumatoid arthritis, where controlled modulation of the immune system is required.

• Regulatory and Commercial Considerations
Given the complexity of immune therapies, regulatory agencies will continue to work closely with developers to ensure that novel OX40-targeted agents meet safety and efficacy benchmarks. Future directions will involve streamlined manufacturing processes and collaborative efforts to address both clinical and commercial hurdles, ultimately ensuring that these therapies can reach a broader patient population.

Conclusion
In conclusion, therapeutic candidates targeting OX40 represent a highly innovative and rapidly evolving area in immunotherapy. Agents such as rocatinlimab, MEDI6469, INCAGN01949, IMG-007, YH002, AGX-501, and BGB-A445 highlight the diverse approaches being used to modulate this critical immune checkpoint. These candidates work primarily by engaging the OX40 receptor to enhance T cell activation, proliferation, and survival, while concurrently reducing the suppressive influence of regulatory T cells. The promising clinical data from early-phase trials—demonstrated by significant improvements in both objective endpoints such as tumor regression and subjective patient-reported outcomes in conditions like atopic dermatitis—underscore the therapeutic potential of OX40 agonists.

However, challenges remain in optimizing dosing schedules, managing immune-related adverse events, and selecting appropriate patient populations based on robust biomarkers. Furthermore, the complexity of combining these agents with other immunotherapies demands innovative trial designs and close monitoring of safety signals. Future research will likely focus on refining these combination strategies, developing novel formats such as bispecific antibodies and fusion proteins, and expanding the application of OX40-targeted therapies to a broader spectrum of diseases, including autoimmune disorders.

In summary, while the journey from preclinical discovery to clinical implementation continues to pose significant challenges, the steady progression of research in this area, supported by detailed synapse-verified data, presents a hopeful scenario. The current landscape, marked by an array of candidates with varied mechanisms of action, extensive clinical trial activity, and a clear path for future research, indicates that OX40-targeted therapies will play an increasingly important role in the next generation of immunomodulatory 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.