Skip to main content
Human Vaccines & Immunotherapeutics logoLink to Human Vaccines & Immunotherapeutics
. 2023 Sep 29;19(2):2256904. doi: 10.1080/21645515.2023.2256904

Principles and current clinical landscape of NK cell engaging bispecific antibody against cancer

Tian Huan a,b,*, Bugao Guan a,*, Hongbo Li a, Xiu Tu a, Chi Zhang a, Bin Tang a,c,
PMCID: PMC10543353  PMID: 37772505

ABSTRACT

Monoclonal antibody-based targeted therapies have greatly improved treatment options for patients by binding to the innate immune system. However, the long-term efficacy of such antibodies is limited by mechanisms of drug resistance. Over the last 50 years, with advances in protein engineering technology, more and more bispecific antibody (bsAb) platforms have been engineered to meet diverse clinical needs. Bispecific NK cell engagers (BiKEs) or tri-specific NK cell engagers (TriKEs) allow for direct targeting of immune cells to tumors, and therefore resistance and serious adverse effects are greatly reduced. Many preclinical and clinical trials are currently underway, depicting the promise of antibody-based natural killer cell engager therapeutics. In this review, we compile worldwide efforts to explore the involvement of NK cells in bispecific antibodies. With a particular emphasis on lessons learned, we focus on preclinical and clinical studies in malignancies and discuss the reasons for the limited success of NK-cell engagers against solid tumors, offering plausible new ideas for curing some advanced cancers shortly.

KEYWORDS: Bispecific antibodies, natural killer cells, bispecific NK cell engager, tri-specific NK cell engager, cancer immunotherapy

NK cell engaging bispecific antibody as an emerging therapy

Immunotherapy is the preferred agent for systemic cancer treatment, and antibody therapy in particular has become the preferred treatment choice for cancer because of its ability to specifically target molecules and low toxicity.1 However, in complex disease pathogenesis, a wide range of mediators help to stimulate separate signaling pathways or promote duplicated signaling cascades, which limit the effectiveness of therapies targeting individual molecules.2 Tumor types are considered to express diverse receptors that interact to activate such tumor cells to proliferate indefinitely, metastasize, and inhibit apoptosis. For this reason, in 1961, Nisonoff et al. made a large class of molecules capable of recognizing two different epitopes or antigens, known as bispecific antibodies, by chemically cross-linking the Fab’ fragments of two polyclonal antibodies that bind different antigens.3 With the rapid development of genetic engineering techniques and new insights into the mechanisms by which tumors evade immune control, the next generation of bispecific or multi-specific antibodies is being developed to eliminate cancer by redirecting immune cells (e.g., T cells, NK cells) to the tumor. Various forms of bispecific antibodies (bsAbs) have been generated, which in clinical practice may show better clinical efficacy than monoclonal antibodies or other conventional anti-tumor therapies.4,5

The redirection of immune cells is not necessarily restricted to T cells. Natural killer (NK) cells are cytotoxic lymphocytes of the innate immune system that can kill tumor or virus-infected cells with effector functions.6 Significantly, a higher percentage of NK cell infiltration in solid tumors is associated with better clinical therapeutic efficacy, as has been demonstrated in breast cancer,7 head and neck cancer,8 and clear cell renal cancer.9 The composition and release of NK lysis granules filled with perforin and granzyme are comparable to that of cytotoxic T cell subpopulations but without strong cytokine release.10 Targeting NK cells by immunotherapy is, therefore, an attractive anti-tumor strategy.

In addition, NK cells could express various activating and inhibitory receptors to respond to the tolerance of or activation signaling of cytotoxicity against the target cells.11 Where the total level of inhibitory receptor signaling surpasses the total level of activating receptor signaling, NK cell activation is thwarted, resulting in the induction of cellular tolerance.12 For instance, NK cells normally upregulate stimulatory ligands for NK cell activation receptors such as natural-killer group 2 (NKG2D) in response to viral infection or transformation. The resulting interaction induces activation of signaling levels that exceed those of receptors at the level of intrinsic signaling via inhibitory signaling (e.g., killer immunoglobulin-like receptors (KIR) and NKG2A), activating the release of NK cell cytokines including products of cytotoxicity to target cells.13

NK cell-mediated antibody-dependent cell-mediated cytotoxicity (ADCC) is another key mechanism for the killing of cancer cells. Several studies have confirmed that the Fc receptor CD16 (CD16A, FcγRIII) is the most effective activating receptor expressed in NK cells, resulting in ADCC-triggered tumor cell lysis when IgG-coated target cells bind to it and occur independently of other co-activating receptors.14,15 Indeed, several therapeutic monoclonal antibodies (mAbs), such as rituximab (Rituxan®), cetuximab (Erbitux®), and trastuzumab (Herceptin®) based on this mechanism are in wide use and have shown the significant therapeutic potential of NK cells16–19(Table 1).

Table 1.

Overview of pre-clinical or clinical trials based on NK cells and MAbs.

Treatment Condition or disease Target Reference or clinical trial
Rituximab-RLIb B-cell malignancies CD20 Vincent et al.20
c.60C3-RLIc Neuroblastoma cells. GD2 Vincent et al.21
Hul4.18-IL2 Neuroblastoma cells GD2 Shusterman et al.22
Delgado et al.23
KM2812 Prostate cancer cells PSMA Sugimoto et al.24
293C3-SDIE Acute myeloid leukemia cells CD133 Koerner et al.25
Riegg et al.26
7C6 mAb Cholangiocarcinoma NKG2D receptor Oliviero et al.27
Rituximab Non-Hodgkin’s lymphoma CD20 Cartron et al.28
Obinutuzumab Chronic lymphocytic leukemia CD20 Goede et al.29
Capuano et al.30
Dinituximab High-risk neuroblastoma GD2 Yu et al.31
Trastuzumab Breast cancer HER2 Junttila et al.32
Cetuximab Metastatic colorectal cancer and head and neck cancer EGFR Juliá et al.33
Hu3F8 GD2+ tumors GD2 NCT01419834
NCT01662804
NCT01757626

A recently revisited approach is to redirect NK cells to tumor cells by targeting one tumor antigen (e.g., CD19) and one CD16A molecule simultaneously, similar to the BiTE® format which consists of a target on the surface of cancer cell and CD3 on the surface of T-cells. Some groups have named these bispecific antibodies “bispecific killer cell engagement (BiKEs).”10 These BiKEs work by bringing NK cells and target cells into similar proximity, forming cytolytic synapses that lead to tumor cell death. Tri-specific killer engagers (TriKEs) are trivalent molecules that redirect the killing of NK cells against different tumor antigens and enhance NK cell functions through the addition of a third single-chain variable fragment (scFv) targeting another cancer target (e.g., CD22) or immune cell-stimulating cytokines, like interleukin (IL)-15.34

Although these molecules vary in structure, they all have small molecular weights and structural domains that bind to target-associated antigens (TAAs) and NK cell-activated receptors, including CD16A, NKG2D, NKp30, and NKp46.35 In this review, we discuss the emergence of Bi- and trispecific NK cell engagers as a new generation of immunotherapy, as well as the advantages, obstacles, and development prospects for future clinical applications.

Bi-specific and tri-specific NK cell engagers mechanism of action

As previously mentioned, NK cells are cytotoxic lymphocytes of the innate immune system and play an important role in tumor immune surveillance. One strategy to re-direct NK cells to cancer cells is based on tandem scFvs, like bi- or tri-specific NK cell engagers,36 which can target both scFv binding CD16A on NK cells and one or two TAA on various solid tumor types (e.g., CD19, CD133, EpCAM, or B7-H3) (Figure 1).37,38 Alternatively, two scFvs can be linked by a modified human IL-15 cross-linker to produce TriKEs that promote the persistence and activation of NK cells in vivo. For example, AFM13, a bispecific antibody targeting CD30 and CD16A, mediates in vivo ADCC and NK cell retention activity, with CD30 being a prominent marker for relapsed/refractory (R/R) Hodgkin lymphoma (HL), increased cytotoxicity.39,40 In addition, revisiting the CD133-expressing cancer stem cells (CSCs), a TriKE expressing IL-15, named 1,615,133, showed in vitro the activity of NK cell efficient cytotoxicity and proliferation.41

Figure 1.

Figure 1.

Design and function of BiKEs and TriKEs (a) BiKEs are consisting of individual heavy (VH) and light (VL) chains of the variable region of each different antibody with the VH and VL structural domains linked via short flexible peptide junctions to prevent dissociation. These scFv are specific for CD16 on NK cells and tumor antigens on tumor cells. (b) TriKEs of the same design including the cytokine, IL-15, are included as a linker between the two scFv components. (c) shown here is a mechanism diagram. BiKEs and TriKEs redirect NK cells to target tumor cells through a tumor-specific target on the one hand and to CD16A-positive NK cells on the other hand, forming lytic immune synapses with cytokines, granzymes, and perforins release, ultimately leading to tumor cell death.

BsAbs can be divided into IgG-like and non-IgG-like based on their structure for the presence of the Fc region of the IgG-like BsAbs with Fc-mediated antibody effect function, such as ADCC. In addition, the retention of the constant region is conducive to the purification of the antibody, improves the solubility and stability of the antibody drug, and in vivo has a higher half-life.42 Although NK cells can also be recruited, what needs to be solved in the process of practical project development is the problem of antibody chain mismatch, which often leads to low efficiency of double antibody industrialization and high impurity proteins. In contrast, non-IgG-like BsAbs like BiKEs or TriKEs are less immunogenic and relatively safer because of their relatively small molecular weight (55-60 kDa).43 Although it has a short half-life, BiKEs have high tumor tissue permeability. In addition, NK cells are safer than T cells compared to BiTEs, and NK cell infusion does not induce graft versus host disease; therefore, adverse events with BiKEs and TriKEs are usually less severe.44 In summary, these BiKEs or TriKEs showed promising translational potential.

Examples of bi-specific and tri-specific NK cell engagers

Hematological malignancies

BiKE has demonstrated promising treatment by targeting specific hematopoietic cell lines and their corresponding malignant counterpart antigens, like CD19, considered a key component of the B-cell receptor complex, as well as CD33 highly expressed by myeloid cells and acute myeloid leukemia (AML) cells.45 For example, CD16 × CD19 BiKEs showed the enhancing effect of in vitro NK cell-mediated cytotoxicity.46 In a study of AML and biphenotypic, ALL blasts from pediatric patients with primary CD33+ blasts, CD16 × CD33 BiKEs elicited the inhibitory effect of KIR signaling and enhanced NK cell-mediated lysis of AML blasts.47 Moreover, CD16 × CD33 BiKEs were also demonstrated to redirect NK cells to myelodysplastic syndrome (MDS), expressing CD33 in pre-clinical studies. Gleason MK et al. published the results about 67 patients with MDS who could be assessed for a response by a pre-clinical study against high-risk MDS to express CD33.48 In this treatment, BiKEs played an essential role in targeting MDS and other myeloid malignancies and significantly enhanced degranulation and cytokine production.

TriKEs serve as an alternative and innovative version, and 161533 TriKEs (GTB-3550), including an anti-CD16 and an anti-CD33 scFv with IL-15 presented as a linker, have anti-leukemic activity in patients with relapsed and/or refractory AML or MDS, according to results from pre-clinical studies and a phase I/II clinical trial (NCT03214666).49,50 CD19 as an antigen target for Trike technology was confirmed in studies assessing its expression in patients with primary chronic lymphocytic leukemia (CLL), and the cytotoxicity of the 161,519 TriKE (CD16 × CD19 × IL-15 TriKEs) showed better results in the lysis of CLL cells in vitro, when compared with that of rituximab or those treated with 1619 BiKE (CD16 × CD19 BiKE).51 Additionally, multiple clinical trials are currently underway for BiKEs or TriKEs treatment of R/R HL patients (e.g., NCT02321592, NCT01221571, NCT03192202, and NCT04074746),39,40,52 or in combination with pembrolizumab (NCT02665650), or for BCMA+ relapsed or refractory multiple myeloma (known as RO7297089, NCT04434469) (Table 2)53–55 are currently underway.

Table 2.

Overview of clinical current trials of bi- and tri-specific NK cell engager.

Drug Condition or disease Targeted antigen ClinicalTrials.gov Identifier Phase status
AFM13
(anti-CD30/CD16A BiKE)
Lymphoma, T-Cell, Cutaneous CD30 NCT03192202 Phase 1/2
Completed
Hodgkin Lymphoma CD30 NCT01221571 Phase 1
Completed
R/R CD30-positive T-cell Lymphoma,
Transformed Mycosis Fungoides
CD30 NCT04101331 Phase 2
Active, not recruiting
R/R Hodgkin lymphoma CD30 NCT02321592 Phase 2
Completed
The combination therapy of AFM13 and pembrolizumab (MK-3475) R/R Hodgkin lymphoma CD30 NCT02665650 Phase 1
Completed
The combination therapy of AFM13 and UCB- derived NK cell therapy R/R Hodgkin or Non-Hodgkin Lymphomas CD30 NCT04074746 Phase 1/2
Recruiting
AFM24 (anti-EGFR/CD16A BiKE) Advanced Solid Tumor EGFR NCT04259450 Phase 1/2
Recruiting
The combination therapy of AFM24 and SNK01 Advanced or metastatic EGFR-expressing cancers EGFR NCT05099549 Phase 1/2
Recruiting
The combination therapy of AFM24 and atezolizumab Advanced Solid Tumor EGFR NCT05109442 Phase 1/2
Recruiting
AFM26 (anti-BCMA/CD16A BiKE) R/R Multiple Myeloma BCMA NCT04434469 Phase 1
Completed
GTB-3550 (anti-CD16/IL-15/CD33 TriKETM) High-risk MDS and R/R AML CD33 NCT03214666 Phase 1/2
Recruiting
The combination therapy of DF1001 (anti-HER2 TriNKETM) and Nivolumab HER2+ solid tumors HER2 NCT04143711 Phase 1/2
Recruiting

*All listed clinical studies are sourced from ClinicalTrials.gov. BCMA B cell maturation antigen, R/R relapsed or refractory, UCB umbilical cord blood, EGFR epithelial growth factor receptor, MDS myelodysplastic syndrome, AML acute myeloid leukemia, HER2 human epidermal growth factor receptor 2.

Solid tumor

One of the most significant issues in the area of NK cell-engaging therapies is whether these treatments are effective outside of hematological malignancies. AFM24 is another novel bispecific, IgG1-scFv fusion antibody developed by Affimed, based in Heidelberg, Germany, that mediates tumor cell depletion by targeting EGFR on tumor cells and CD16A on, NK cells, i.e., innate immune cells and macrophages.56 A pre-clinical investigation has emphasized robustly enhanced killing of tumor cells expressing varying levels of EGFR, regardless of KRAS/BRAF mutational status both in vitro and in vivo with AFM24.57 Importantly, there was the absence of systemic immune activation or any other toxicity found in the cynomolgus monkey model in vivo; thus, the promising potential clinic trial (NCT04259450) of AFM24 is currently enrolling patients (Table 2).

The epithelial cell-adhesion molecule (EpCAM) is a mutated TAA that is expressed in various types of carcinomas like prostate cancers, colon, breast, and ovary. In 2013, Vallera DA and colleagues showed strong preclinical evidence that CD16 × EpCAM BiKEs were previously presented in vitro with significantly higher NK cell cytotoxicity at low effect target ratios.35,38 A sufficient NK cell proliferative capacity seems to be a further prerequisite for NK cell cytolytic activity and correlates with the level of inflammatory cytokine production. Thus, to enhance the efficacy of specific engagers, an EpCAM TriKE that incorporates an IL-15-crosslinker into a BiKE construct relative to CD16 × EpCAM BiKEs is preferable.58 These authors proposed that the CD16 × EpCAM × IL-15 TriKEs markedly enhance its therapeutic potential and NK cell proliferation in a manner like exogenous IL-15 although its intramolecular conformation. In line with the TriKEs platform, B7-H3 expressed by prostate cancer, ovarian cancer, and lung cancer,59 and tumor endothelial marker 8 (TEM8) expressed by lung cancer is also currently being tested preclinically.60 These data all suggest that TriKEs may be a valuable NK cell-based immunotherapy for patients with various solid tumors.

Taken together, advances in BiKEs and TriKEs technology could substantially improve the treatment of hematologic and solid tumors.

Limitations of bi-specific and tri-specific NK cell engagers

NK cell engagers are considered less immunogenic and relatively safer than Fc-mediated ADCC compared to conventional IgG because of their relatively small molecular size.61,62 In contrast to antibody-drug conjugates (ADCs), which utilize NK cell redirection for cytotoxic effects and are more dependent on the host’s immune system, they have a better safety profile.63,64

A deeper understanding of the mechanisms of structural selection, tumor escape, resistance, and toxicity confronted in Bi-specific and Tri-specific NK cell engagers may identify opportunities to enhance the effectiveness of NK cell-based therapies and the long-term outcomes of cancer patients. Notably, to compensate for these shortcomings, researchers are currently performing several different modifications to improve their efficacy (Figure 2).

Figure 2.

Figure 2.

Different formats of NK cell engagers. A BiKE molecule consisting of two single chain fragment variables (scFvs), including one targeting a target-associated antigen (TAA) and (a) CD16A, (b) NKG2D, (c) NKp30 on NK cells; a TriKE adding (d) an interleukin (IL)-15-crosslinker or (e) another TAA against tumor cells; (f) a Tetraspecific killer engager (TetraKE) comprising three scFvs and an IL-15-crosslinker; (g) BiKEs and the PD-1/PD-L1 blockade combination therapy; (h) in addition, oncolytic viruses (OVs) have been modified for NK cell engagers, forming OVs-armed BiKEs.

Selection of structure

It is generally known that bsAbs can simultaneously engage two different antigens together. This increased functionality may effectively lead to fewer side effects and fewer injections compared to conventional mAbs, which can only bind a single epitope.65,66 It is important to understand how to design and stabilize bsAbs in vivo for developing effective NK cell-mediated anti-cancer therapeutics. The design of these structures is a complex process and with the rapid development of protein engineering technology in recent years, researchers have become increasingly interested in nanobodies.

Nanobodies are known to be univariate immunoglobulin structural domains originating from naturally occurring heavy chain antibodies (hcAbs) in camels.67,68 It has been shown that nanobody-based BiKEs (nano-BiKEs, approx. 30 kDa) are smaller in size and more soluble than scFv-based BiKEs (approx. 50 kDa), resulting in superior stability and greater penetration of tissues in vivo.69 Antibodies binding selectively to CD16A have an essential role in improving treatment efficacy and decreasing off-target toxicity. Nikkhoi et al. designed a functional bispecific killer cell engager (BiKE) with high affinity and specificity/selectivity for the CD16A receptor using anti-CD16A and anti-HER2 nanobodies.70 This BiKE induced higher ADCC on ovarian and breast cancer cells compared to trastuzumab. Besides, Van Faassen H and his colleagues constructed bi-specific sdAb-sdAb and sdAb-scFv BiKEs bound by fusion with tumor-specific sdAb or scFv (anti-CD19, anti-HER2, or anti-EGFR) by isolating camel anti-CD16 monoclonal antibodies. The experimental results suggest that the smaller size of sdAbs may confer better tumor penetration to sdAb-based BiKEs and TriKEs compared to full-length IgGs, Fabs, or scFv; also, the fact that the nanobodies have only heavy chains without the need for light-heavy chain pairing makes them suitable for modular assemblies, allowing for the easy construction of multispecific and multivalent molecules.68

Despite the advantages of hcAbs over full-length IgG, Fabs, or scFvs in the design of NK cell engagers, their smaller size which is below the kidney filtration barrier leads to a shorter serum half-life in vivo. To prolong in vivo half-life, several research groups have designed nano-BiKE with an extended half-life (HLE nano-BiKE), that is, nano-based BiKE is fused into albumin-specific nanobody.71,72 Recently, Hambach J et al. reported that CD38-specific HLE-nano-BiKEs recognizing three distinct and non-overlapping epitopes of CD38-induced NK92 hCD16-mediated cell lysis more efficiently than the conventional CD38-specific antibody daratumumab (ADCC) and induced in vitro CD38-positive myeloma cell line cytotoxicity.73 These nanobody-trimers bind CD38 on myeloma cells (N-terminal nanobody), bind and activate NK cells via targeting CD16 with central nanobodies, and bind to albumin via a C-terminal nanobody. Thus, the whole trimer not only targets the antigen and activates NK cells but also extends the half-life of the construct.

In general, different NK cell engagers have their structural advantages and shortcomings, and the selection and development of next-generation NK cell engagers are still the centers of future research.

Tumor drug resistance

Antigen escape

The first step in generating BiKEs is the identification of suitable tumor cell-specific antigens. Ideally, the target antigen needs to be clearly and predominantly expressed in tumor cells, rather than in normal cells to prevent nonspecific toxicity, and they are closely associated with malignant phenotypes or signaling pathways to prevent immune tolerance caused by antigen mutations. However, most cancer targets do not meet these criteria.10 CD19, the most representative TAA, is restricted to specific immune cell lineages and is a key antigenic target for BiKEs-based immunotherapy. It has been reported that anti-CD19 × CD3 BiTEs in B-ALL therapy result in loss of extracellular structural domains due to mutations in CD19, conformational changes, and ultimately therapeutic failure.74,75 For this reason, some researchers have come up with the idea of TriKEs. It can simultaneously recognize multiple antigens on the surface of cancer target cells and is expected to improve tumor targeting specificity and exert better anti-tumor efficacy through synergistic signal inhibition and accelerated tumor cell degradation. For example, generating a tri-specific antibody, which recognizes both CD33 and CD123 on AML cancer cells.76 Similarly, Schmohl et al. recently developed a tetraspecific killer engager (TetraKE), named 1615EpCAM133, which targets an anti-CD16 scFv on NK cells, an anti-EpCAM scFv on carcinoma cells and an anti-CD133 scFv on cancer stem cells to promote dual-antigen ADCC.77 Importantly, although an IL-15-crosslinker enhanced NK cell-related superior efficacy and cytokine production, there is no evidence of excessive cytokine release.

Furthermore, the search for tumor-specific antigens (TSAs) and the identification of the associated intrinsic mechanisms is challenging but imminent, paving the way for improved anti-tumor strategies.78 Although most immunotherapies target TAAs as they are usually overexpressed in a group of tumor types, numerous clinical reports of TAA-based therapies have shown their potential damage to normal tissues and are accompanied by unsatisfactory clinical outcomes.78 However, most TSAs are derived from non-synonymous somatic mutations that are completely absent in normal cells by contrast. It can be selectively expressed in tumor cells, providing an opportunity for personalized TSA-dependent immunotherapy due to differences between individuals.10,79

The suppression of NK cells is another explanation for tumor escape. Thus, another attractive option is the addition of scFvs that block checkpoint receptors such as PD-1, KIR, NKG2A, or TIGIT through structural improvements to TriKEs to bypass checkpoint blockade and further drive NK-mediated anti-tumor responses.80 More recently, one study has shown that the use of a PD-1 blocking antibody (CT-011) enhances target cell killing and cytokine production by NK cells in response to PD-L1 expressing primary multiple myeloma cells or myeloma cell lines in vitro.81

Immunosuppressive microenvironment

The ability of NK cells to kill tumor cells is greatly hampered in the face of sensitivity to the multiple immunosuppressive mechanisms active within TME although their activity in tumor growth control. It could limit the persistence of NK cells and inhibit their cytotoxic functions, by producing immune-suppressive cells and secreting factors, IL-6, IL-10, prostaglandin E2 (PGE2), and indoleamine 2,3-dioxygenase (IDO).82,83 To benefit patients, several potential strategies are a focus of future research.

ADAM17 (a disintegrin and metalloprotease 17), an enzyme that cleaves the outer membrane of L-selectin, promotes the decay of NK cell function by cleaving CD16 from the cell surface and is another mechanism by which MDSC suppresses antitumor immunity.84,85 Wiernik et al. found that combining CD16 × 33 BiKEs with an ADAM17 inhibitor increased both NK cell toxicity and cytokine secretion, preventing the failure of NK cell therapy due to the immunosuppressive effects of the TME.37

Generally, while the current generation of TriKEs triggers NK responses through CD16 ligation and cytokine signaling, other components can be added to enhance the NK cell anti-tumor response. For example, another mechanism by which NK cells evade tumors is by shedding soluble NKG2D ligands.86 Therefore, ligand expression on the tumor surface is reduced, decreasing the susceptibility to NK-mediated tumor cell killing. Tregs have been reported to downregulate NKG2D and NCR NKp30 expression on NK cells through cell–cell contacts, membrane-bound TGF-β, and soluble TGF-β.87 In the future, including an scFv blockade of TGF-β to reduce negative signaling in the tumor microenvironment might enhance NK cell-mediated tumor killing based on a bispecific antibody platform.

Pharmacokinetics and toxicity

Although a variety of NK cell engagers have demonstrated good in vitro targeting capabilities, their safety including preclinical studies to clinical trials and finally to the formal approval of the product for the treatment of tumors is a key point that must be considered and evaluated. BiKEs or BiTEs have a short half-life in serum, thus one of the drawbacks of BiKEs or BiTEs is the need for continuous intravenous infusion. The development of arming oncolytic viruses (OVs) with a therapeutic transgene encoding BiTE is an attractive platform for bsAbs delivery.88 OVs enter cells by interacting with specific surface receptors that are highly expressed in cancer cells. Viral replication is achieved, targeting tumor cells while leaving normal cells undamaged. For example, Fajardo et al. engineered the adenovirus ICOVIR-15K to express antibodies against BiTE (cBiTE) targeting EGFR, and ICOVIR-15K-cBiTE induced the persistence and accumulation of tumor infiltration in vivo.89 In another similar construct, Freedman et al. modified an anti-EpCAM × CD3 BiTE-armed oncolytic group B adenovirus EnAdenotucirev (EnAd) that could enhance infiltration and activation of CD4+ and CD8+ T cells, thereby enhancing T cell-mediated tumor killing in clinical tissue biopsies.90 Although no relevant engineered BiKE-producing OVs are currently undergoing preclinical evaluation, future applications are extremely valuable.

In addition, TriKEs have also been reported to potentially activate systemic immune responses by triggering cytokine cascades, leading to a risk of systemic toxicity.91 Thus, ongoing development of treatments is imperative, both in terms of alternative and potentially safer ways of detecting cytokine pathway activation.

Conclusions and future directions

The utilization of immunotherapy to target malignant cells has emerged as a novel approach for the treatment of various cancers. Especially, NK cells serve as a unique set of anti-tumor response agents with non-MHC-restricted cytotoxicity, and cytokine production. The success of preclinical studies by multiple groups and the results of ongoing clinical studies with BiKEs have demonstrated specific killing activity. However, in the long term, a large proportion of patients will not benefit from immunotherapy based on bi-specific and tri-specific NK cell engagers.

The major challenge is the treatment of solid tumors involving immunosuppressive TME. Since inhibitory receptors, downregulation of activated receptors, molecular checkpoints, and inhibitors of the tumor environment all lead to immune escape of NK cells, combinations of immune checkpoint inhibitors targeting the synergistic response of NK cells are being attempted. The observation that the antitumor activity of a CD47 × CD19 bispecific antibody (NI-1701) was improved in the presence of NK cells by CD47/SIRPα blockade in vivo,92 and the evidence that CD47 antibody treatment could thereby promote activation of systemic and tumor-associated NK cells.93

Another important aspect of NK cell therapy is to increase the lifespan of NK cells, and TriKEs provide another choice for NK cell persistence in vivo regardless of genetic modification. However, insight is needed into the biological effects of TriKEs on NK cells, including their activation, maturation, and depletion; their activity after cessation of administration; and their tendency to develop into a long-life memory cell population.

For example, a novel multifunctional molecule ( ANKET4 )based on the versatile NK cell engager platform is available in preclinical development that binds an Interleukin-2 (IL-2) variant that enhances NK cell activation and proliferation through cis-binding of NKp46, CD16a, and IL-2 Rβ (CD122) without binding to IL-2 Rα (CD25) in Tregs or endothelial cells.94 In testing with non-human primates, targeting CD20 to ANKET4 showed that ANKET4 not only has no clinical signs of toxicity but also severe depletion of circulating B cells with a favorable safety profile. During clinical trials, as more bsAbs and cellular therapies are investigated, understanding of the optimal design and/or construct for targeting specific tumor types is increasing. We expect that as more NK cell-based immunotherapy is applied in clinical treatment in the future, more and more patients will benefit from painful diseases and endless treatment processes.

Funding Statement

This work was supported by the Health Research Project of Huai’an City in 2020, General Program, Jiangsu, China. Grant number [33].

Disclosure statement

No potential conflict of interest was reported by the author(s).

Author contributions

Tian Huan and Bugao Guan collected the data and wrote the manuscript. Hongbo Li, Xiu Tu, and Chi Zhang provided substantial contributions to revise the paper critically for important intellectual content. Bugao Guan and Bin Tang contributed to the scientific discussion and critical review of the manuscript. All authors contributed to the article and approved the submitted version.

References

  • 1.Chames P, Van Regenmortel M, Weiss E, Baty D.. Therapeutic antibodies: successes, limitations and hopes for the future. Br J Pharmacol. 2009;157(2):220–10. doi: 10.1111/j.1476-5381.2009.00190.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Acheampong DO, Adokoh CK, Ampomah P, Agyirifor DS, Dadzie I, Ackah FA, Asiamah EA. Bispecific antibodies (bsAbs): promising immunotherapeutic agents for cancer therapy. Protein Pept Lett. 2017;24(5):456–65. doi: 10.2174/0929866524666170120095128. [DOI] [PubMed] [Google Scholar]
  • 3.Henricks LM, Schellens JH, Huitema AD, Beijnen JH. The use of combinations of monoclonal antibodies in clinical oncology. Cancer Treat Rev. 2015;41(10):859–67. doi: 10.1016/j.ctrv.2015.10.008. [DOI] [PubMed] [Google Scholar]
  • 4.Felices M, Lenvik TR, Kodal B, Lenvik AJ, Hinderlie P, Bendzick LE, Schirm DK, Kaminski MF, McElmurry RT, Geller MA, et al. Potent cytolytic activity and specific IL15 delivery in a second-generation trispecific killer engager. Cancer Immunol Res. 2020;8(9):1139–49. doi: 10.1158/2326-6066.CIR-19-0837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Allen C, Zeidan AM, Bewersdorf JP. BiTEs, DARTS, BiKEs and TriKEs—are antibody based therapies changing the future treatment of AML? Life (Basel). 2021;11(6):11. doi: 10.3390/life11060465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Imai K, Matsuyama S, Miyake S, Suga K, Nakachi K. Natural cytotoxic activity of peripheral-blood lymphocytes and cancer incidence: an 11-year follow-up study of a general population. Lancet. 2000;356(9244):1795–9. doi: 10.1016/S0140-6736(00)03231-1. [DOI] [PubMed] [Google Scholar]
  • 7.Slattery K, Woods E, Zaiatz-Bittencourt V, Marks S, Chew S, Conroy M, Goggin C, MacEochagain C, Kennedy J, Lucas S, et al. TGFβ drives NK cell metabolic dysfunction in human metastatic breast cancer. J Immunother Cancer. 2021;9(2):e002044. doi: 10.1136/jitc-2020-002044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Greene S, Robbins Y, Mydlarz WK, Huynh AP, Schmitt NC, Friedman J, Horn LA, Palena C, Schlom J, Maeda DY, et al. Inhibition of MDSC trafficking with SX-682, a CXCR1/2 inhibitor, enhances NK-Cell immunotherapy in head and neck cancer models. Clin Cancer Res. 2020;26(6):1420–31. doi: 10.1158/1078-0432.CCR-19-2625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Ziblat A, Iraolagoitia XLR, Nunez SY, Torres NI, Secchiari F, Sierra JM, Spallanzani RG, Rovegno A, Secin FP, Fuertes MB, et al. Circulating and tumor-infiltrating NK cells from clear cell renal cell carcinoma patients exhibit a predominantly inhibitory phenotype characterized by overexpression of CD85j, CD45, CD48 and PD-1. Front Immunol. 2021;12:681615. doi: 10.3389/fimmu.2021.681615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Li H, Er Saw P, Song E. Challenges and strategies for next-generation bispecific antibody-based antitumor therapeutics. Cell Mol Immunol. 2020;17(5):451–61. doi: 10.1038/s41423-020-0417-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Karre K. NK cells, MHC class I molecules and the missing self. Scand J Immunol. 2002;55(3):221–8. doi: 10.1046/j.1365-3083.2002.01053.x. [DOI] [PubMed] [Google Scholar]
  • 12.Bryceson YT, March ME, Ljunggren HG, Long EO. Activation, coactivation, and costimulation of resting human natural killer cells. Immunol Rev. 2006;214(1):73–91. doi: 10.1111/j.1600-065X.2006.00457.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Lopez-Larrea C, Suarez-Alvarez B, Lopez-Soto A, Lopez-Vazquez A, Gonzalez S. The NKG2D receptor: sensing stressed cells. Trends Mol Med. 2008;14(4):179–89. doi: 10.1016/j.molmed.2008.02.004. [DOI] [PubMed] [Google Scholar]
  • 14.Wallin RP, Screpanti V, Michaelsson J, Grandien A, Ljunggren HG. Regulation of perforin-independent NK cell-mediated cytotoxicity. Eur J Immunol. 2003;33(10):2727–35. doi: 10.1002/eji.200324070. [DOI] [PubMed] [Google Scholar]
  • 15.Farag SS, Caligiuri MA. Human natural killer cell development and biology. Blood Rev. 2006;20(3):123–37. doi: 10.1016/j.blre.2005.10.001. [DOI] [PubMed] [Google Scholar]
  • 16.Dixon KJ, Wu J, Walcheck B. Engineering anti-tumor monoclonal antibodies and Fc receptors to enhance ADCC by human NK cells. Cancers Basel. 2021;13(2):312. doi: 10.3390/cancers13020312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Frampton JE. Rituximab: a review in pemphigus vulgaris. Am J Clin Dermatol. 2020;21(1):149–56. doi: 10.1007/s40257-019-00497-9. [DOI] [PubMed] [Google Scholar]
  • 18.Mazzarella L, Guida A, Curigliano G. Cetuximab for treating non-small cell lung cancer. Expert Opin Biol Ther. 2018;18(4):483–93. doi: 10.1080/14712598.2018.1452906. [DOI] [PubMed] [Google Scholar]
  • 19.Keam SJ. Trastuzumab deruxtecan: first approval. Drugs. 2020;80(5):501–8. doi: 10.1007/s40265-020-01281-4. [DOI] [PubMed] [Google Scholar]
  • 20.Vincent M, Teppaz G, Lajoie L, Sole V, Bessard A, Maillasson M, Loisel S, Béchard D, Clémenceau B, Thibault G, et al. Highly potent anti-CD20-RLI immunocytokine targeting established human B lymphoma in SCID mouse. MAbs. 2014;6(4):1026–37. doi: 10.4161/mabs.28699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Vincent M, Bessard A, Cochonneau D, Teppaz G, Sole V, Maillasson M, Birklé S, Garrigue-Antar L, Quéméner A, Jacques Y, et al. Tumor targeting of the IL-15 superagonist RLI by an anti-GD2 antibody strongly enhances its antitumor potency. Int J Cancer. 2013;133(3):757–65. doi: 10.1002/ijc.28059. [DOI] [PubMed] [Google Scholar]
  • 22.Shusterman S, London WB, Gillies SD, Hank JA, Voss SD, Seeger RC, Reynolds CP, Kimball J, Albertini MR, Wagner B, et al. Antitumor activity of hu14.18-IL2 in patients with relapsed/refractory neuroblastoma: a children’s oncology group (COG) phase II study. J Clin Oncol. 2010;28(33):4969–75. doi: 10.1200/JCO.2009.27.8861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Delgado DC, Hank JA, Kolesar J, Lorentzen D, Gan J, Seo S, Kim K, Shusterman S, Gillies SD, Reisfeld RA, et al. Genotypes of NK cell KIR receptors, their ligands, and Fcγ receptors in the response of neuroblastoma patients to Hu14.18-IL2 immunotherapy. Cancer Res. 2010;70(23):9554–61. doi: 10.1158/0008-5472.CAN-10-2211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Sugimoto Y, Hirota M, Yoshikawa K, Sumitomo M, Nakamura K, Ueda R, Niwa R, Suzawa T, Yamasaki M, Shitara K, et al. The therapeutic potential of a novel PSMA antibody and its IL-2 conjugate in prostate cancer. Anticancer Res. 2014;34(1):89–97. [PubMed] [Google Scholar]
  • 25.Koerner SP, Andre MC, Leibold JS, Kousis PC, Kubler A, Pal M, Haen SP, Bühring H-J, Grosse-Hovest L, Jung G, et al. An Fc-optimized CD133 antibody for induction of NK cell reactivity against myeloid leukemia. Leukemia. 2017;31(2):459–69. doi: 10.1038/leu.2016.194. [DOI] [PubMed] [Google Scholar]
  • 26.Riegg F, Lutz MS, Schmied BJ, Heitmann JS, Queudeville M, Lang P, Jung G, Salih HR, Märklin M. An Fc-optimized CD133 antibody for induction of NK cell reactivity against B cell acute lymphoblastic leukemia. Cancers Basel. 2021;13(7):1632. doi: 10.3390/cancers13071632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Oliviero B, Varchetta S, Mele D, Pessino G, Maiello R, Falleni M, Tosi D, Donadon M, Soldani C, Franceschini B, et al. MICA/B-targeted antibody promotes NK cell–driven tumor immunity in patients with intrahepatic cholangiocarcinoma. Oncoimmunology. 2022;11(1):2035919. doi: 10.1080/2162402X.2022.2035919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Cartron G, Dacheux L, Salles G, Solal-Celigny P, Bardos P, Colombat P, Watier H. Therapeutic activity of humanized anti-CD20 monoclonal antibody and polymorphism in IgG Fc receptor FcγRIIIa gene. Blood. 2002;99(3):754–8. doi: 10.1182/blood.V99.3.754. [DOI] [PubMed] [Google Scholar]
  • 29.Goede V, Fischer K, Busch R, Engelke A, Eichhorst B, Wendtner CM, Chagorova T, de la Serna J, Dilhuydy M-S, Illmer T, et al. Obinutuzumab plus chlorambucil in patients with CLL and coexisting conditions. N Engl J Med. 2014;370(12):1101–10. doi: 10.1056/NEJMoa1313984. [DOI] [PubMed] [Google Scholar]
  • 30.Capuano C, Pighi C, Molfetta R, Paolini R, Battella S, Palmieri G, Giannini G, Belardinilli F, Santoni A, Galandrini R, et al. Obinutuzumab-mediated high-affinity ligation of FcγRIIIA/CD16 primes NK cells for IFNγ production. Oncoimmunology. 2017;6(3):e1290037. doi: 10.1080/2162402X.2017.1290037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Yu AL, Gilman AL, Ozkaynak MF, London WB, Kreissman SG, Chen HX, Smith M, Anderson B, Villablanca JG, Matthay KK, et al. Anti-GD2 antibody with GM-CSF, interleukin-2, and isotretinoin for neuroblastoma. N Engl J Med. 2010;363(14):1324–34. doi: 10.1056/NEJMoa0911123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Junttila TT, Parsons K, Olsson C, Lu Y, Xin Y, Theriault J, Crocker L, Pabonan O, Baginski T, Meng G, et al. Superior in vivo efficacy of afucosylated trastuzumab in the treatment of HER2-amplified breast cancer. Cancer Res. 2010;70(11):4481–9. doi: 10.1158/0008-5472.CAN-09-3704. [DOI] [PubMed] [Google Scholar]
  • 33.Julia EP, Mordoh J, Levy EM. Cetuximab and IL-15 promote NK and dendritic cell activation in vitro in triple negative breast cancer. Cells. 2020;9(7):1573. doi: 10.3390/cells9071573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Felices M, Lenvik TR, Davis ZB, Miller JS, Vallera DA. Generation of BiKEs and TriKEs to improve NK cell-mediated targeting of tumor cells. Methods Mol Biol. 2016;1441:333–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Phung SK, Miller JS, Felices M. Bi-specific and tri-specific NK cell engagers: the new avenue of targeted NK cell immunotherapy. Mol Diagn Ther. 2021;25(5):577–92. doi: 10.1007/s40291-021-00550-6. [DOI] [PubMed] [Google Scholar]
  • 36.Hodgins JJ, Khan ST, Park MM, Auer RC, Ardolino M. Killers 2.0: NK cell therapies at the forefront of cancer control. J Clin Invest. 2019;129(9):3499–510. doi: 10.1172/JCI129338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Wiernik A, Foley B, Zhang B, Verneris MR, Warlick E, Gleason MK, Ross JA, Luo X, Weisdorf DJ, Walcheck B, et al. Targeting natural killer cells to acute myeloid leukemia in vitro with a CD16 × 33 bispecific killer cell engager and ADAM17 inhibition. Clin Cancer Res. 2013;19(14):3844–55. doi: 10.1158/1078-0432.CCR-13-0505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Vallera DA, Zhang B, Gleason MK, Oh S, Weiner LM, Kaufman DS, McCullar V, Miller JS, Verneris MR. Heterodimeric bispecific single-chain variable-fragment antibodies against EpCAM and CD16 induce effective antibody-dependent cellular cytotoxicity against human carcinoma cells. Cancer Biother Radiopharm. 2013;28(4):274–82. doi: 10.1089/cbr.2012.1329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Rothe A, Sasse S, Topp MS, Eichenauer DA, Hummel H, Reiners KS, Dietlein M, Kuhnert G, Kessler J, Buerkle C, et al. A phase 1 study of the bispecific anti-CD30/CD16A antibody construct AFM13 in patients with relapsed or refractory Hodgkin lymphoma. Blood. 2015;125(26):4024–31. doi: 10.1182/blood-2014-12-614636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Bartlett NL, Herrera AF, Domingo-Domenech E, Mehta A, Forero-Torres A, Garcia-Sanz R, Armand P, Devata S, Izquierdo AR, Lossos IS, et al. A phase 1b study of AFM13 in combination with pembrolizumab in patients with relapsed or refractory Hodgkin lymphoma. Blood. 2020;136(21):2401–9. doi: 10.1182/blood.2019004701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Schmohl JU, Felices M, Oh F, Lenvik AJ, Lebeau AM, Panyam J, Miller JS, Vallera DA. Engineering of anti-CD133 Trispecific molecule capable of inducing NK expansion and driving antibody-dependent cell-mediated cytotoxicity. Cancer Res Treat. 2017;49(4):1140–52. doi: 10.4143/crt.2016.491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Ma J, Mo Y, Tang M, Shen J, Qi Y, Zhao W, Huang Y, Xu Y, Qian C. Bispecific antibodies: from research to clinical application. Front Immunol. 2021;12:626616. doi: 10.3389/fimmu.2021.626616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Lancman G, Richter J, Chari A. Bispecifics, trispecifics, and other novel immune treatments in myeloma. Hematol Am Soc Hematol Educ Program. 2020;2020(1):264–71. doi: 10.1182/hematology.2020000110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Kang J, Sun T, Zhang Y. Immunotherapeutic progress and application of bispecific antibody in cancer. Front Immunol. 2022;13:1020003. doi: 10.3389/fimmu.2022.1020003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Scheuermann RH, Racila E. CD19 antigen in leukemia and lymphoma diagnosis and immunotherapy. Leuk Lymphoma. 1995;18(5–6):385–97. doi: 10.3109/10428199509059636. [DOI] [PubMed] [Google Scholar]
  • 46.Gleason MK, Verneris MR, Todhunter DA, Zhang B, McCullar V, Zhou SX, Panoskaltsis-Mortari A, Weiner LM, Vallera DA, Miller JS, et al. Bispecific and trispecific killer cell engagers directly activate human NK cells through CD16 signaling and induce cytotoxicity and cytokine production. Mol Cancer Ther. 2012;11(12):2674–84. doi: 10.1158/1535-7163.MCT-12-0692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Reusing SB, Vallera DA, Manser AR, Vatrin T, Bhatia S, Felices M, Miller JS, Uhrberg M, Babor F. CD16xCD33 bispecific killer cell engager (BiKE) as potential immunotherapeutic in pediatric patients with AML and biphenotypic ALL. Cancer Immunol Immunother. 2021;70(12):3701–8. doi: 10.1007/s00262-021-03008-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Gleason MK, Ross JA, Warlick ED, Lund TC, Verneris MR, Wiernik A, Spellman S, Haagenson MD, Lenvik AJ, Litzow MR, et al. CD16xCD33 bispecific killer cell engager (BiKE) activates NK cells against primary MDS and MDSC CD33+ targets. Blood. 2014;123(19):3016–26. doi: 10.1182/blood-2013-10-533398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Vallera DA, Felices M, McElmurry R, McCullar V, Zhou X, Schmohl JU, Zhang B, Lenvik AJ, Panoskaltsis-Mortari A, Verneris MR, et al. IL15 Trispecific killer engagers (TriKE) make natural killer cells specific to CD33+ targets while also inducing persistence, in vivo expansion, and enhanced function. Clin Cancer Re. 2016;22(14):3440–50. doi: 10.1158/1078-0432.CCR-15-2710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Yun HD, Felices M, Vallera DA, Hinderlie P, Cooley S, Arock M, Gotlib J, Ustun C, Miller JS. Trispecific killer engager CD16xIL15xCD33 potently induces NK cell activation and cytotoxicity against neoplastic mast cells. Blood Adv. 2018;2(13):1580–4. doi: 10.1182/bloodadvances.2018018176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Cheng Y, Zheng X, Wang X, Chen Y, Wei H, Sun R, Tian Z, Sun H. Trispecific killer engager 161519 enhances natural killer cell function and provides anti-tumor activity against CD19-positive cancers. Cancer Biol Med. 2020;17(4):1026–38. doi: 10.20892/j.issn.2095-3941.2020.0399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Sasse S, Brockelmann PJ, Momotow J, Plutschow A, Huttmann A, Basara N, Koenecke C, Martin S, Bentz M, Grosse-Thie C, et al. AFM13 in patients with relapsed or refractory classical Hodgkin lymphoma: final results of an open-label, randomized, multicenter phase II trial. Leuk Lymphoma. 2022;63(8):1871–8. doi: 10.1080/10428194.2022.2095623. [DOI] [PubMed] [Google Scholar]
  • 53.Kakiuchi-Kiyota S, Ross T, Wallweber HA, Kiefer JR, Schutten MM, Adedeji AO, Cai H, Hendricks R, Cohen S, Myneni S, et al. A BCMA/CD16A bispecific innate cell engager for the treatment of multiple myeloma. Leukemia. 2022;36(4):1006–14. doi: 10.1038/s41375-021-01478-w. [DOI] [PubMed] [Google Scholar]
  • 54.Cai H, Kakiuchi-Kiyota S, Hendricks R, Zhong S, Liu L, Adedeji AO, Chan P, Schutten MM, Kamath AV, Ovacik MA, et al. Nonclinical pharmacokinetics, pharmacodynamics, and translational model of RO7297089, a novel anti-BCMA/CD16A bispecific tetravalent antibody for the treatment of multiple myeloma. AAPS J. 2022;24(6):100. doi: 10.1208/s12248-022-00744-8. [DOI] [PubMed] [Google Scholar]
  • 55.Plesner T, Harrison SJ, Quach H, Lee C, Bryant A, Vangsted A, Estell J, Delforge M, Offner F, Twomey P, et al. Phase I study of safety and pharmacokinetics of RO7297089, an anti-BCMA/CD16a bispecific antibody, in patients with relapsed, refractory multiple myeloma. Clin Hematol Int. 2023;5(1):43–51. doi: 10.1007/s44228-022-00023-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Ellwanger K, Reusch U, Fucek I, Wingert S, Ross T, Muller T, Schniegler-Mattox U, Haneke T, Rajkovic E, Koch J, et al. Redirected optimized cell killing (ROCK®): a highly versatile multispecific fit-for-purpose antibody platform for engaging innate immunity. MAbs. 2019;11(5):899–918. doi: 10.1080/19420862.2019.1616506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Wingert S, Reusch U, Knackmuss S, Kluge M, Damrat M, Pahl J, Schniegler-Mattox U, Mueller T, Fucek I, Ellwanger K, et al. Preclinical evaluation of AFM24, a novel CD16A-specific innate immune cell engager targeting EGFR-positive tumors. MAbs. 2021;13(1):1950264. doi: 10.1080/19420862.2021.1950264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Schmohl JU, Felices M, Taras E, Miller JS, Vallera DA. Enhanced ADCC and NK cell activation of an anticarcinoma bispecific antibody by genetic insertion of a modified IL-15 cross-linker. Mol Ther. 2016;24(7):1312–22. doi: 10.1038/mt.2016.88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Vallera DA, Ferrone S, Kodal B, Hinderlie P, Bendzick L, Ettestad B, Hallstrom C, Zorko NA, Rao A, Fujioka N, et al. NK-Cell-mediated targeting of various solid tumors using a B7-H3 tri-specific killer engager in vitro and in vivo. Cancers Basel. 2020;12(9):12. doi: 10.3390/cancers12092659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Kaminski MF, Bendzick L, Hopps R, Kauffman M, Kodal B, Soignier Y, Hinderlie P, Walker JT, Lenvik TR, Geller MA, et al. TEM8 tri-specific killer engager binds both tumor and tumor stroma to specifically engage natural killer cell anti-tumor activity. J Immunother Cancer. 2022;10(9):10. doi: 10.1136/jitc-2022-004725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Kapelski S, Cleiren E, Attar RM, Philippar U, Hasler J, Chiu ML. Influence of the bispecific antibody IgG subclass on T cell redirection. MAbs. 2019;11(6):1012–24. doi: 10.1080/19420862.2019.1624464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Perales-Puchalt A, Duperret EK, Yang X, Hernandez P, Wojtak K, Zhu X, Jung S-H, Tello-Ruiz E, Wise MC, Montaner LJ, et al. DNA-encoded bispecific T cell engagers and antibodies present long-term antitumor activity. JCI Insight. 2019;4(8):4. doi: 10.1172/jci.insight.126086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Li J, Stagg NJ, Johnston J, Harris MJ, Menzies SA, DiCara D, Clark V, Hristopoulos M, Cook R, Slaga D, et al. Membrane-proximal epitope facilitates efficient T cell synapse formation by anti-FcRH5/CD3 and is a requirement for myeloma cell killing. Cancer Cell. 2017;31(3):383–95. doi: 10.1016/j.ccell.2017.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Autio KA, Boni V, Humphrey RW, Naing A. Probody therapeutics: an emerging class of therapies designed to enhance on-target effects with reduced off-tumor toxicity for use in immuno-oncology. Clin Cancer Res. 2020;26(5):984–9. doi: 10.1158/1078-0432.CCR-19-1457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Wang Q, Chen Y, Park J, Liu X, Hu Y, Wang T, McFarland K, Betenbaugh MJ. Design and production of bispecific antibodies. Antibodies (Basel). 2019;8(3):43. doi: 10.3390/antib8030043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Brinkmann U, Kontermann RE. The making of bispecific antibodies. MAbs. 2017;9(2):182–212. doi: 10.1080/19420862.2016.1268307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Muyldermans S, Smider VV. Distinct antibody species: structural differences creating therapeutic opportunities. Curr Opin Immunol. 2016;40:7–13. doi: 10.1016/j.coi.2016.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.van Faassen H, Jo DH, Ryan S, Lowden MJ, Raphael S, MacKenzie CR, van Faassen H, Lee S-H, Hussack G, Henry KA. Incorporation of a novel CD16-specific single-domain antibody into multispecific natural killer cell engagers with potent ADCC. Mol Pharm. 2021;18(6):2375–84. doi: 10.1021/acs.molpharmaceut.1c00208. [DOI] [PubMed] [Google Scholar]
  • 69.Bannas P, Hambach J, Koch-Nolte F. Nanobodies and nanobody-based human heavy chain antibodies as antitumor therapeutics. Front Immunol. 2017;8:1603. doi: 10.3389/fimmu.2017.01603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Nikkhoi SK, Li G, Eleya S, Yang G, Vandavasi VG, Hatefi A. Bispecific killer cell engager with high affinity and specificity toward CD16a on NK cells for cancer immunotherapy. Front Immunol. 2022;13:1039969. doi: 10.3389/fimmu.2022.1039969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Terryn S, Francart A, Rommelaere H, Stortelers C, Van Gucht S, Rupprecht CE. Post-exposure treatment with anti-rabies VHH and vaccine significantly improves protection of mice from lethal rabies infection. PLoS Negl Trop Dis. 2016;10(8):e0004902. doi: 10.1371/journal.pntd.0004902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Zorzi A, Linciano S, Angelini A. Non-covalent albumin-binding ligands for extending the circulating half-life of small biotherapeutics. Medchemcomm. 2019;10(7):1068–81. doi: 10.1039/C9MD00018F. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Hambach J, Fumey W, Stahler T, Gebhardt AJ, Adam G, Weisel K, Koch-Nolte F, Bannas P. Half-life extended nanobody-based CD38-specific bispecific killercell engagers induce killing of multiple myeloma cells. Front Immunol. 2022;13:838406. doi: 10.3389/fimmu.2022.838406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Sotillo E, Barrett DM, Black KL, Bagashev A, Oldridge D, Wu G, Sussman R, Lanauze C, Ruella M, Gazzara MR, et al. Convergence of acquired mutations and alternative splicing of CD19 enables resistance to CART-19 immunotherapy. Cancer Discov. 2015;5(12):1282–95. doi: 10.1158/2159-8290.CD-15-1020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Braig F, Brandt A, Goebeler M, Tony HP, Kurze AK, Nollau P, Bumm T, Böttcher S, Bargou RC, Binder M, et al. Resistance to anti-CD19/CD3 BiTE in acute lymphoblastic leukemia may be mediated by disrupted CD19 membrane trafficking. Blood. 2017;129(1):100–4. doi: 10.1182/blood-2016-05-718395. [DOI] [PubMed] [Google Scholar]
  • 76.Dimasi N, Fleming R, Hay C, Woods R, Xu L, Wu H, Gao C. Development of a trispecific antibody designed to simultaneously and efficiently target three different antigens on tumor cells. Mol Pharm. 2015;12(9):3490–501. doi: 10.1021/acs.molpharmaceut.5b00268. [DOI] [PubMed] [Google Scholar]
  • 77.Schmohl JU, Felices M, Todhunter D, Taras E, Miller JS, Vallera DA. Tetraspecific scFv construct provides NK cell mediated ADCC and self-sustaining stimuli via insertion of IL-15 as a cross-linker. Oncotarget. 2016;7(45):73830–44. doi: 10.18632/oncotarget.12073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Zhou S, Liu M, Ren F, Meng X, Yu J. The landscape of bispecific T cell engager in cancer treatment. Biomark Res. 2021;9(1):38. doi: 10.1186/s40364-021-00294-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Liu XS, Mardis ER. Applications of immunogenomics to cancer. Cell. 2017;168(4):600–12. doi: 10.1016/j.cell.2017.01.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Davis ZB, Vallera DA, Miller JS, Felices M. Natural killer cells unleashed: checkpoint receptor blockade and BiKE/TriKE utilization in NK-mediated anti-tumor immunotherapy. Semin Immunol. 2017;31:64–75. doi: 10.1016/j.smim.2017.07.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Benson DM Jr., Bakan CE, Mishra A, Hofmeister CC, Efebera Y, Becknell B, Baiocchi RA, Zhang J, Yu J, Smith MK, et al. The PD-1/PD-L1 axis modulates the natural killer cell versus multiple myeloma effect: a therapeutic target for CT-011, a novel monoclonal anti–PD-1 antibody. Blood. 2010;116(13):2286–94. doi: 10.1182/blood-2010-02-271874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Tong L, Jimenez-Cortegana C, Tay AHM, Wickstrom S, Galluzzi L, Lundqvist A. NK cells and solid tumors: therapeutic potential and persisting obstacles. Mol Cancer. 2022;21(1):206. doi: 10.1186/s12943-022-01672-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Melaiu O, Lucarini V, Cifaldi L, Fruci D. Influence of the tumor microenvironment on NK cell function in solid tumors. Front Immunol. 2019;10:3038. doi: 10.3389/fimmu.2019.03038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol. 2009;9(3):162–74. doi: 10.1038/nri2506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Hanson EM, Clements VK, Sinha P, Ilkovitch D, Ostrand-Rosenberg S. Myeloid-derived suppressor cells down-regulate L-selectin expression on CD4+ and CD8+ T cells. J Immunol. 2009;183(2):937–44. doi: 10.4049/jimmunol.0804253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Chitadze G, Bhat J, Lettau M, Janssen O, Kabelitz D. Generation of soluble NKG2D ligands: proteolytic cleavage, exosome secretion and functional implications. Scand J Immunol. 2013;78(2):120–9. doi: 10.1111/sji.12072. [DOI] [PubMed] [Google Scholar]
  • 87.Pedroza-Pacheco I, Madrigal A, Saudemont A. Interaction between natural killer cells and regulatory T cells: perspectives for immunotherapy. Cell Mol Immunol. 2013;10(3):222–9. doi: 10.1038/cmi.2013.2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Guo ZS, Lotze MT, Zhu Z, Storkus WJ, Song XT. Bi- and tri-specific T cell engager-armed oncolytic viruses: next-generation cancer immunotherapy. Biomedicines. 2020;8(7):8. doi: 10.3390/biomedicines8070204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Fajardo CA, Guedan S, Rojas LA, Moreno R, Arias-Badia M, de Sostoa J, June CH, Alemany R. Oncolytic adenoviral delivery of an EGFR-Targeting T-cell engager improves antitumor efficacy. Cancer Res. 2017;77(8):2052–63. doi: 10.1158/0008-5472.CAN-16-1708. [DOI] [PubMed] [Google Scholar]
  • 90.Freedman JD, Hagel J, Scott EM, Psallidas I, Gupta A, Spiers L, Miller P, Kanellakis N, Ashfield R, Fisher KD, et al. Oncolytic adenovirus expressing bispecific antibody targets T-cell cytotoxicity in cancer biopsies. EMBO Mol Med. 2017;9(8):1067–87. doi: 10.15252/emmm.201707567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Christodoulou I, Ho WJ, Marple A, Ravich JW, Tam A, Rahnama R, Fearnow A, Rietberg C, Yanik S, Solomou EE, et al. Engineering CAR-NK cells to secrete IL-15 sustains their anti-AML functionality but is associated with systemic toxicities. J Immunother Cancer. 2021;9(12):e003894. doi: 10.1136/jitc-2021-003894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Chauchet X, Cons L, Chatel L, Daubeuf B, Didelot G, Moine V, Chollet D, Malinge P, Pontini G, Masternak K, et al. CD47xCD19 bispecific antibody triggers recruitment and activation of innate immune effector cells in a B-cell lymphoma xenograft model. Exp Hematol Oncol. 2022;11(1):26. doi: 10.1186/s40164-022-00279-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Nath PR, Pal-Nath D, Mandal A, Cam MC, Schwartz AL, Roberts DD. Natural killer cell recruitment and activation are regulated by CD47 expression in the tumor microenvironment. Cancer Immunol Res. 2019;7(9):1547–61. doi: 10.1158/2326-6066.CIR-18-0367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Demaria O, Gauthier L, Vetizou M, Blanchard Alvarez A, Vagne C, Habif G, Batista L, Baron W, Belaïd N, Girard-Madoux M, et al. Antitumor immunity induced by antibody-based natural killer cell engager therapeutics armed with not-alpha IL-2 variant. Cell Rep Med. 2022;3(10):100783. doi: 10.1016/j.xcrm.2022.100783. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Human Vaccines & Immunotherapeutics are provided here courtesy of Taylor & Francis

RESOURCES