For what indications are Bispecific killer cell engager (BiKE) being investigated?

17 March 2025
Introduction to Bispecific Killer Cell Engagers (BiKEs)

Definition and Mechanism of Action
Bispecific killer cell engagers (BiKEs) are engineered antibody constructs that possess dual specificity, allowing them to simultaneously bind to an antigen expressed on tumor cells and to an activating receptor on natural killer (NK) cells, most commonly CD16. This dual binding creates an immunologic synapse between NK cells and target cancer cells, thereby triggering NK cell activation, degranulation, cytokine release, and ultimately antibody‐dependent cellular cytotoxicity (ADCC) against malignant cells. Functionally, by directly engaging CD16, BiKEs bypass the need for conventional Fc-mediated interactions and overcome limitations associated with serum IgG competition, making them highly efficient at activating NK cells even in an environment with endogenous antibodies. Their modular molecular design—often constructed from linked single-chain variable fragments (scFv) or nanobody formats—has allowed significant improvements both in targeting specificity and in enhancing NK cell function. The overall mechanism positions BiKEs as highly promising agents for targeted cancer immunotherapy.

Overview of BiKE Development
Over the past two decades, there has been a rapid clinical evolution of bispecific antibodies, including those engaging NK cells. Early studies demonstrated the feasibility of redirecting NK cell cytotoxicity to tumor targets using BiKEs, and subsequent research focused on refining their molecular design and efficacy. For instance, pioneering studies engineered CD16×CD33 BiKEs that effectively activated NK cells against CD33+ acute myeloid leukemia (AML) cell lines and primary blasts; these constructs laid the groundwork for later clinical development. Moreover, innovations have led to the development of trispecific killer cell engagers (TriKEs), which integrate an interleukin-15 (IL-15) crosslinker between scFv modules to further induce NK cell expansion, activation, and in vivo persistence. The evolution of BiKE technology has been largely driven by the need to overcome challenges associated with low immune infiltration, limited in vivo NK cell expansion, and the immunosuppressive tumor microenvironment—factors which impede successful immunotherapy in both hematological and solid tumors.

Current Clinical Indications for BiKEs

Approved Indications
Although many bispecific antibody-based therapies have received regulatory approval, the first wave of clinical approvals was primarily seen in Bi-specific T cell engagers (BiTEs), such as blinatumomab for B-cell acute lymphoblastic leukemia (ALL). For BiKEs, even though several constructs are in various stages of clinical development for many indications, there are no BiKEs as monotherapy that have yet reached full regulatory market authorization. Certain BiKEs that target NK cells, notably the CD16×CD33 BiKE, have shown promising preclinical data in activating NK cells against malignant cells in myeloid diseases, and early-phase clinical evaluations have been initiated in the hematological setting. Therefore, while no BiKE has been formally “approved” in the same sense as some T-cell engagers, the development pathway indicates eventual approvals are anticipated, particularly given the robust data emerging in hematologic malignancies.

Ongoing Clinical Trials
A significant focus has been on evaluating BiKEs in clinical trials for hematological malignancies. For example, the CD16×CD33 BiKE is being investigated for its therapeutic potential in acute myeloid leukemia (AML) as well as in myelodysplastic syndromes (MDS). Preclinical and early-phase clinical studies have shown that these agents not only enhance NK cell-mediated cytotoxicity but also restore NK cell effector functions in patients with abnormal NK cell phenotypes. In pediatric patients with AML and biphenotypic acute lymphoblastic leukemia (ALL), CD16×CD33 BiKEs have been tested for their ability to reverse NK cell deficiency and improve outcomes when combined with allogeneic NK cell infusion.
In addition, early clinical investigations focusing on NK cell-engaging therapies include trials with TriKE constructs such as the 161533 TriKE (comprising anti-CD16, anti-CD33, and an IL-15 crosslinker) that have already progressed into phase I/II clinical evaluations for high-risk AML and MDS patients. Moreover, exploratory studies are being conducted in other hematologic malignancies where alternate targets such as CD19 have been incorporated into the BiKE/TriKE platforms, indicating potential utility in chronic lymphocytic leukemia (CLL) and B-cell leukemias.
Clinical trial registries (as per the involved organizations and published data on the synapse platform) show multiple ongoing studies aimed at evaluating NK cell engagers in combination with established therapies (e.g., checkpoint inhibitors) to determine improved synergistic anti-tumor effects. These trials provide evidence that the clinical indication spectrum for BiKEs is beginning with hematological malignancies, although the robust activity in preclinical models has set the stage to expand into other domains as well.

Potential Future Indications

Emerging Research Areas
Beyond hematologic malignancies, there is increasing interest and emerging evidence to pursue the use of BiKEs in solid tumors—an area historically challenging for NK cell-based therapies. Preclinical studies have demonstrated that modifications to BiKE constructs can overcome the hurdles of the immunosuppressive tumor microenvironment characteristic of many solid tumors. For instance, trispecific constructs such as 133EpCAM16 have been engineered to simultaneously engage NK cells via CD16 and tumor cells expressing both CD133 and EpCAM, thereby targeting both differentiated tumor cells and cancer stem cells (CSC). In vitro studies using Caco-2 colorectal carcinoma cell lines have confirmed that these engineered TriKEs induce potent ADCC with moderate cytokine secretion, suggesting a favorable therapeutic window.
Similarly, targets such as HER2, EGFR, and EpCAM in epithelial carcinomas have been explored as solid tumor antigens for BiKEs, with early preclinical evidence showing a marked enhancement in NK cell-mediated tumor cell cytotoxicity. Targeting certain immune checkpoint molecules expressed on tumor cells, like B7-H3, have also been investigated by redirecting NK cells through BiKEs in non-small cell lung cancer (NSCLC) and other solid tumors, highlighting future directions in solid tumor immunotherapy. Moreover, innovative designs that further enhance tumor specificity—by adding a third binding domain to address heterogeneity in antigen expression—are being developed and could extend the application of BiKEs to a broader range of solid tumor indications.

Preclinical Studies and Findings
Preclinical models have provided compelling evidence supporting the extension of BiKE use into multiple new indications. Animal models and in vitro studies have demonstrated that the activation of NK cells via BiKEs can lead to significant tumor reduction in both blood-borne and solid tumor models. For example, the CD16×CD33 BiKE has been associated with potent NK cell cytotoxicity against CD33+ leukemic cells in xenograft models, and its activity extends to targeting myeloid-derived suppressor cells (MDSCs) that contribute to immune evasion in the bone marrow microenvironment.
In addition, similar constructs targeting CD19 and CD133 have shown preliminary effectiveness against B-cell leukemias and carcinoma cells, respectively, in both in vitro cytotoxicity assays and in vivo tumor models. The incorporation of IL-15 in TriKEs enhances not only NK cell activation but also their in vivo persistence and expansion, making these constructs promising candidates to treat relapsed and refractory forms of hematologic cancers.
Another compelling area of investigation is the potential for BiKEs in the treatment of pediatric malignancies. Preclinical studies and early clinical data in pediatric cohorts have indicated that BiKEs might restore compromised NK cell activity, a vital requirement for effective immunotherapy in children with conditions like AML and biphenotypic ALL.
Furthermore, the expanding interest in cancer stem cell (CSC) targeting—as CSCs are implicated in tumor recurrence and treatment resistance—has led researchers to develop BiKEs and TriKEs with dual binding domains for CSC markers such as CD133, in combination with tumor-associated antigens like EpCAM. These constructs have demonstrated enhanced NK cell-mediated lysis of CSC-enriched populations in preclinical studies, suggesting that they might be ushering in a new therapeutic approach for solid tumors where CSCs play a pivotal role.
Overall, the preclinical data suggest that the indications for BiKEs could eventually span a diverse range of cancers—from hematological malignancies like AML, MDS, ALL, and CLL to solid tumors including colorectal, NSCLC, breast, and gliomas—while also offering promise in eradicating CSCs and overcoming tumor heterogeneity.

Challenges and Considerations

Safety and Efficacy Concerns
While the preclinical and early clinical data for BiKEs are promising, several challenges must be addressed to ensure safety and efficacy in a broader patient population. One key safety concern is the potential for sudden and extensive cytokine release following robust NK cell activation, which could lead to systemic inflammation and toxicity. Although many studies report only moderate cytokine production upon NK cell activation using BiKEs, systematic evaluation is needed to confirm that the immune activation remains within a clinically tolerable range.
Another challenge is the risk of NK cell fratricide, which refers to NK cells killing each other due to unintended crosslinking of CD16 molecules. Engineering strategies, such as employing specific nanobodies with high affinity—while being independent of Fc-receptor allotype—have been developed to circumvent these risks; however, continuous monitoring during clinical trials is essential.
Efficacy concerns include ensuring that BiKE-mediated NK cell activation translates into meaningful clinical benefits rather than just transient activation. The immunosuppressive tumor microenvironment (TME), especially in solid tumors, can impair NK cell infiltration and function. Strategies that combine BiKEs with other therapeutic modalities (e.g., oncolytic viruses, checkpoint inhibitors, or cytokine adjuvants such as IL-15) are being investigated to overcome these hurdles.
Furthermore, variability in antigen expression on tumor cells may impact the effectiveness of BiKEs. Heterogeneous expression could enable tumor escape from NK cell-mediated lysis if only a subpopulation of tumor cells is recognized. In response, trispecific constructs that target multiple antigens or incorporate costimulatory molecules have been designed, but these also add complexity to the safety evaluation.

Regulatory and Ethical Considerations
The regulatory framework for BiKEs is evolving alongside the technology. Given that BiKEs are classified as biologics with unique structures and mechanisms of action, regulatory agencies require extensive nonclinical safety assessments, proof of efficacy, and stringent manufacturing controls to ensure consistency and quality. Regulatory challenges include the need to standardize assays for NK cell activation, cytotoxicity measures, and cytokine release profiling—all of which are essential for regulatory approval.
Ethically, the use of novel immunotherapeutics like BiKEs requires careful consideration, particularly when early-phase trials involve pediatric populations or heavily pretreated patients. Informed consent processes must thoroughly explain the possible risks related to immune activation, cytokine release, and potential off-target effects. Additionally, the possibility of combining BiKEs with other emerging immunotherapies raises ethical questions regarding complex combination regimens that may complicate the attribution of observed effects to individual agents.

Future Directions and Innovations

Advancements in BiKE Technology
The future of BiKEs is being shaped by continuous technological innovations aimed at improving specificity, potency, and durability of NK cell-mediated therapy. One significant advancement has been the incorporation of an IL-15 crosslinker into the BiKE construct, thereby transforming it into a trispecific killer cell engager (TriKE). This modification not only maintains the antigen specificity and NK cell engagement via CD16 but also confers the dual benefit of inducing NK cell proliferation and increased in vivo persistence. Early-phase clinical studies with IL-15 augmented TriKEs (such as the 161533 TriKE) have shown enhanced anti-leukemic activity and improved NK cell survival in xenograft models.
Another notable innovation is the design of bispecific and trispecific constructs using nanobody-based scaffolds instead of conventional scFv fragments. These nanobodies offer improved solubility, stability, and ease of manufacturing. Furthermore, their ability to bind CD16 independent of serum IgG occupancy minimizes competition and enhances NK cell activation, which is critical when considering fast half-life and bio-distribution profiles.
Advances in recombinant protein engineering and in silico modeling are also providing the tools necessary to optimize binding affinities for both tumor antigens and NK cell receptors, ensuring that BiKEs can operate efficiently even in heterogeneous tumor environments. Such advancements are expected to enhance the translational potential of BiKEs across a wider spectrum of cancers.

Prospective Research and Development
Based on the strong preclinical foundation and early clinical results observed primarily in hematologic malignancies, prospective research will likely push BiKE development into additional indications. Future trials may explore combination therapies, where BiKEs are employed alongside checkpoint inhibitors, cytokine therapies, or oncolytic viruses to combat the immunosuppressive tumor microenvironment and heighten the overall immune response in solid tumor settings.
There is also significant research interest in optimizing BiKE design for the targeting of cancer stem cells (CSCs). CSCs are thought to contribute to relapse and resistance in many cancers, and studies with constructs such as the CD16×133 BiKE and 133EpCAM16 indicate that targeting CSC-specific markers could produce more durable responses and prevent treatment resistance.
Furthermore, expanding the indication of BiKEs to address pediatric malignancies—especially in cases of AML and biphenotypic ALL where NK cell dysfunction is prevalent—could offer an impactful therapeutic avenue. Emerging clinical data suggest that restoration of NK cell function using BiKEs in pediatric populations holds promise, though further research is needed to overcome the inherent challenges of limited perforin and granzyme expression in some patients.
The next generation of research is also expected to involve adaptive trial designs that allow for adjustments in dosing, patient selection, and combination regimens based on early biomarker signals and interim efficacy data. Given the complexity of immune activation and potential off-target effects, such innovative trial frameworks could optimize the clinical development process, reduce risks, and accelerate the translation of promising BiKE constructs into clinical practice.

Detailed Conclusion

In summary, Bispecific Killer Cell Engagers (BiKEs) are emerging as a versatile and innovative approach in cancer immunotherapy. They function by simultaneously engaging an activating receptor (most notably CD16) on NK cells with specific antigens on the surface of malignant cells, thereby inducing a potent ADCC response. Initially developed for hematological malignancies, particularly diseases such as acute myeloid leukemia (AML), myelodysplastic syndromes (MDS), and biphenotypic acute lymphoblastic leukemia (ALL), BiKEs are already showing promising activity in early clinical trials. The incorporation of additional modules, such as IL-15 in the design of TriKEs, has further enhanced their potential by promoting NK cell activation, expansion, and persistence.

Current clinical investigations are concentrated on hematological indications, with CD16×CD33 BiKEs and related TriKEs being tested in high-risk AML and MDS patients, including pediatric cohorts where immune deficits are prevalent. Concurrently, early-phase research and preclinical models indicate that the scope of BiKE applications may extend to solid tumors. Emerging targets for solid tumors include epithelial antigens such as EpCAM, HER2, EGFR, and B7-H3, which are being explored in advanced constructs designed to overcome the immunosuppressive microenvironment and heterogeneity of tumor antigen expression. Furthermore, innovative constructs that target cancer stem cells through dual antigen engagement (e.g., CD133 and EpCAM) have shown enhanced cytotoxicity and could play a crucial role in preventing tumor recurrence and resistance.

Despite these advances, several challenges remain. Safety issues—such as the risk of cytokine release syndrome, NK cell fratricide, and the potential for off-target effects—necessitate rigorous preclinical safety evaluations. Additionally, the regulatory landscape for these novel biologics is still evolving, demanding high standards in manufacturing and thorough characterization of pharmacodynamic and pharmacokinetic profiles. Ethical considerations, especially in vulnerable populations like pediatrics, add another layer of complexity that must be navigated with care.

Looking ahead, continued innovations such as enhanced affinity engineering, nanobody-based formats, and novel combination strategies are set to propel BiKE technology forward. With adaptive clinical trial designs and prospective research initiatives focusing on both hematologic and solid tumor indications, the future of BiKEs appears promising. They represent a new frontier in precision oncology that not only harnesses the innate power of NK cells but also holds the potential to synergize with other immunotherapeutic modalities for improved patient outcomes.

In conclusion, based on the extensive research and multiple perspectives provided by current preclinical and early clinical studies, BiKEs are being investigated for:
- Hematological Malignancies: Including AML, MDS, biphenotypic ALL, and potentially CLL and B-cell leukemias where markers such as CD33 and CD19 are relevant.
- Solid Tumors: Early-stage studies have extended into targeting epithelial antigens in carcinomas (e.g., via EpCAM, HER2, EGFR, and B7-H3), as well as cancer stem cells to prevent relapse and resistance in colorectal, non-small cell lung cancer, and gliomas.
- Pediatric Indications: Particularly in pediatric patients with AML and biphenotypic ALL, where restoration of NK cell activity via BiKEs can be a promising therapeutic intervention.

Thus, the evolving landscape of BiKE technology underscores the potential for broad indications across both hematologic and solid tumor malignancies, with ongoing trials and preclinical studies paving the way for future clinical application. Continued efforts to enhance the molecular design and safety profile of BiKEs, coupled with innovative regulatory and adaptive clinical trial frameworks, will ultimately determine the scope and success of these therapies in the fight against cancer.

Curious to see how Eureka LS fits into your workflow? 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.