What are the different types of drugs available for TriTAC?

17 March 2025
Introduction to TriTAC

Definition and Basic Concepts
TriTACs, or Tri-specific T cell Activating Constructs, represent an innovative class of engineered proteins that harness and redirect the patient’s own T-cells to recognize and kill tumor cells. Unlike traditional bispecific T-cell engagers (BiTEs), TriTACs are modular constructs that include three distinct binding domains. Typically, one domain binds to a specific antigen expressed on tumor cells, another domain binds to T cells (usually via the CD3 receptor), and a third domain is designed for half-life extension—often by binding transiently to human serum albumin. This modular design not only confers potent anti-tumor activity but also provides improved serum stability and potentially reduced off-target toxicities. In essence, TriTACs aim to combine the attractive features of small-molecule immune activators (such as the efficient killing at low antigen expression) with the favorable pharmacokinetic properties that are often challenging in smaller antibody fragments.

Overview of TriTAC Technology in Drug Development
In the field of immuno-oncology, the TriTAC platform is one example of how modular protein engineering is revolutionizing targeted therapies. The technology is focused on achieving efficacious T-cell engagement while overcoming key limitations associated with earlier generations of T-cell engagers—most notably the need for continuous intravenous infusion due to their short half-life. By integrating a half-life extending domain—often via binding to serum albumin—TriTAC molecules can maintain sufficient bioavailability and can be administered less frequently, thereby offering a significant improvement in clinical convenience and safety profile. The TriTAC design also enables the tailoring of its functional domains to suit different tumor targets, which is critical in addressing the diversity among solid tumors and hematologic malignancies. This approach underscores a broader movement in drug development toward highly engineered, precision medicines that act by bridging the body’s immune effector cells directly to the tumor cells while simultaneously mitigating adverse effects.

Types of TriTAC Drugs

Classification of TriTAC Drugs
Current developments in TriTAC technology have led to the emergence of three distinct platforms or “types” of TriTAC drugs, each designed with unique characteristics to address specific clinical challenges. Broadly, the classification includes:

1. Constitutively Active TriTAC:
This is the first and most direct form of the TriTAC product candidate. Constitutively active TriTACs are designed to remain in an active state upon administration. Their structure maximizes immediate T-cell activation by continuously engaging both the T-cell receptor and the tumor cell antigen simultaneously. Because these molecules do not require any in vivo conversion or activation steps, they tend to have robust on-target activity, which is ideal for tumors with limited on-target liabilities. However, the continuous state of activity also poses a risk for potential off-target toxicities if not carefully managed.

2. ProTriTAC:
ProTriTACs introduce an additional level of control by incorporating a “prodrug” concept into the TriTAC platform. In this design, the molecule is maintained in an inactive state until it reaches the tumor microenvironment. The spatial control is achieved by masking the active domains of the TriTAC until they are triggered by specific cues present in the tumor milieu, such as particular enzymes or pH conditions. This targeting strategy is intended to minimize on-target tissue damage by restricting activation of T-cell engagement predominantly to the tumor site. In effect, ProTriTAC drugs aim to reduce systemic toxicity and improve the therapeutic window, making them particularly useful for vulnerable or sensitive tissues.

3. TriTAC-XR (Extended Release TriTAC):
The TriTAC-XR platform represents a further refined approach that focuses on temporal control of drug activation. These extended-release formulations are designed such that the active TriTAC is gradually released in the systemic circulation at a predefined rate. The objective here is to mitigate cytokine release syndrome (CRS), a potentially dangerous side effect observed with rapid and high concentrations of T-cell engagers. By modulating the release kinetics, TriTAC-XR allows for a controlled exposure to the active molecule, reducing the peak (C_max) levels that are typically associated with severe CRS and other adverse events. This controlled activation is particularly advantageous for patients who might be at higher risk for systemic inflammatory responses.

Beyond the three fundamental platforms, TriTAC drug candidates are also frequently differentiated based on their target antigen profiles. For example, in the clinical pipeline, specific molecules have been designed to target various tumor-associated antigens such as:

- HPN424: Targeting Prostate Specific Membrane Antigen (PSMA) for metastatic castration-resistant prostate cancer.
- HPN536: Targeting mesothelin, which is relevant in ovarian and pancreatic cancers.
- HPN217: Targeting B Cell Maturation Antigen (BCMA) for the treatment of relapsed/refractory multiple myeloma.
- HPN328: Targeting DLL3, often associated with small cell lung cancer and other DLL3-expressing tumors.

This bifurcation into platform type and target specificity allows for a highly customizable therapeutic strategy. It ensures that the appropriate balancing of activation, safety, and pharmacokinetic properties is achieved while also tuning the molecular construct to engage specific tumor cell antigens and T lymphocyte subsets.

Mechanism of Action
Despite differences in platform design, the mechanism of action for all TriTAC drugs shares a common underlying principle: the formation of a ternary complex that bridges the T cell to the tumor cell. The general steps are as follows:

1. Simultaneous Binding:
- One domain of the TriTAC binds to a tumor-associated antigen on the cancer cell.
- A second domain engages the T-cell receptor complex (typically via CD3 on T cells), thereby recruiting the T cell into proximity with the target cell.
- The third domain binds to circulating human serum albumin. This binding extends the molecule’s half-life by taking advantage of albumin’s long circulatory residence time, leading to improved bioavailability.

2. Facilitation of T-cell Activation:
Once the ternary complex (tumor cell–TriTAC–T cell) is formed, the T cell becomes activated. The T cell then deploys its cytotoxic machinery—releasing perforin, granzymes, and other cytolytic proteins—to induce apoptosis in the targeted tumor cell. This process is not necessarily dependent on antigen presentation via major histocompatibility complex (MHC) molecules, which is a key advantage over traditional T-cell receptor engagement tactics.

3. Control of Activation in ProTriTAC and TriTAC-XR:
- In the case of ProTriTACs, the active binding sites of the molecule are masked until the construct encounters an activating factor in the tumor microenvironment. This spatial regulation ensures that T cells are activated locally rather than systemically, thereby reducing the risk of widespread immune-related toxicities.
- For TriTAC-XR formulations, the kinetic release properties are engineered such that after administration, the active drug is released slowly and steadily. This prevents the high plasma concentrations that might trigger high levels of cytokine release, thereby offering a safer profile especially for systemic applications.

4. Modular Adaptability:
TriTACs are designed in a modular fashion so that by substituting the tumor-binding domain, the same platform can be redirected against different cancer cell types. This modularity not only accelerates the development timeline for new indications but also allows for fine-tuning of the binding affinities and valencies, optimizing the efficacy-to-toxicity ratio.

Current Development and Clinical Trials

Approved TriTAC Drugs
At present, while the TriTAC technology has demonstrated significant promise in preclinical and early-phase clinical evaluations, there are no TriTAC drugs that have received full regulatory approval for commercial use. The first T-cell engager BiTE was approved in 2014 for acute lymphocytic leukemia, but TriTAC constructs represent a new generation of these therapies. Given their relatively recent introduction and innovative design, TriTACs are still in clinical development, with several candidates progressing through Phase I and Phase I/II trials. The absence of an approved label does not detract from the potential of these drugs; rather, it underscores the cutting-edge nature of their technology as they continue compared side-by-side in clinical environments with existing immune therapies.

TriTAC Drugs in Clinical Trials
A number of TriTAC candidates are currently in the clinical trial pipeline, with several promising candidates already being evaluated:

- HPN424:
This candidate targets the Prostate Specific Membrane Antigen (PSMA), which is present in a high percentage (approximately 85% to 90%) of patients with advanced metastatic prostate cancer. HPN424 is undergoing Phase I dose-escalation trials, with preliminary data indicating that it can activate T cells effectively and may exhibit a favorable pharmacokinetic profile that supports weekly intravenous administration. The construct is based on the constitutively active TriTAC platform.

- HPN536:
Targeting mesothelin, HPN536 is being evaluated primarily in the context of platinum-refractory ovarian cancer and, more recently, in metastatic pancreatic cancer patients. HPN536 is the first mesothelin-targeted T-cell engager to be developed and entered into clinical studies. The candidate is structured to provide potent T-cell activation while leveraging the prodrug aspects of the ProTriTAC design, thus ensuring that the drug mainly becomes active in the tumor microenvironment, potentially reducing systemic side effects.

- HPN217:
This candidate engages B Cell Maturation Antigen (BCMA), which is a key target in multiple myeloma. HPN217 is being assessed in a Phase I/II trial targeting patients with relapsed/refractory multiple myeloma. The design of this candidate leverages the TriTAC technology’s advantages in rapidly recruiting T cells to tumor cells while maintaining a manageable safety profile, thanks in part to the half-life extension mechanisms incorporated into the molecule.

- HPN328:
Targeting DLL3, HPN328 is designed for use in small cell lung cancer and other tumor types that are associated with DLL3 expression. DLL3 as a tumor antigen has been targeted by a limited number of T-cell engager technologies previously, and HPN328 is significant because it is the first TriTAC aimed at this target. This candidate’s clinical trial is structured as a Phase I/II study, wherein clinical endpoints will evaluate safety, target engagement, and initial signs of efficacy.

These candidates represent a spectrum of TriTAC drug types in development, highlighting the flexibility of the platform. The diversity in targets—from PSMA in prostate cancer to mesothelin in ovarian and pancreatic cancers, to BCMA in multiple myeloma and DLL3 in lung cancer—demonstrates how the TriTAC platform can be adapted to address varying tumor biology. Importantly, these studies are designed to explore not only the therapeutic activity, but also the pharmacokinetic and pharmacodynamic profiles that underscore the advantages of the TriTAC approach, such as extended half-life and improved safety margins compared to traditional T-cell engagers.

Challenges and Future Directions

Current Challenges in TriTAC Drug Development
Despite the significant promise inherent in TriTAC technology, several challenges remain that need to be addressed as these therapies move further along the clinical development pathway:

1. Off-Target Toxicity and Cytokine Release Syndrome (CRS):
One of the foremost challenges with T-cell engagers, including TriTACs, is the risk of off-target T-cell activation that can lead to systemic cytokine release syndrome—a potentially life-threatening condition. While the constitutively active TriTACs provide robust T-cell engagement, their continuous mode of action may increase the risk of CRS. This risk has driven the development of the TriTAC-XR platform, which seeks to mitigate these adverse effects by controlling the release rate of the active drug. However, balancing immediate efficacy with safety remains a critical hurdle.

2. Tumor Heterogeneity and Antigen Escape:
Tumors often exhibit heterogeneity in antigen expression, and there is a risk that a tumor may downregulate the targeted antigen after therapy begins. This antigen escape can reduce the efficacy of a TriTAC drug that is designed to engage a specific tumor marker. Multi-targeted or modular approaches that allow for rapid re-engineering of the tumor-binding domain are being considered to overcome this challenge.

3. Manufacturing and Stability:
As sophisticated protein-based biotherapeutics, TriTACs require precise manufacturing processes to ensure proper folding, stability, and quality control. Given their modular design and relatively small size (around 50 kilodaltons, approximately one-third the size of a conventional antibody), maintaining consistency during production is critical. Further, the integration of half-life extension domains and activation control mechanisms must be finely tuned to avoid loss of activity or unwanted immunogenicity.

4. Pharmacokinetic Challenges:
Even though the inclusion of albumin-binding domains has significantly improved the half-life of TriTAC constructs, achieving an optimal pharmacokinetic profile without compromising tissue penetration remains a balancing act. In solid tumors, effective tumor penetration is vital, and smaller proteins could theoretically diffuse more effectively than larger antibodies. However, the trade-off is that overly rapid diffusion might also increase off-target exposure. Thus, fine-tuning the pharmacokinetic parameters through controlled release formulations like TriTAC-XR continues to be a critical area of translational research.

5. Regulatory and Clinical Trial Design Considerations:
As with any emerging drug modality, the regulatory frameworks for TriTAC drugs are still evolving. Defining biomarkers of response, establishing dosing regimens, and designing bridging studies for different patient populations are all challenges that must be met to streamline clinical development. The design of early-phase clinical trials must adequately measure both safety and efficacy, particularly given the dual challenges of immunotherapy-related toxicities and tumor resistance mechanisms.

Future Prospects and Research Directions
Looking ahead, the future of TriTAC drug development appears promising, with several avenues of research and development likely to drive further innovation:

1. Improved Engineering for Enhanced Safety and Specificity:
Ongoing research is focused on further refining the design of TriTAC molecules to maximize tumor specificity and reduce the risk of systemic immune activation. Advances in protein engineering could lead to constructs that have improved conformational stability and binding specificity, thereby reducing the likelihood of off-target effects and cytokine-mediated toxicities. For instance, additional modifications to the ProTriTAC design could further restrict drug activation exclusively to the tumor microenvironment, increasing the therapeutic window.

2. Combination Therapies:
Given the complex immunobiology of cancer, it is increasingly likely that TriTAC drugs will be used in combination with other immunotherapies or targeted therapies. Combination regimens could synergize the effects of TriTAC-mediated T-cell activation with checkpoint inhibitors or other modalities that modulate the immune response, potentially overcoming issues like antigen escape and increasing overall efficacy. Early clinical trials may explore these combinations to identify regimens that maximize anti-tumor activity while minimizing toxicity.

3. Expanding Target Profiles:
The modular nature of TriTACs means that once the foundational platform is established, it can be adapted relatively quickly to target new tumor-associated antigens. Research is anticipated to continue exploring novel targets for which there is a strong scientific rationale but where classical antibody-based therapies have shown limitations. This flexibility is expected to lead to an expanding portfolio of TriTAC drugs addressing a broader range of solid tumors and hematologic malignancies.

4. Refinement of Extended-Release and Prodrug Strategies:
Future research may further improve the TriTAC-XR platform by refining the kinetics of drug release. By using a more sophisticated understanding of the interplay between drug concentration, T-cell activation, and cytokine release, developers can design formulations that optimize the timing and amount of active drug release. This could significantly reduce the incidence of adverse events such as CRS while maintaining potent anti-tumor activity. Enhanced prodrug technologies might also allow for better spatial control, ensuring that active drug molecules are released exclusively within the tumor microenvironment.

5. Biomarkers and Patient Stratification:
Future clinical trials are likely to incorporate more robust biomarker analyses to predict which patients are most likely to benefit from TriTAC therapy. This could include genomic, transcriptomic, and proteomic profiling to determine tumor antigen expression and immune cell infiltration. Stratifying patients based on these biomarkers will help in optimizing clinical trial design, selecting appropriate dosing regimens, and ultimately guiding individualized therapy. Improved biomarker-driven stratification is a critical component for enhancing the clinical success of any targeted immunotherapy.

6. Regulatory Pathway Evolution:
As more data emerge from ongoing clinical trials, regulatory bodies are expected to develop more precise guidelines tailored to complex biologics like TriTAC drugs. This regulatory evolution should help streamline the approval process by clarifying issues related to safety monitoring, pharmacokinetic modeling, and efficacy endpoints. Collaborative efforts between regulators, industry, and academic researchers will be essential to establishing best practices that can accelerate the clinical translation of TriTAC technology.

7. Manufacturing Innovations:
To meet the growing demand for TriTAC products, advancements in manufacturing processes will be crucial. Innovations in bioprocessing, quality control, and scale-up procedures will help ensure that these complex molecules can be reliably produced in a cost-effective manner without compromising their functional integrity. This will be particularly important as TriTAC drugs move from early-stage trials into broader clinical applications.

Conclusion
In summary, TriTAC drugs represent a groundbreaking advancement in immunotherapy that leverages a tri-specific design to harness and direct T-cell mediated cytotoxicity against tumor cells. They are classified into three main types based on their platform design: constitutively active TriTACs provide immediate T-cell engagement but with the risk of off-target toxicities; ProTriTACs incorporate a prodrug approach to achieve spatial control of activation, limiting systemic exposure and reducing toxicity; and TriTAC-XR formulations offer extended-release kinetics to further mitigate peaks in active drug concentration and reduce cytokine release syndrome.

From a mechanistic perspective, all TriTACs operate by forming a ternary complex—bridging the tumor cell, the T cell, and a half-life extending moiety such as albumin—which results in targeted T-cell activation and subsequent tumor cell lysis. This modular and adaptable design not only allows for rapid re-targeting to different tumor antigens but also provides opportunities to optimize safety and pharmacokinetic profiles.

Current clinical development features several promising candidates such as HPN424, HPN536, HPN217, and HPN328, each tailored to specific tumor-associated antigens like PSMA, mesothelin, BCMA, and DLL3. Although none of these candidates have yet received regulatory approval, they are advancing through Phase I and Phase I/II clinical trials, demonstrating preliminary signs of efficacy and manageable safety profiles. Importantly, the emerging clinical data, combined with advanced engineering and manufacturing technologies, promise to address several of the inherent challenges in T-cell engager development.

Looking forward, the future prospects for TriTAC drugs are robust, with ongoing research focused on improving specificity, minimizing toxicity, enhancing pharmacokinetic control, and exploring combination therapies to overcome tumor heterogeneity and antigen escape. Additional efforts in biomarker development, regulatory pathway refinement, and manufacturing innovation are expected to further facilitate the clinical translation and broad application of TriTAC drugs.

In conclusion, the evolution of TriTAC technology exemplifies the rapid innovation in the field of immuno-oncology. By overcoming the limitations of earlier T-cell engager platforms, TriTACs have the potential to provide more effective, safer, and more versatile treatments for multiple cancer types. Their ability to be rapidly adapted to various tumor antigens, combined with advanced prodrug and extended-release strategies, positions them as a promising frontier for targeted cancer therapy. As the clinical and manufacturing challenges are addressed through ongoing research and development, TriTAC drugs are poised to play a significant role in the future landscape of cancer treatment, offering new hope to patients with previously difficult-to-treat malignancies.

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.