How many FDA approved Trispecific T-cell engager (TriTE) are there?

17 March 2025

Introduction to Trispecific T-cell Engagers (TriTE)

Trispecific T-cell engagers, often abbreviated as TriTEs, are advanced engineered antibody-based molecules designed to simultaneously bind three distinct epitopes. Typically, one binding arm engages CD3 on T cells, a second binds to a tumor-associated antigen (TAA) on target cancer cells, and the third can be directed either towards an additional antigen on the tumor for enhanced specificity or a costimulatory receptor on the T cell to improve its activation and cytotoxic functions. This architecture enables these molecules not only to redirect T-cell cytotoxicity towards cancer cells in a manner similar to bispecific T-cell engagers (BiTEs) but also to potentially overcome issues such as antigen escape and insufficient T-cell priming. The third binding domain, depending on the design, may add layers of selectivity or function by simultaneously engaging costimulatory signals (e.g., CD28 or CD137) or targeting additional tumor markers. This multifaceted engagement is theorized to provide an enhanced activation profile and potentially mitigate the immune-suppressive elements in the tumor microenvironment.

Evolution of T-cell Engaging Therapies 
Traditional T-cell engaging therapies began with the development of bispecific molecules such as blinatumomab, a CD19 × CD3 bispecific T-cell engager that received FDA approval in 2014 for acute lymphoblastic leukemia (ALL). Although blinatumomab was a breakthrough, it is important to note that it operates via a two-binding-domain mechanism, effectively bridging T cells and cancer cells without additional costimulatory enhancement. Subsequent research and development have shifted toward more complex modalities like TriTEs, which build upon the success of BiTEs by integrating an extra binding moiety to either target a second tumor-associated antigen or to incorporate an agonistic costimulatory signal. These advanced formats are generally still in the experimental or early clinical stages of development, being tested in both hematologic malignancies and solid tumors. The evolution from bispecific to trispecific molecules signals an attempt to address some of the limitations observed with early T-cell engagers—such as tumor escape due to antigen loss, insufficient T-cell persistence, and on-target off-tumor toxicities—by designing molecules with multi-antigen specificity and integrated signaling functions.

FDA Approval Process for Biologics

Overview of FDA Approval Stages 
The U.S. Food and Drug Administration (FDA) approval process for biologics is a rigorous, multi-step pathway designed to ensure safety, efficacy, and quality. The process begins with preclinical studies, often involving in vitro assays and animal models to assess pharmacodynamics, pharmacokinetics, and toxicity profiles. Once preclinical data convincingly support the therapeutic potential, the investigational new drug (IND) application is submitted to the FDA, after which Phase 1 clinical trials are initiated to examine safety and dosing in a small cohort of patients. This is followed by sequential Phase 2 and Phase 3 clinical trials which expand the patient population and evaluate both efficacy and long-term safety. Finally, a Biologics License Application (BLA) is submitted for FDA review. The regulatory review process involves detailed scrutiny of clinical data, manufacturing practices, and labeling recommendations. Only upon satisfactorily passing through these stages can the biologic become FDA approved and available for clinical use.

Specific Requirements for TriTE Approval 
For a TriTE to earn FDA approval, it must meet not only the general biopharmaceutical standards but also clinical benchmarks that reflect the complexity of its multi-specific mechanism. Specific requirements include: 
- Demonstrated Safety and Tolerability: Given the potent T-cell activating capacity, the drug must show an acceptable toxicity profile, with controlled cytokine release and avoidance of immune-mediated adverse events such as neurotoxicity or severe off-tumor activity. 
- Efficacy in Target Populations: The TriTE must show statistically significant clinical benefits over existing treatments, especially in its targeted indications, whether these are hematologic malignancies or solid tumors. Clinical endpoints might include objective response rates, progression-free survival, or overall survival improvements. 
- Manufacturing Consistency: Production of a complex molecule like a TriTE requires robust manufacturing protocols ensuring batch-to-batch consistency, proper folding, and bioactivity of all three binding domains. The FDA evaluates these parameters carefully under Good Manufacturing Practice (GMP) conditions. 
- Pharmacokinetic and Pharmacodynamic Profiles: The extended half-life enhancements observed in second-generation T-cell engagers must be substantiated with data showing that dosing regimens are practical for patient use without continuous infusion requirements—a challenge noted with earlier BiTE molecules. 
- Risk Mitigation Strategies: Post-market surveillance plans and risk mitigation strategies are necessary to manage any emerging adverse events related to multispecific engagement, especially given the novel mechanisms involved.

Since TriTEs are still emerging in the translational pipeline, much of their current data comes from preclinical studies and early-phase clinical trials. This means they are yet to fully satisfy the long and rigorous FDA approval process which was first successfully navigated by simpler bispecific formats.

Current FDA Approved TriTEs

List and Description of Approved TriTEs 
Based on the current evidence and the compiled references, there are no FDA approved Trispecific T-cell engagers (TriTEs) as of now. All of the tri-specific formats discussed in the literature—whether aimed at targeting dual tumor antigens or combining tumor targeting with costimulatory signals—remain in the preclinical or early clinical testing phases. 
- Preclinical and Experimental TriTEs: For example, reference discusses a novel tandem T-cell recruiting TriTE for the treatment of colorectal cancer, and reference outlines the construction of a tri-specific T cell engager targeting CD19, demonstrating promising in vitro and in vivo activity. However, none of these have received FDA approval yet because such advanced candidates are still undergoing safety and efficacy evaluations in clinical trials. 
- Comparison with Approved BiTEs: To date, the only T cell engagers that have moved through the FDA approval process successfully are bispecific molecules such as blinatumomab and the more recent CD20 X CD3 and BCMA X CD3 molecules (mosunetuzumab, epcoritamab, and teclistamab) approved in 2014, 2022, and 2023 respectively. These molecules operate via a bispecific mode of action rather than the more complex trispecific engagement.

Thus, when examining the current landscape of FDA approvals and referencing structured reports from synapse, there is clear evidence that while tri-specific formats are a hot topic in current research, the FDA-approved T-cell engagers in routine clinical use are exclusively bispecific in nature. No TriTE has, as of now, passed through the entire regulatory process to obtain FDA approval.

Clinical Indications and Applications 
FDA-approved T-cell engagers, being primarily bispecific, are currently indicated for hematologic malignancies such as B-cell acute lymphoblastic leukemia (ALL), non-Hodgkin’s lymphoma (NHL), and multiple myeloma. Their design leverages the ability to rapidly engage T cells in patients and induce potent cytolytic activity against cancer cells that express the target antigen. However, the clinical applications of TriTEs—once approved—are projected to extend into realms where dual antigen targeting could reduce the risk of tumor antigen escape and enhance specificity, for example: 
- Solid Tumors: Given the heterogeneity and immune-suppressive microenvironments often observed in solid tumors, dual or triple targeting may improve the precision with which T cells home in on malignant cells while sparing normal tissues. 
- Hematologic Malignancies with High Immune Evasion: TriTE designs might be particularly beneficial in diseases like acute myeloid leukemia (AML) or certain lymphomas where the tumor cells can modulate immune checkpoint expression, hence the additional binding domain could be useful for enhancing T-cell activation or costimulatory signaling.

In clinical practice, if a TriTE were to be FDA approved, it would likely be accompanied by detailed population-specific studies establishing its safety and efficacy in comparison or in synergy with existing immunotherapies. Presently, the clinical indications for T-cell engaging therapies remain dominated by their bispecific counterparts, and TriTEs are still positioned at the cutting edge of translational research with significant potential for future therapeutic application.

Challenges and Future Prospects

Current Challenges in TriTE Development 
The transition from promising preclinical data to FDA approval for complex multispecific biologics like TriTEs is fraught with technical and clinical challenges. Among the primary hurdles are: 
- Molecular Complexity and Production: TriTEs incorporate three functional binding domains, which increases the complexity of protein folding, stability, and batch consistency. Manufacturing processes must ensure that all three domains maintain their specific binding activity and that the therapeutic molecule remains stable in vivo. Inefficiencies or variability at the production stage can impede clinical translation and regulatory approval. 
- Safety Concerns: With the addition of a third binding module, there is an increased risk of off-target effects and unexpected toxicity. For example, more robust T-cell activation can lead to cytokine release syndrome (CRS) or neurotoxicity if not carefully controlled in clinical settings. 
- Optimizing Pharmacokinetics: Earlier TCEs like BiTEs faced challenges with short half-lives, necessitating continuous intravenous infusions. Although modifications (such as Fc fusion or PEGylation) have improved the half-life of subsequent constructs, similar optimizations must be made for TriTEs without compromising their function. The longer half-life is critical for clinical usability and patient compliance. 
- Efficacy in Heterogeneous Tumor Microenvironments: Tumor cells can demonstrate significant antigen heterogeneity. While TriTEs aim to counteract antigen escape by targeting multiple TAAs or combining costimulatory signals, the diversity and dynamic nature of tumor microenvironments continue to be a challenge. Efficacy in real-world conditions must be rigorously established across patient populations. 
- Regulatory and Clinical Trial Design: Given that TriTEs are a relatively new class of therapeutics, designing clinical trials that adequately capture both safety and efficacy endpoints becomes challenging. Regulatory agencies require robust data that demonstrate clear benefits over existing therapies. These challenges slow the process of clinical development and eventual FDA approval.

Future Directions in TriTE Research 
Looking ahead, TriTEs represent a next-generation strategy with tremendous potential. Future research directions include: 
- Refinement of Molecular Design: Researchers are continuously exploring novel formats to optimize the binding affinity, stability, and functional synergy of the three binding domains. Innovative approaches such as nanobody-based TriTEs or modular designs that allow for flexible swapping of binding domains are being actively investigated. 
- Combination Therapeutic Strategies: Given the complex interplay between tumors and the immune system, there is significant interest in combining TriTEs with other immunotherapies. For instance, pairing TriTEs with immune checkpoint inhibitors or costimulatory agonists may enhance the overall antitumor response while mitigating the risk of immune tolerance or exhaustion. 
- Enhanced Preclinical Models and Clinical Trial Designs: Advanced preclinical models that better simulate human tumor microenvironments, including patient-derived xenografts and organoid-based systems, will be crucial to accurately predict the efficacy and safety of TriTEs. Moreover, inclusion of immune profiling and biomarkers in clinical trial designs will help identify patient subpopulations most likely to benefit from these therapies. 
- Addressing Safety via Conditional Activation: One novel approach involves designing TriTEs that are conditionally active only within the tumor microenvironment. This can be achieved by engineering molecular switches or incorporating protease-cleavable linkers that activate only in the presence of tumor-specific signals. Such strategies may reduce systemic toxicity and enhance the therapeutic window. 
- Broadening Therapeutic Applications: In addition to traditional hematologic malignancies, there is increasing interest in applying TriTEs to solid tumors that are traditionally resistant to T-cell based immunotherapies. The dual targeting of tumor antigens along with the provision of costimulatory signals may help overcome the immunosuppressive barriers typical of solid tumor microenvironments.

Although the pathway to FDA approval for TriTEs remains challenging, continued advancements in protein engineering, immuno-oncology, and clinical trial design are paving the way toward overcoming these obstacles. The current research suggests that while tri-specific T-cell engagers hold great promise, they have yet to complete the regulatory journey that has been successfully navigated by earlier bispecific T-cell engagers.

Conclusion 
In summary, based on the current body of literature and the structured information available from reliable sources such as synapse, there are presently no FDA approved trispecific T-cell engagers (TriTEs). The approved molecules in current clinical use are exclusively bispecific T-cell engagers such as blinatumomab, mosunetuzumab, epcoritamab, and teclistamab, which have achieved regulatory success for hematologic malignancies. TriTEs are still in the experimental and early clinical stages, being thoroughly investigated for their potential advantages over bispecific formats. 

From a general perspective, the remarkable evolution from bispecific to trispecific therapeutics reflects the continuous drive to enhance targeted immunotherapy by incorporating additional binding domains that potentially increase efficacy and overcome resistance mechanisms. Specifically, the added complexity of a TriTE could theoretically offer improved tumor specificity and provide enhanced costimulatory signals, thereby facilitating robust T-cell activation even in immune-suppressive environments. However, detailed preclinical studies and early-phase clinical trials to date have yet to produce sufficient evidence for FDA approval. 

Looking at the regulatory pathway, the advanced requirements necessary for the approval of a novel multi-specific biologic mean that TriTEs must demonstrate a superior benefit-to-risk ratio over existing therapies. Safety concerns, manufacturing challenges, and the need for an acceptable pharmacokinetic profile remain pivotal issues that researchers are actively addressing. The FDA approval process, defined by strict preclinical and clinical benchmark requirements, has successfully navigated simpler bispecific modalities — an achievement that sets a high bar for the more complex TriTEs.

In conclusion, while trispecific T-cell engagers represent an exciting frontier with significant potential to redefine immunotherapy options for both hematologic and solid tumors, as of now, no TriTE has achieved FDA approval. Future research directions remain promising, with efforts to optimize molecular design, reduce systemic toxicity, and improve clinical outcomes. As the field progresses and more robust early-phase data becomes available, it is anticipated that TriTEs may eventually join the clinical armamentarium, further advancing the fight against cancer.

The current status, therefore, is that the clinically available T-cell engaging therapies approved by the FDA are bispecific, and while work on TriTEs is progressing rapidly, they are not yet a part of approved immunotherapeutic agents. This conclusion highlights both the innovation in the field and the rigorous demands of the FDA approval process which currently preclude TriTEs from being utilized in routine clinical practice.

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.