For what indications are Trispecific T-cell engager (TriTE) being investigated?

17 March 2025
Introduction to Trispecific T-cell Engagers (TriTEs)

Definition and Mechanism of Action
Trispecific T-cell engagers (TriTEs) are advanced engineered antibody constructs designed to simultaneously bind to three distinct antigens. Typically, one part of the molecule binds to a T-cell antigen such as CD3 (sometimes combined with a costimulatory molecule like CD28), while the other two binding domains are directed against two separate tumor-associated antigens. This structure permits the formation of a bridge between T cells and cancer cells, thereby eliciting potent T-cell activation and consequent cytotoxicity toward target tumor cells. Their unique design not only fosters direct tumor cell lysis but also amplifies signaling through costimulatory engagement, which can improve T-cell activation even in immunosuppressive microenvironments.

Overview of T-cell Engagers in Immunotherapy
T-cell engagers, historically represented by bispecific T-cell engagers (BiTEs) such as blinatumomab, have significantly altered the landscape of immunotherapy by redirecting a patient’s own cytotoxic T cells to specifically target malignant cells. As research has advanced, the scope of these molecules has expanded into multispecific formats like TriTEs, which integrate additional targeting or modulatory functions into a single construct. By harnessing the inherent specificity and potency of T cells, TriTEs are emerging as promising next-generation immunotherapeutic agents that might overcome some of the limitations observed with earlier bispecific configurations, such as cytokine release syndrome (CRS) and “off‐tumor” toxicity. These new constructs are designed to enhance the overall therapeutic index and target a broader array of cancer indications.

Current Research on TriTEs

Indications Under Investigation
The current research across numerous studies and patents indicates that TriTEs are being evaluated for a wide spectrum of indications, spanning both hematologic malignancies and solid tumors.

1. Hematologic Malignancies
- Acute Lymphoblastic Leukemia (ALL) and B-cell Malignancies:
In one study, a novel trispecific T-cell engager was constructed to target CD19—an antigen widely expressed on B-lineage cells—and demonstrated significant cytotoxicity against CD19-positive tumor cell lines. This investigation incorporates the concept of redirecting T cells specifically to leukemia cells by bridging CD3 on T cells and CD19 on malignant cells.
- Acute Myeloid Leukemia (AML) and Myelodysplastic Syndromes (MDS):
Other reports have highlighted trispecific formats that combine targeting of antigens such as CD33—which is found on AML blasts—with T-cell receptors. Experimental data showed that these constructs not only bound specifically to the leukemic cells but also induced robust T-cell proliferation and enhanced cytotoxicity, even in the presence of high PD-L1 expression.
- Multiple Myeloma (MM):
TriTEs that incorporate binding modules directed at antigens such as CD38, and costimulatory molecules like CD28, are also under active investigation for multiple myeloma. For instance, one study leveraged a trispecific antibody format binding CD3, CD28, and CD38. This design is intended to overcome the typical bell-shaped dose response observed with bispecific formats and enhance T-cell mediated killing at clinically relevant doses.
- Non-Hodgkin’s Lymphoma (NHL):
Although less common in early reports, the modularity of TriTE constructs suggests applications in other B-cell lymphomas as well. The design principles illustrated in early-phase studies indicate potential use in broader lymphoid malignancies where targeting CD19 or other lineage-specific markers alongside T-cell activation can elicit significant anti-tumor responses.

2. Solid Tumors
- Colorectal Cancer (CRC):
A notable study described the development of a TriTE that simultaneously targets two tumor-associated antigens, namely epidermal growth factor receptor (EGFR) and epithelial cell adhesion molecule (EpCAM), in addition to CD3 on T cells. This construct displayed a 100-fold increased potency against double-positive colorectal cancer cells versus single-positive targets. This dual tumor-targeting approach using a trispecific format seeks to address the antigen heterogeneity commonly observed in solid tumors, thereby reducing the chance of tumor escape due to antigen loss.
- Ovarian and Pancreatic Cancers:
In preclinical trial data, companies like Harpoon Therapeutics have utilized proprietary platforms (e.g., TriTAC®) to develop trispecific constructs. One candidate targets mesothelin, a frequently overexpressed antigen in ovarian and pancreatic cancers, by engaging both T cells and mesothelin-positive tumor cells. Early-phase clinical trials are underway, with indications suggesting that these constructs could offer a novel therapy for mesothelin-expressing solid tumors.
- Small Cell Lung Cancer (SCLC) and DLL3-Associated Tumors:
Another target in the solid tumor realm is DLL3, often overexpressed in small cell lung cancer. TriTEs targeting DLL3 are currently being studied in Phase 1/2 trials, offering a promising approach for tumors that have limited treatment options due to their aggressive nature and antigenic profile.
- Prostate Cancer:
Although more commonly targeted by other immunotherapeutic approaches like CAR-T cells and checkpoint inhibitors, indications from recent news releases suggest that platforms such as TriTAC® are being applied to prostate cancer, particularly metastatic castration-resistant prostate cancer (mCRPC) by targeting the prostate-specific membrane antigen (PSMA).
- Broader Applications in Solid Tumor Microenvironments:
Additional studies have explored the modification of TriTEs through integration with novel modalities such as nanobody-based formats (Nb-TriTE). These are designed specifically to overcome the immunosuppressive tumor microenvironment prevalent in many solid tumors by coupling T-cell engagement with immune checkpoint modulation (e.g., anti-PD-1). This suggests that TriTEs could be used as a universal platform for a variety of challenging solid tumor indications where immune escape is a key barrier.

3. Other Emerging Indications
- Combination Immunotherapeutics:
There is a growing interest in using TriTEs in combination with other immunotherapies to improve efficacy while mitigating undue toxicity. For example, integrating TriTE modalities with checkpoint inhibitors or adoptive T-cell therapies (such as CAR-T cells) is an avenue currently under investigation to harness synergistic effects, particularly in refractory cancer cases.
- Overcoming Antigen Heterogeneity and Immunosuppression:
In addition to directly targeting tumor antigens, some TriTE designs aim to address the tumor heterogeneity issue by including two separate tumor antigen-binding domains, thus providing a broader coverage against variant tumor cell populations. This dual targeting is particularly attractive in heterogeneous solid tumors like colorectal cancer, where antigen loss and tumor escape mechanisms are common.
- T-cell Engager-Armed Oncolytic Viruses:
Although not strictly a TriTE per se, emerging research has investigated the integration of T-cell engager functionalities into oncolytic virus platforms. This next-generation strategy combines the direct tumor-lytic effects of oncolytic viruses with the immunomodulatory potential of trispecific antibodies. Such a strategy holds promise for a broad range of tumor types, both hematologic and solid, by simultaneously stimulating an immune response and directly lysing tumor cells.

Preclinical and Clinical Trial Data
Preclinical models have consistently demonstrated that TriTEs can achieve enhanced potency and selectivity compared to their bispecific counterparts. For instance, in colorectal cancer models, the bispecific engagement of tumor markers like EGFR and EpCAM by a TriTE not only potentiated T-cell cytotoxicity but also led to a dramatic prolongation of survival in animal models. These studies utilized sophisticated in vitro assays and in vivo murine models to establish the kinetics of T-cell activation, receptor occupancy, and tumor cell killing.

In addition to solid tumor models, advances in hematologic malignancy models have shown that TriTEs targeting CD3, CD28, and CD38 can bypass the typical bell-shaped dose-response observed with bispecific constructs. Quantitative systems pharmacology (QSP) models have been employed to predict that the inclusion of a CD28-binding arm yields a plateau in the effective killing capacity at higher doses, suggesting a wider therapeutic index. Furthermore, clinical trials with TriTE constructs based on the Harpoon TriTAC® platform are in early stages. These studies are evaluating candidates such as HPN217 (a BCMA-targeted TriTAC) for multiple myeloma and HPN328 (a DLL3-targeted TriTAC) for small cell lung cancer, with reported Phase 1/2 data demonstrating safety profiles that are supportive of further dose-escalation studies.

The preclinical evaluations have also extended to nanobody-based TriTEs (Nb-TriTEs), where the smaller size of nanobodies allows for better tissue penetration and may lead to enhanced efficacy in the solid tumor microenvironment. In these studies, Nb-TriTE constructs targeting fibroblast activation protein (FAP) and incorporating an anti-PD-1 module induced robust tumor antigen-specific killing, enhanced T-cell activation, and led to improved survival in mouse xenograft models of solid tumors. Clinical data, although still emerging, indicate that these platforms might safely overcome immunosuppression without triggering excessive systemic cytokine release.

Potential and Challenges of TriTEs

Therapeutic Potential in Various Diseases
TriTEs show considerable promise in transforming cancer immunotherapy. Their design allows for increased selectivity by requiring the simultaneous engagement of two tumor-associated antigens. This dual antigen recognition minimizes off-tumor effects and ensures that T-cell activation is predominantly confined to the tumor microenvironment. Furthermore, the addition of a costimulatory arm like CD28 or the inclusion of cytokine-binding domains in some designs (for instance, an IL-6R-targeted domain to mitigate cytokine release syndrome) provides an extra layer of safety and efficacy.

From a therapeutic standpoint, the benefits of TriTEs are manifold:
- Enhanced Tumor Selectivity: The simultaneous targeting of two tumor antigens theoretically reduces the “on-target off-tumor” toxicity, which is especially critical in solid tumors that often express low levels of target antigens in normal tissues.
- Overcoming Tumor Heterogeneity: Many solid tumors, such as colorectal cancer, demonstrate antigen heterogeneity. TriTEs can be designed to target two different antigens, thereby addressing the challenge of tumor escape due to antigen loss.
- Improved T-cell Activation: The incorporation of additional binding sites, particularly costimulatory molecules, can enhance T-cell proliferation, activation, and persistence. This aspect is crucial when targeting immunosuppressive microenvironments often seen in solid tumors like ovarian, pancreatic, and lung cancers.
- Flexibility in Modifying Immune Response: TriTE platforms can be integrated with other therapeutic strategies, such as checkpoint inhibition or oncolytic virotherapy. This combinatorial potential can lead to improved clinical outcomes, especially in tumors refractory to single-agent therapies.

Challenges in Development and Application
Despite their promise, the development and clinical application of TriTEs are not without challenges. A key challenge concerns the delicate balance between achieving potent tumor cell lysis and avoiding systemic toxicity. For example, clinical application of T-cell engagers has historically been associated with cytokine release syndrome (CRS) and other immune-related adverse events. Even though TriTEs aim to mitigate these risks by incorporating costimulatory molecules or IL-6R-binding modules, the risk of immune overstimulation remains an ongoing concern that needs careful dose-finding and patient monitoring.

Other challenges include:
- Manufacturing Complexity: The engineering of trispecific constructs is inherently more complex than bispecific antibodies. Ensuring proper folding, stability, and manufacturing reproducibility is critical for clinical translation and scalability. Emerging patent literature stresses the importance of advanced protein engineering and manufacturing processes to maintain consistency and purity of these molecules.
- Pharmacokinetics and Biodistribution: TriTEs, due to their larger molecular structure compared to traditional BiTEs, might demonstrate different pharmacokinetic properties such as half-life and tissue penetration. The optimization of these parameters is vital to achieve the desired therapeutic index without compromising safety.
- Immunogenicity and Tolerance: As with any engineered biologic, there is always the potential for immunogenicity. The immune system might recognize these novel constructs as foreign, which could lead to the generation of anti-drug antibodies that neutralize the therapeutic effects of the TriTE while also possibly causing adverse reactions.
- Clinical Trial Design: The novel mechanism of action of TriTEs demands that clinical trial designs be adapted to capture not only traditional endpoints such as tumor shrinkage or overall survival but also measures of T-cell activation, cytokine profiles, and immune modulation. Early-phase trials must therefore incorporate comprehensive biomarker assessments to characterize the therapeutic window.

Future Directions and Innovations

Emerging Trends in TriTE Research
As the field advances, several emerging trends are shaping the future development of TriTEs. One significant trend is the move toward incorporating modular design features that allow for on-demand modifications of the T-cell engager. For instance, nanobody-based platforms are being explored to improve tissue penetration and reduce molecular weight while preserving efficacy. Additionally, innovative formats that incorporate built-in safety measures such as attenuated activation domains aim to limit the risk of CRS without sacrificing antitumor potency.

Another trend is the integration of computational modeling and quantitative systems pharmacology (QSP). This approach allows researchers to predict dose-response relationships and optimize dosing regimens before clinical implementation. Models have already shown promise in predicting that trispecific antibodies with a CD28 arm achieve a plateau in cytotoxicity at higher doses, which points to a broader therapeutic window compared to bispecific constructs. This trend not only assists in the rational design of TriTEs but also helps in understanding the complex in vivo interactions and guides patient-specific treatment strategies.

Moreover, there is growing interest in combining TriTEs with other immunotherapies. Early clinical and preclinical studies are exploring synergistic effects when TriTEs are used in conjunction with checkpoint inhibitors or adoptive cell therapies such as CAR-T cells. Such combination strategies may further enhance efficacy by counteracting the immunosuppressive microenvironment, which is particularly prevalent in solid tumors such as ovarian, pancreatic, and colorectal cancers.

Future Prospects in Immunotherapy
Looking ahead, TriTEs are poised to play a critical role in next-generation immunotherapy. Their ability to target two tumor-associated antigens simultaneously means that they could potentially address the issue of tumor antigen heterogeneity, a challenge that has limited the efficacy of many current immunotherapies. In hematologic malignancies, this implies that resistant disease due to antigen modulation or loss could be better managed with a dual-targeting approach.

In the realm of solid tumors, TriTEs offer hope by directly addressing the challenges of immunosuppressive tumor microenvironments through enhanced T-cell activation and multifunctional engagement of tumor cells. For example, the targeting of DLL3 in small cell lung cancer represents an innovative approach for a notoriously difficult-to-treat tumor type, while simultaneous engagement of mesothelin in ovarian and pancreatic cancers broadens the clinical applicability of TriTE constructs.

Furthermore, the versatility of TriTE constructs paves the way for personalized immunotherapy regimens. By tuning the affinities of the individual binding domains and adjusting the costimulatory signals, these agents can be tailored to the specific antigen expression patterns and immune status of individual patients. Such a strategy has the potential to maximize therapeutic efficacy while minimizing adverse effects—a critical consideration in the treatment of both hematologic cancers and a diverse array of solid tumors.

The clinical development of TriTEs is rapidly advancing as more first-in-human studies are initiated. Early-phase clinical trials are providing valuable data on safety, pharmacokinetics, and efficacy. For example, Phase 1/2 studies involving TriTAC platforms report promising signs of disease stabilization and measurable anti-tumor responses while avoiding severe CRS typically associated with T-cell engagers. As these trials progress, long-term outcomes, including overall survival and durable remission rates, will help clarify the role that TriTEs may play in the standard-of-care treatment paradigm for various malignancies.

Another exciting avenue is the use of TriTEs in combination with oncolytic viruses. This strategy leverages the direct tumor-lytic capability of oncolytic viruses while simultaneously engaging T cells to sustain long-term tumor control through immune surveillance. Preclinical data support the potential for such combinatorial approaches, and early translational research is beginning to explore these models. This dual modality approach might represent a paradigm shift in immunotherapy, particularly for tumors that have been refractory to conventional treatments.

Finally, the field is also witnessing innovations in manufacturing and delivery. New techniques in protein engineering and formulation are being developed to improve the stability, solubility, and in vivo half-life of these complex constructs. Such advances are critical not only for ensuring the reproducibility of clinical results but also for making these therapies accessible on a larger scale. The evolution of manufacturing platforms, as highlighted in several patents, is expected to reduce production costs and improve the overall feasibility of TriTEs as therapeutic agents.

Conclusion
In summary, trispecific T-cell engagers (TriTEs) are being investigated for a diverse range of indications across both hematologic malignancies and solid tumors. In hematologic malignancies, TriTEs have been evaluated for the treatment of acute lymphoblastic leukemia, acute myeloid leukemia, myelodysplastic syndromes, multiple myeloma, and potentially other B-cell lymphomas by targeting antigens such as CD19, CD33, CD38, and engaging T-cell antigens like CD3 and CD28. In the realm of solid tumors, considerable research is focused on colorectal cancer, where dual targeting of EGFR and EpCAM yields significant improvements in cytotoxic efficacy; ovarian and pancreatic cancers through targeting mesothelin; small cell lung cancer via DLL3; and even prostate cancer through PSMA targeting are being actively investigated.

The preclinical studies have provided robust evidence that TriTEs can overcome issues of tumor heterogeneity and immunosuppression by enabling dual antigen targeting with potent T-cell activation. QSP models and multiscale simulations have contributed to understanding dose-response relationships and identifying key metrics such as “effective receptor occupancy,” which further guides the clinical development of these agents. Moreover, early-phase clinical trials have reported encouraging safety and tolerability data, with some studies demonstrating the potential for enhanced therapeutic activity without excessive cytokine release syndrome.

While the potential of TriTEs is immense, challenges remain in balancing efficacy with safety, optimizing manufacturing processes, and addressing pharmacokinetic issues. The complexity of engineering trispecific molecules demands rigorous design and production strategies to ensure reproducible and safe products. Innovative approaches, such as nanobody-based TriTE formats and integration with oncolytic virus platforms, are being explored to further expand their therapeutic window and overall clinical utility.

From a future perspective, the promise of TriTEs lies in their inherent modularity, which allows for customization of targets based on the specific antigenic landscape of different tumors. The ability to combine T-cell engagement with additional immune checkpoint modulation or costimulatory signaling creates a robust platform for next-generation immunotherapies that may offer superior efficacy compared to current approaches. Combining TriTEs with other therapeutic modalities, such as CAR-T cells, checkpoint inhibitors, or even traditional chemotherapies, remains an exciting avenue for future investigation and could pave the way for more personalized and adaptive cancer treatment strategies.

In conclusion, the current body of literature and ongoing clinical research underscore that TriTEs are being actively investigated for an array of indications from hematologic malignancies—including ALL, AML, MDS, MM, and lymphomas—to various solid tumors such as colorectal, ovarian, pancreatic, lung, and prostate cancers. These agents represent a significant evolution in the field of T-cell engagers, with the potential to enhance clinical outcomes through improved tumor selectivity, overcoming antigen heterogeneity, and mitigating adverse immune events. As more clinical data become available and as innovations in design and delivery continue to evolve, TriTEs are likely to become a cornerstone in the expanding repertoire of immunotherapeutic strategies, ultimately offering hope for patients with cancers that are currently challenging to treat.

By addressing multiple targets simultaneously and incorporating costimulatory functions, TriTEs not only boost T-cell mediated cytotoxicity but also hold the promise of a broader therapeutic index with reduced off-target effects. This integrated approach—from preclinical design through early-phase clinical trials—demonstrates their versatile potential and lays the groundwork for future combination therapies that may significantly advance the field of immunotherapy in oncology and beyond.

The detailed preclinical and clinical evidence, combined with emerging trends in protein engineering and immunomodulation, provides a comprehensive picture of how TriTEs could soon transition from experimental therapeutics to mainstream clinical applications. Their ability to recruit and activate T cells in a specific and controlled manner, along with their innovative dual-target approach, positions them as a highly promising strategy in the next generation of cancer immunotherapy.

Ultimately, while challenges in manufacturing, immunogenicity, and precise dosing remain, the future of TriTE research appears bright. Continued advancements in both molecular design and clinical evaluation strategies are expected to address these hurdles, facilitating the successful translation of TriTEs into effective treatments for a wide range of cancer indications. The ongoing collaboration between academic researchers, industry partners, and regulatory bodies will be critical to harnessing the full potential of these multi-specific agents and ensuring that their promise is effectively realized in clinical practice.

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