what are the top Trispecific killer cell engager (TriKE) companies?

20 March 2025

Introduction to Trispecific Killer Cell Engagers (TriKEs)
 
Trispecific killer cell engagers (TriKEs) are a novel class of engineered immunotherapeutics designed to harness and potentiate the cytotoxic capabilities of natural killer (NK) cells against cancer. In contrast to traditional monoclonal antibodies that bind to a single antigen, TriKEs are composed of three distinct binding domains. Typically, one arm of the molecule targets the activating receptor CD16 on NK cells, another arm binds to a tumor-associated antigen (TAA) specific to malignant cells, and the third domain consists of an interleukin-15 (IL-15) moiety that serves as a cytokine signal to promote NK cell expansion, survival, and enhanced cytotoxic activity. This triple interaction facilitates the creation of an immunologic synapse between NK cells and tumor cells, thereby enabling more precise and sustained immune-mediated cell killing. The inclusion of IL-15, in particular, addresses NK cell persistence issues typically seen in adoptive therapies, ensuring that the effector cells remain active and proliferative in the tumor microenvironment.

Importance in Cancer Immunotherapy 
The strategic importance of TriKEs in cancer immunotherapy lies in their dual ability to overcome two critical challenges: the lack of specificity and the limited in vivo persistence of NK cells. First, by simultaneously engaging a TAA and an activating receptor on NK cells, TriKEs significantly enhance the specificity of the immune response against malignant cells, thereby lowering the off-target effects commonly associated with other immunotherapeutic strategies. Second, the local delivery of IL-15 through the TriKE format uniquely drives NK cell expansion directly at the tumor site, reducing the need for systemic cytokine administration that might result in severe toxicities. These features not only expand the potential applicability of NK cell-based therapies to various hematological malignancies but also hold promise for tackling solid tumors where the immunosuppressive microenvironment has historically limited therapeutic efficacy. Collectively, TriKEs represent a transformative step in immunotherapy, merging the strengths of targeted antibodies with the potent effector functions of NK cells.

Leading Companies in TriKE Development

Top Companies Overview 
In the rapidly evolving landscape of NK cell engaging therapies, several companies have emerged as leaders in the development and clinical translation of TriKE platforms. Foremost among these is GT Biopharma, Inc. GT Biopharma is recognized for its proprietary TriKE® technology platform, which has led to the development of multiple TriKE candidates such as GTB-3550 (a CD16/IL-15/CD33 TriKE) for high-risk hematological malignancies and its solid tumor-focused variants (GTB-4550 targeting PD-1 and GTB-5550 targeting B7-H3). The company has successfully advanced these molecules through preclinical models as well as early phase clinical trials, indicating robust target engagement, effective NK cell activation, and significant antitumor activity.

Another key player in the emerging TriKE space is Harpoon Therapeutics. While Harpoon is primarily known for its innovative TriTAC designs, which are diabody-based trispecific constructs that similarly target cancer cells, the company’s approach closely parallels that of traditional NK cell engager strategies. Harpoon’s TriTACs, such as HPN 328 and HPN217, are designed using a nanobody for specific tumor antigen recognition combined with an anti-CD3 single-chain variable fragment (scFv) and a nanobody against human serum albumin (HSA) for improved half-life. Although not strictly classical NK cell engagers, these molecules contribute to the broad landscape of multispecific therapeutics by engaging immune effector cells and demonstrating promising clinical profiles in early stage trials.

The Swiss company Numab is also making strides in the development of trispecific molecules. Their construct, NM21-1480, while differing in mechanism from standard NK cell engagers, employs an scFv-based format to simultaneously bind PD-L1, 4-1BB, and HSA. Although NM21-1480 does not engage NK cells in the traditional sense, it exemplifies the trispecific antibody paradigm by linking costimulatory signals with tumor-associated targets to enhance immune activation. This strategy represents an alternative but complementary approach to leveraging multispecific formats for cancer immunotherapy.

In addition to these leading companies, strategic collaborations between established biopharmaceutical companies and specialized molecular partners are contributing to innovation in this domain. Amgen, through partnerships targeting designed ankyrin repeat proteins (DARPins), has been involved in developing antibody-mimetic polypeptides that function in a manner similar to TriKEs, further diversifying the technological approaches to NK cell activation. Such collaborations underscore the dynamic nature of the field, where companies are not only developing standalone TriKE platforms but are also partnering to integrate novel biomarkers, targeting moieties, and cytokine signals for improved safety and efficacy.

Key Technologies and Innovations 
The technological foundations supporting TriKE development encompass advanced protein engineering, recombinant fusion protein technology, and state-of-the-art cytokine delivery mechanisms. GT Biopharma’s TriKE platforms leverage a modular design wherein each domain is optimized for stability, specificity, and activity. Utilizing a single-domain antibody (sdAb) approach for targeting CD16 allows for a more refined engagement of NK cells, reducing the potential for off-target activation compared to conventional scFv formats. The incorporation of IL-15 into these constructs is particularly innovative because, unlike systemic IL-15 therapy that can trigger widespread immune activation and toxicity, the TriKE format localizes the cytokine signal precisely where it is most needed—at the interface between the NK cell and the tumor target.

From a manufacturing standpoint, scalability and consistency are critical. GT Biopharma has invested in processes to rapidly produce IND-ready TriKE candidates which can be advanced into clinical trials with confidence in batch-to-batch reproducibility. The diabody-engineered TriTACs developed by Harpoon Therapeutics exemplify another angle of innovation. Their use of nanobody scaffolds facilitates the creation of molecules that are smaller, more stable, and potentially more penetrative within solid tumors. This is a crucial consideration, given that many immunotherapies face resistance due to limited tumor microenvironment accessibility.

Furthermore, companies like Numab are breaking new ground by incorporating costimulatory receptor targeting into their trispecific designs. The simultaneous binding to PD-L1 and 4-1BB not only blocks immune checkpoint pathways but also actively stimulates T or NK cell proliferation and activation, thereby overcoming tumor-induced immunosuppression. Other innovations include the deployment of engineered ankyrin repeat proteins by molecular partners allied with Amgen. These DARPins mimic antibody functionality but offer advantages in terms of reduced molecular weight and improved tissue penetration, key factors in the successful treatment of solid tumors.

In summary, the leading TriKE companies excel through the integration of precision binding, innovative cytokine delivery mechanisms, and a deep understanding of immune cell biology. Their designs are rooted in rigorous preclinical validations and early clinical successes, positioning them as frontrunners in next-generation immunotherapies.

Market Landscape

Market Trends and Dynamics 
The market for NK cell engaging therapies, and TriKEs in particular, is experiencing robust growth, spurred by the increasing recognition of NK cells as potent mediators of antitumor immunity. The integration of tri-specific modalities into cancer treatment regimens has been propelled by the need to address the limitations of current immunotherapies. In recent years, there has been a notable shift towards molecules that can simultaneously engage immune effector functions and provide localized cytokine support—a trend that is driving the development of TriKEs.

Market dynamics in this space are characterized by a strong emphasis on hematological malignancies, where early clinical trials have demonstrated promising safety and efficacy profiles. However, the landscape is rapidly evolving to include solid tumors, supported by the innovation in targeting different TAAs such as PD-1, B7-H3, and CLEC12A. The development of TriKEs that aim to reinvigorate NK cells in cold tumor microenvironments, such as metastatic castration-resistant prostate cancer (mCRPC), further exemplifies the expanding potential of these agents.

From an investor perspective, companies like GT Biopharma are attracting significant interest due to their advanced pipeline and the promising clinical data emerging from early phase trials. The trend towards scalable, off-the-shelf therapeutic products also positions TriKE developers favorably within the broader immuno-oncology market, which is expected to continue its rapid expansion over the next decade. Additionally, strategic licensing agreements, such as the exclusive worldwide license held by GT Biopharma with the University of Minnesota for the TriKE technology, further validate the commercial potential of these platforms.

Competitive Analysis 
The competitive landscape in TriKE development is marked by a mix of established clinical stage biopharmaceutical companies and emerging biotech startups. GT Biopharma, Inc. remains the clear leader in this niche, owing to its pioneering TriKE platform and the breadth of clinical candidates in its pipeline. With multiple TriKE molecules addressing various tumor antigens and indications, GT Biopharma’s approach underscores its commitment to translating preclinical innovations into tangible clinical benefits.

Harpoon Therapeutics represents a compelling competitor, although its focus on TriTAC designs and T cell engaging modalities positions it slightly differently. Nonetheless, Harpoon’s expertise in diabody engineering and its ability to extend the half-life of these molecules through nanobody integration provide a robust competitive challenge. Their pipeline includes constructs such as HPN 328 and HPN217, which, while not strictly conventional NK cell engagers, demonstrate the company’s capability to innovate multispecific approaches for immune cell activation.

Numab, with its unique NM21-1480, pushes the envelope further by incorporating receptor costimulation (PD-L1, 4-1BB, and HSA binding) into its trispecific format. This innovation addresses both immune checkpoint blockade and the enhancement of immune cell effector functions, distinguishing it from traditional NK cell engager strategies. Meanwhile, Amgen’s strategic collaborations leveraging DARPins add another competitive element, particularly as these engineered proteins offer advantages in terms of size, tumor penetration, and manufacturing scalability.

In the market, differentiation is based not only on clinical data but also on technological superiority, scalability, and the ability to position the product as a monotherapy or as part of combination regimens. Companies that achieve these milestones are better placed to capture market share in an increasingly crowded immuno-oncology space. The competitive dynamics are further influenced by regulatory approvals, intellectual property portfolios, and the robustness of clinical trial data demonstrating improved NK cell function and patient outcomes.

Overall, while GT Biopharma is currently at the forefront, the continuous innovation by competitors such as Harpoon Therapeutics, Numab, and collaborative efforts involving companies like Amgen signals a dynamic market where technological evolution and clinical validation will be critical to long-term success.

Challenges and Future Prospects

Current Challenges in TriKE Development 
Despite their immense promise, TriKE development faces several technical and clinical hurdles. One of the primary challenges is ensuring the optimal balance between NK cell activation and safety. While IL-15 is a potent stimulant for NK cell proliferation, its incorporation must be carefully calibrated to avoid excessive cytokine release or off-target toxicities that are sometimes observed with systemic cytokine therapies. Early clinical and preclinical data indicate that localized delivery via the TriKE format can mitigate some of these issues, but fine-tuning the dose and delivery remains an area of active research.

Another challenge is addressing tumor heterogeneity, particularly in the context of solid tumors. The expression levels of TAAs can vary significantly both within and between tumor types, necessitating the design of TriKEs with high affinity and specificity. While GT Biopharma and its competitors have developed molecules targeting antigens such as CD33, PD-1, B7-H3, and CLEC12A, ensuring consistent efficacy across diverse patient populations is a complex task. Additionally, the immunosuppressive tumor microenvironment, characterized by factors such as hypoxia and the presence of myeloid-derived suppressor cells (MDSCs), poses further barriers to NK cell infiltration and activity. Recent studies have shown that tailored TriKE constructs, such as the PSMA TriKE designed for mCRPC, can relieve NK cell suppression even under adverse conditions; however, this remains a significant challenge in broader clinical settings.

Manufacturing and scalability represent further obstacles. The complexity of producing multispecific recombinant proteins that retain high stability and bioactivity requires advanced biotechnological manufacturing solutions. Companies must invest not only in process development but also in ensuring regulatory compliance and quality control across large-scale production batches. GT Biopharma, for instance, has made considerable progress in establishing scalable processes, yet the industry as a whole continues to work towards more efficient production methods.

Intellectual property (IP) and competitive differentiation are also challenges. With multiple companies racing to patent various TriKE constructs and related technologies, navigating the IP landscape can be challenging. Companies must continuously innovate and secure robust patent protections to maintain a competitive edge in what is becoming an increasingly crowded field. Finally, integrating TriKE therapies into existing treatment paradigms and defining their role—whether as standalone therapies or in combination with other modalities like CAR T cells or checkpoint inhibitors—requires further clinical validation and collaboration across the oncology community.

Future Directions and Opportunities 
Looking ahead, the future of TriKEs in cancer immunotherapy appears promising, with numerous opportunities to advance both their design and clinical application. One key direction is the further refinement of cytokine fusion strategies. Future TriKEs may incorporate next-generation cytokines or modified IL-15 moieties engineered to optimize NK cell activation while minimizing systemic toxicities. This could involve the development of IL-15 variants with altered receptor-binding characteristics or the combination of IL-15 with other costimulatory molecules to achieve synergistic effects.

Expanding the scope of target antigens is another important avenue for future research. Although current TriKE constructs often target markers such as CD33 or B7-H3, there is significant potential to broaden the applicability of these agents to a wider range of cancers, including those with heterogeneous antigen expression. This might involve using dual-targeting strategies where two different TAAs are simultaneously engaged, thereby reducing the chances of immune escape and enhancing the precision of NK cell-mediated cytotoxicity.

Combination therapies represent a fertile area of development for TriKEs. Given the multifaceted nature of cancer, it is unlikely that a single agent will provide a complete cure. Instead, TriKEs could be combined with immune checkpoint inhibitors, CAR T cell therapies, or conventional chemotherapeutics to maximize antitumor efficacy. Preclinical models have already demonstrated the potential for such synergistic combinations, and future clinical trials are expected to explore these strategies in greater detail. Additionally, real-time biomarker monitoring and adaptive trial designs will help optimize treatment regimens and personalize therapy based on patient-specific tumor characteristics and immune profiles.

From a technological perspective, advances in protein engineering and manufacturing will further enhance the durability, stability, and bioactivity of TriKE molecules. Methods such as the incorporation of designed ankyrin repeat proteins (DARPins) or other antibody-mimetic scaffolds are likely to improve tissue penetration and reduce the immunogenicity of these constructs. Such innovations will not only refine the therapeutic window but also facilitate the development of off-the-shelf products that can be rapidly deployed in clinical settings.

In addition, the development of novel delivery platforms—including nanoparticle-based systems or local depot formulations—could revolutionize the bioavailability and pharmacokinetics of TriKEs. These technologies may allow for controlled, sustained release of the therapeutic agent directly into the tumor microenvironment, thus optimizing NK cell activation and minimizing adverse events. Furthermore, enhanced imaging and tracking systems in clinical trials will provide valuable insights into the in vivo behavior of TriKEs, guiding the iterative design process and enabling more precise patient stratification.

On the regulatory front, as TriKE candidates progress through clinical trials and generate robust efficacy data, regulatory agencies are expected to provide clearer pathways for approval. This is especially pertinent in light of recent successes with bispecific therapeutics and the increasing comfort of regulators with multispecific antibody formats. A favorable regulatory environment will likely accelerate the transition of TriKEs from experimental treatments to mainstream clinical options.

Finally, economic and strategic partnerships are set to play a crucial role in the future development of TriKEs. Collaborations between biotech companies, academic institutions, and larger pharmaceutical firms—exemplified by the exclusive licensing agreements and co-development partnerships seen in companies like GT Biopharma—will be critical in overcoming the financial and technical barriers to clinical translation. These partnerships can provide the necessary capital, expertise, and access to clinical trial networks to bring next-generation TriKEs to market, ultimately expanding the reach and impact of these therapies globally.

Conclusion

In conclusion, the landscape of trispecific killer cell engager (TriKE) companies is both dynamic and multi-faceted. GT Biopharma, Inc. emerges as the clear leader, driven by its proprietary TriKE® platform that integrates CD16 targeting for NK cell engagement, tumor antigen specificity, and localized IL-15 delivery. This technology has shown promise across multiple indications, particularly in hematological malignancies, and is expanding into solid tumor applications through various constructs such as GTB-3550, GTB-4550, and GTB-5550. 

Harpoon Therapeutics, while primarily focused on diabody-based TriTACs for T cell engagement, offers complementary innovations that further underscore the potential of multispecific antibody approaches in immunotherapy. Similarly, Numab introduces a unique approach with its NM21-1480 construct that simultaneously targets immune checkpoint molecules and provides costimulatory signals, embodying the progress in trispecific designs beyond traditional NK cell engagers. Collaborative efforts involving companies like Amgen and strategic partnerships leveraging designed ankyrin repeat proteins (DARPins) demonstrate that innovation in this field is not limited to a single modality; instead, it is a concerted effort across the biopharmaceutical sector to harness and enhance innate immunity for cancer therapy.

The market landscape for TriKEs is buoyed by positive clinical data, promising preclinical models, and a growing understanding of the importance of targeted cytokine delivery. Market dynamics are characterized by an increasing shift toward precision immunotherapies that address both specificity and in vivo persistence of NK cells, essential for overcoming the challenges posed by heterogeneous tumor biology and immunosuppressive microenvironments. Although significant technical, manufacturing, and regulatory challenges remain, ongoing innovation, robust clinical trials, and strategic collaborations will continue to drive the evolution of this therapeutic class.

Looking forward, future directions for TriKE development involve refining cytokine integration, broadening antigen targeting, and formulating combination strategies to synergize with other immunotherapeutic modalities. Advances in protein engineering, delivery technologies, and scalable manufacturing processes will be critical to translating early successes into clinically approved therapies. As the competitive landscape matures and more companies enter the space, the continued innovation and strategic partnerships witnessed today promise to transform TriKEs into a cornerstone of future cancer immunotherapy.

Through a general-specific-general narrative, we can see that TriKE companies are not only addressing the complex challenges of cancer treatment by leveraging the innate power of NK cells but are also pioneering technologies that promise to transform therapeutic paradigms. In balancing efficacy, specificity, and safety, these companies are paving the way for innovative treatments that could ultimately improve patient outcomes across a range of malignancies. The combined efforts of GT Biopharma, Harpoon Therapeutics, Numab, and allied technological collaborations represent a multifaceted approach to next-generation immuno-oncology, providing hope and tangible progress in the fight against cancer.

Overall, while GT Biopharma currently stands out as the frontrunner in the TriKE arena, the coordinated advances by other companies highlight a competitive and innovative ecosystem. This ecosystem is well poised to address current challenges and exploit future opportunities, ensuring that TriKEs—and the broader multispecific antibody technologies—will continue to evolve and redefine cancer immunotherapy in the years ahead.

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.