For what indications are Trispecific killer cell engager (TriKE) being investigated?

17 March 2025
Introduction to Trispecific Killer Cell Engagers (TriKEs)

Tri-specific killer cell engagers (TriKEs) are an emerging class of immunotherapeutic molecules designed to harness the potent cytotoxic abilities of natural killer (NK) cells. These engineered protein constructs bind simultaneously to three different targets in a single molecular entity, thereby orchestrating a more robust and directed antitumor immune response. In their classic design, one of the binding arms engages the CD16 receptor on NK cells, which is critical for antibody‐dependent cell‐mediated cytotoxicity (ADCC), while the other binding domains recognize tumor-associated antigens on cancer cells. A third functional domain, often incorporating the cytokine interleukin-15 (IL-15), is included to stimulate NK cell proliferation and persistence in vivo. This multi-component assembly not only directs NK cell cytotoxic capacity toward malignant targets but also ensures improved survival and expansion of the effector cell population, overcoming some limitations noted in earlier NK-based immunotherapy formats.

Definition and Mechanism of Action

TriKEs are designed to redirect NK cells to specifically eliminate tumor cells by creating an immune synapse where cytotoxic signaling is strongly triggered. The binding of the anti-CD16 component to NK cells results in the activation of these cells by mimicking natural antibody Fc receptor interactions; simultaneously, if the tumor-associated antigen is present on a cancer cell, the other targeting moiety binds it, bridging the NK cell and the tumor cell in close proximity. The integrated IL-15 moiety plays a dual role: it not only enhances the activation signal through autocrine and paracrine interactions but also leads to the expansion and prolonged survival of NK cells in the circulation. Such a mechanism is particularly attractive as it bypasses some of the major barriers associated with conventional antibody therapies, such as the need for prolonged dosing and limitations in achieving sustained immune activity.

Overview of TriKEs in Immunotherapy

Over the past decade, immunotherapy has revolutionized cancer treatment, and TriKEs represent one of the newest frontiers in this field. Unlike bispecific killer cell engagers (BiKEs) that combine engagement of NK cells with a single tumor antigen recognition, TriKEs add a third arm that actively supports NK cell expansion through IL-15 signaling. This feature is critical in the hostile tumor microenvironment, where immunosuppressive factors can dampen NK cell function. By promoting in vivo NK cell homeostasis, TriKEs not only facilitate targeted cytotoxicity but also sustain a durable immune response against tumor cells. Their modular design allows for rapid engineering to target a variety of tumor antigens, thus offering a versatile platform for the treatment of both hematologic malignancies and solid tumors.

Investigational Indications for TriKEs

TriKEs are being investigated for a broad range of malignancies. Their versatility is underscored by the fact that they have been tailored to target antigens expressed in various cancers, thereby addressing both hematologic and solid tumor indications. The dual functionality of these molecules—simultaneously engaging NK cells and delivering IL-15 support—has opened opportunities in several clinical domains.

Hematologic Malignancies

In the context of blood cancers, TriKEs are being intensively studied to overcome the challenges associated with relapsed or refractory diseases. One of the major areas of investigation is acute myeloid leukemia (AML). AML cells generally express myeloid antigens, such as CD33, and numerous TriKE constructs have been engineered with anti-CD16 and anti-CD33 binding regions coupled with IL-15 to specifically activate NK cells against CD33+ leukemic cells. For instance, the CD16 × IL-15 × CD33 TriKE (commonly referred to as GTB-3550) has been evaluated in preclinical models of AML and myelodysplastic syndromes (MDS). These studies have demonstrated that TriKE-based therapy not only increases NK cell proliferation but also leads to potent NK cell-mediated killing of AML blasts in vitro, with promising in vivo data in xenograft models.

Moreover, TriKEs targeting other antigens in hematologic malignancies are under investigation. A variant aimed at targeting CLEC12A, which is expressed on leukemic stem cells in AML, has yielded encouraging evidence that such targeting can spare normal hematopoietic progenitors while eliminating leukemic cells. In addition to AML, other hematologic indications such as B-cell lymphoid malignancies are also being explored using TriKE constructs. For example, TriKEs incorporating anti-CD19 and even dual antigens (such as anti-CD19 and anti-CD22) are studied in order to overcome relapse driven by antigen escape—a common problem in treatments using bispecific molecules or CAR-T therapies.

The clinical rationale behind pursuing TriKEs in hematologic malignancies lies in their ability to both activate and expand NK cells. Compared with the use of bispecific formats alone, the integration of the IL-15 element supports long-term remissions by maintaining a robust NK cell presence. This is particularly important in diseases like AML and MDS, where the tumor microenvironment is often immunosuppressive and where conventional chemotherapies have limited efficacy upon relapse.

Solid Tumors

Solid tumors present a unique set of challenges to immunotherapy, including physical barriers, heterogeneous antigen expression, and a highly immunosuppressive tumor microenvironment. Despite these challenges, TriKEs are being developed for several solid tumor indications with encouraging preclinical data.

One of the early successes reported in the literature involves a TriKE targeting B7-H3, a costimulatory molecule highly expressed on several solid tumors, including ovarian cancer. In one study, an engineered B7-H3-targeted TriKE demonstrated that NK cells could be specifically expanded and activated to recognize and kill B7-H3+ tumor cells in vitro. Moreover, xenogeneic mouse models confirmed that this approach resulted in significant control of ovarian cancer growth, thereby suggesting a promising therapeutic angle for otherwise difficult-to-treat cancers.

Additional TriKE constructs have been tailored to target HER2, an antigen overexpressed in certain breast and ovarian cancers. The CAM1615HER2 TriKE, for instance, has been tested in both in vitro and in vivo models, where it effectively promoted NK cell expansion and enhanced cytotoxicity against HER2-positive cancer cells. This is particularly relevant in malignancies such as HER2-positive breast cancer or ovarian cancer, where traditional HER2-targeting antibodies have had limited success, thereby opening up the possibility of using TriKEs to potentiate NK cell-mediated immune responses and potentially improve patient outcomes.

Moreover, TriKEs are being investigated in the context of prostate cancer. A study presented at the Society for Immunotherapy of Cancer (SITC) demonstrated that TriKEs targeting prostate-specific antigens, including PSMA and B7H3, enhanced both the degranulation and cytotoxic responses of NK cells derived from both healthy donors and prostate cancer patients. The data indicated that TriKE treatment could overcome resistance even under conditions such as enzalutamide resistance and hypoxia, thereby underscoring the potential of these agents in a tumor type that traditionally presents with therapy-resistant features.

Beyond these specific examples, investigations are ongoing to evaluate TriKEs for other types of solid tumors as well. For instance, studies are examining their efficacy in lung cancer, head and neck squamous cell carcinoma (HNSCC), and even in rare solid tumor indications where conventional therapies fail. The flexibility of the TriKE platform in incorporating different tumor-specific binding domains means that new constructs can be rapidly developed and optimized to address the antigenic landscape of various solid malignancies.

Clinical Trials and Studies

The clinical development of TriKEs is underway, with several ongoing early-phase clinical trials and encouraging initial results emerging from preclinical and early clinical studies. These trials are not only assessing the safety and dosing of these agents but are also providing preliminary insights into their efficacy in both hematologic and solid tumor settings.

Current Clinical Trials

One of the most notable clinical trials in the hematologic malignancy arena is the Phase I/II trial of the CD16 × IL-15 × CD33 TriKE (commonly designated as GTB-3550) for patients with relapsed or refractory AML and high-risk MDS. Interim clinical data have shown that treatment with GTB-3550 is associated with a reduction in cancer cell burden and an expansion of activated NK cells, all while maintaining a favorable safety profile with no evidence of cytokine release syndrome (CRS). The study is designed to determine the maximum tolerated dose, optimal dosing schedule, and overall safety, and early data are promising in terms of tolerability and efficacy.

In the realm of solid tumors, early-phase trials are exploring TriKEs such as the B7-H3-targeted TriKE for the treatment of ovarian cancer and perhaps other B7-H3-positive malignancies. Preclinical models have supported the idea that this approach can mitigate immune suppression within the tumor microenvironment and result in significant tumor control, leading several companies, including GT Biopharma, to advance these constructs into clinical studies. Although these trials are at a nascent stage, the encouraging preclinical data have fostered optimism that TriKEs could fill a substantial unmet need in solid tumor immunotherapy.

Additionally, there have been reports of studies involving HER2-targeted TriKEs. The CAM1615HER2 TriKE, for example, has progressed through preclinical stages and is now under consideration for clinical evaluation in HER2-positive cancers, particularly ovarian cancer, where conventional therapies have not yielded durable responses. This study is expected to provide important data on the efficacy of NK cell-driven cytotoxicity in a solid tumor setting and may pave the way for broader application in other HER2-positive malignancies.

Results from Early-Phase Studies

Early-phase studies of TriKEs have provided important insights regarding both their biological activity and clinical potential. For hematologic malignancies, preclinical studies have consistently shown that TriKE treatment results in a significant expansion and activation of NK cells. For instance, the CD33-targeted TriKE has been demonstrated to increase NK cell degranulation and cytokine production in vitro, leading to effective killing of AML cells. In xenograft models, these TriKEs have produced durable responses without significant off-target toxicity, supporting the rationale for clinical translation.

In the solid tumor context, early studies using the B7-H3 TriKE have shown that the molecule induces robust NK cell-mediated cytotoxicity against various B7-H3-positive carcinoma models, including ovarian cancer. In vitro assays have confirmed that NK cells exposed to B7-H3 TriKE expand preferentially compared to controls, and animal studies have demonstrated their ability to control tumor growth while effectively homing to the tumor site. Likewise, the HER2-targeted TriKE has been associated with significant antitumor effects in animal models, where NK cell activation leads to tumor regression even in aggressive cancer models that are resistant to other therapies.

Clinical trial data, though still early, have begun to demonstrate that these preclinical benefits can translate to human subjects. In the Phase I/II study of the CD33-targeted TriKE, initial data reported a significant reduction in the leukemic blast count in patients with AML/MDS, with a majority of patients experiencing stable disease or partial responses. Importantly, the safety profile was favorable, and the incorporation of IL-15 in the TriKE construct appears to mitigate some of the limitations of previous NK cell-based therapies by promoting sustained NK cell activity.

Furthermore, advanced preclinical models simulating the tumor microenvironment of solid tumors have underscored the potential advantages of TriKE therapy. The ability to activate NK cells in the presence of high levels of immunosuppressive cytokines and myeloid-derived suppressor cells (MDSCs) is especially noteworthy, given that many solid tumors create an immunologically “cold” microenvironment that is resistant to conventional checkpoint inhibitors. The data showing that TriKEs can overcome these barriers by delivering IL-15 specifically to NK cells through CD16 engagement is one of the pivotal preclinical findings that support their further evaluation in clinical studies.

Challenges and Future Directions

Despite the promising data, there remain numerous scientific and clinical challenges that must be addressed to fully realize the potential of TriKEs. Researchers are actively investigating these hurdles and devising novel strategies to optimize these molecules for clinical use.

Scientific and Clinical Challenges

One of the primary challenges in the development of TriKEs is the inherent complexity of designing a molecule that maintains balanced affinities to three distinct targets. The optimal configuration must ensure that the NK cell is efficiently activated without causing off-target immune responses or exhaustion of the effector cell population. Achieving the precise molecular geometry where the binding domains function synergistically is a complex task, and any imbalance in binding affinities could lead to suboptimal activation or unwanted toxicities.

In addition, the pharmacokinetic properties of TriKEs must be carefully managed. The inclusion of the IL-15 moiety to promote NK cell proliferation introduces an additional variable in the molecule’s half-life and bioavailability. Although IL-15 has the potential to markedly enhance NK cell persistence, uncontrolled systemic exposure to IL-15 can lead to cytokine-mediated toxicities. Therefore, ensuring that IL-15 is delivered in a targeted manner only to the desired NK cell population is critical. Preclinical studies have helped to delineate these parameters, but real-world clinical application will require rigorous dose-escalation studies and close monitoring.

Another challenge lies within the tumor microenvironment itself, especially in solid tumors. Solid tumors often develop mechanisms to evade immune recognition, including the upregulation of inhibitory ligands and recruitment of suppressive cell types such as regulatory T cells and MDSCs. TriKEs must not only be able to activate NK cells but also overcome these immunosuppressive cues. Studies have shown that TriKE-induced NK cell activation can indeed counteract some of these effects, but it remains to be seen whether this will be sufficient in all tumor types or whether combination strategies with checkpoint inhibitors or other immune modulators will be necessary.

Furthermore, while early clinical data are encouraging, the long-term effects of TriKE therapy are still unknown. There is a need to monitor for potential immune-related adverse events, development of resistance, and the possibility of antigen loss on tumor cells following sustained immune pressure. The durability of responses and whether TriKEs can induce immunological memory that would prevent relapse are significant questions that are currently under investigation.

Future Prospects in TriKE Research

Looking forward, the future of TriKE research is extremely promising. Ongoing efforts are focused on refining the engineering of these molecules to enhance their specificity, potency, and safety profile. One major innovation is the integration of nanobodies as the targeting elements instead of conventional single-chain variable fragments (scFvs). Nanobodies offer improved stability, a smaller size, and enhanced tissue penetration, which could significantly improve the ability of TriKEs to reach tumor sites, particularly in solid tumors.

In the hematologic malignancy space, further clinical trials are planned to optimize dosing regimens and evaluate combinations with other treatments, such as chemotherapeutic agents and checkpoint inhibitors. There is considerable interest in investigating whether TriKEs can be used as part of multi-agent regimens to provide a synergistic effect that might overcome resistance seen with single-agent therapy. In this context, ongoing Phase I/II studies of the CD33-targeted TriKE are expected to provide valuable insights into how best to integrate these agents into existing treatment paradigms.

For solid tumors, the future direction includes expanding the repertoire of tumor antigens targeted by TriKEs. With the rapid progress in genomic profiling and precision oncology, it is becoming increasingly feasible to design TriKEs that are customized for an individual patient’s tumor antigen profile. This personalized approach could greatly enhance therapeutic efficacy and reduce off-target effects. Furthermore, combinatorial approaches that integrate TriKEs with other modalities—such as radiotherapy, conventional chemotherapy, or other forms of immunotherapy—are under active investigation, with the hope of synergistically overcoming the immunosuppressive tumor microenvironment.

Another exciting avenue is the potential use of TriKEs in earlier lines of therapy or even as maintenance therapy following initial remission. The ability of TriKEs to sustain NK cell populations means they might help in preventing relapse by continuously surveilling for and eliminating residual tumor cells. This is particularly relevant for diseases like AML and MDS, where relapse rates remain high despite initial responses to therapy.

Finally, as clinical trials progress, the integration of advanced imaging techniques and biomarker studies will be critical to refining patient selection and optimizing treatment. For example, real-time tracking of NK cell expansion and tumor infiltration, coupled with molecular assessments of the tumor microenvironment, will help clinicians understand the mechanistic underpinnings of response and resistance. This comprehensive approach may lead to better stratification of patients who are most likely to benefit from TriKE therapy, ultimately contributing to more personalized and effective treatment algorithms.

Conclusion

In summary, Trispecific Killer Cell Engagers (TriKEs) represent a groundbreaking approach to cancer immunotherapy by combining three key functions into a single molecule: targeted engagement of NK cells via the CD16 receptor, precise recognition of tumor-associated antigens on malignant cells, and the delivery of IL-15 to promote NK cell expansion. This multi-pronged strategy is designed to overcome some of the intrinsic limitations of prior immunotherapy approaches, including insufficient NK cell persistence and the immunosuppressive microenvironments of tumors.

TriKEs are being investigated for a broad spectrum of indications. In hematologic malignancies, especially acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS), TriKEs such as the CD16 × IL-15 × CD33 construct have shown promising preclinical and early clinical results, demonstrating robust NK cell activation and effective killing of leukemic cells. These studies support further clinical development and optimization in blood cancers, where antigen targeting (e.g., CD33, CLEC12A, CD19, and CD22) has significant therapeutic potential.

In parallel, the utility of TriKEs is also being explored in solid tumors. Preclinical data indicate that TriKEs targeting antigens such as B7-H3 and HER2 can effectively activate NK cells to control tumor growth in models of ovarian cancer, breast cancer, and even prostate cancer. Early data from these studies not only highlight the ability of TriKEs to overcome the immune suppressive milieus characteristic of solid tumors but also suggest potential for combination with other therapeutic modalities. The versatility of the TriKE platform, including the potential for rapid re-design to target newly identified antigens, offers an adaptable and personalized approach to cancer therapy.

Clinical trials to date, including early-phase studies of the CD33-targeted TriKE (GTB-3550) as well as emerging investigations in solid tumor settings, are already demonstrating safety and early signs of efficacy. Importantly, the incorporation of IL-15 within the molecule appears to be pivotal in driving NK cell expansion and function, translating into measurable antitumor responses both in vitro and in vivo. These advancements underscore the potential of TriKEs to serve as both monotherapy and as part of combinatorial treatment regimens, potentially revolutionizing how both hematologic and solid tumors are managed.

Nevertheless, challenges remain. The complexity of molecule design, optimal dosing, management of systemic cytokine exposure, and overcoming the immunosuppressive tumor microenvironment are significant hurdles that necessitate further research. Future directions in TriKE research include refining molecular architecture—potentially through the use of nanobody technology—to improve stability and tissue penetration, as well as developing combination strategies to enhance overall efficacy. With ongoing and future clinical trials, these obstacles are progressively being addressed, promising a new era of NK cell-based immunotherapy that could have far-reaching implications across multiple cancer types.

In conclusion, current evidence underscores a broad and exciting investigational landscape for TriKEs, spanning both hematologic malignancies and solid tumors. These novel agents offer a promising new avenue to harness the full cytotoxic potential of NK cells, addressing unmet clinical needs in diseases that have thus far been refractory to conventional therapies. As clinical development advances and our understanding of NK cell biology deepens, TriKEs may well emerge as a central component of future cancer immunotherapy regimens, offering durable, targeted, and less toxic treatment options for patients across a wide range of malignancies.

Overall, TriKEs embody the evolution from conventional antibody-based therapies to next-generation immunomodulators, providing a versatile and potent platform for targeting cancer in both hematologic and solid disease settings. Continued research, supported by rigorous clinical trials and sophisticated preclinical modeling, is essential to fully elucidate their therapeutic potential. The ongoing integration of advanced engineering techniques, comprehensive biomarker analyses, and combination treatment strategies will likely pave the way for the successful translation of TriKEs into clinical practice, ultimately improving patient outcomes and expanding the armamentarium of cancer immunotherapy.

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.