1 Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata", 00133 Rome, Italy
2 Department of Experimental Medicine, University of Rome “Sapienza", 00161 Rome, Italy
The treatment of adult and pediatric solid tumors remains a major challenge for oncologists. Considerable attention has focused on the adoptive transfer of ex vivo expanded and activated natural killer (NK) cells, defined as an excellent “off-the-shelf” product for novel cell-based anti-cancer therapeutic strategies [1].
Notably, NK cells are cytotoxic lymphocytes that participate in innate immune responses and recognize virus-infected and transformed cells without prior specific sensitization, exceptional specificity, or acquisition of long-term memory.
NK cell-mediated recognition and lysis of cancer cells are strongly dependent on the tumor cell surface expression of ligands recognized by NK cell-activating receptors [1, 2]. The ligands for Natural-Killer receptor group 2, member D (NKG2D), are cellular stress-inducible major histocompatibility complex (MHC)-I-related proteins such as MICA, MICB, and six members of the UL16-binding protein (ULBP) family. The NKG2D receptor consists of a homodimer of two disulfide-associated transmembrane proteins that lack the ability to signal via intracellular domains. However, the adaptor protein DAP10 ensures signaling activation after NKG2D binding in humans [3]. The ligands for DNAX accessory molecule-1 (DNAM-1), such as the poliovirus receptor (PVR; nectin-like molecule, CD155) and Nectin-2 (CD112), are highly expressed in solid tumor cells and weakly expressed in normal tissue cells. Notably, inhibitory receptors such as T-cell immunoglobulin and ITIM domain (TIGIT), CD96 (TACTILE; T-cell activation, increased late expression) and PVRIG compete with DNAM-1 for binding to PVR and Nectin-2 ligands (PVR is recognized by TIGIT, and CD96 and Nectin-2 are recognized by TIGIT and PVRIG) [4]. Therefore, the antitumor efficacy of NK cells depends on their phenotype, which is governed by the expression of activating/inhibitory receptors, thus indicating the activated/exhausted status of the cells [4]. On the other hand, defective expression of NK cell-activating receptors has been reported in cancer patients [5], thus supporting the idea that rescue of NK cell-mediated signaling constitutes a rationale for the development of new NK cell-based immunotherapies.
Current good manufacturing practice (GMP) for the adoptive transfer of NK cells involve the expansion of NK cells with high activation and low exhaustion and with increased trafficking and killing performance [2]. The results of several clinical trials have demonstrated that NK cell-based immunotherapy in combination with cytokines, monoclonal antibodies (mAbs), and immune checkpoint inhibitors is an effective and safe anticancer treatment strategy (ClinicalTrials.gov and [2]). Moreover, we recently reported that a low dose of polyphenols improves NK cell functions in vitro [6]. In addition, the use of NK cells armed with chimeric antigen receptors (CARs), which recognize specific molecules expressed on the surface of cancer cells, has shown numerous advantages by ensuring the recognition and eradication of several types of cancer cells [7].
The biological rationale that led to the development of engineered NK cells was
based on a wide range of limitations previously reported from the clinical use of
CAR-T cells, that require stringent haploidentical mismatch conditions, which are
difficult to apply to a wide range of cancer patients [8]. In contrast, NK cells
can be used therapeutically in allogeneic settings with a major safety, since,
unlike T cells, they showed a low risk of proliferation and fewer side effects
[9]. The use of CAR-T cells showed limitations associated with antigen escape,
poor tumor infiltration and trafficking, and high toxicity leading to the risk of
graft-versus-host disease (GvHD) and cytokine release syndrome [8], pathological
conditions that have never been reported after adoptive transfer of allogeneic NK
cells (except for a few cases of cotransfusion with hematopoietic stem cells
(HSCs), which are subsequently responsible for the development of GvHD) [9].
Different sources of NK cells have been explored, from established cell lines to
allogeneic and alloreactive primary cells isolated from peripheral and umbilical
cord blood or from induced pluripotent stem cells (iPSC), thus circumventing
ethical issues that effectively limit the use of human embryonic stem cells
(hESC), whose use remains restricted in some countries. Moreover, improved
ex vivo amplification and transfection methods have made it possible to
obtain more stable and efficient CAR-NK cells [1]. However, limits of in
vivo cell persistence, transduction efficiency and infiltration
capacity into the tumor microenvironment (TME) have not yet been completely
overcome [2]. TME is composed of immunosuppressive cells that negatively regulate
NK cells activity; their neutralization by monoclonal antibodies, in association
with immune checkpoint inhibitors, should therefore support CAR-NK cell-mediated
antitumor efficacy [2]. CARs expressed by NK cells are engineered membrane fusion
proteins consisting of an extracellular domain (scFv; single-chain fragment
variable) that targets tumor-associated and spacer/transmembrane domains that are
linked to an intracellular region containing the primary activator (for example,
the CD3
Fig. 1.
Schematic model of chimeric antigen receptors (CARs) and chimeric-activating receptors-engineered Natural Killer (NK) cells and their clinical applications. Upper panel: Description of chimeric antigen receptors (CARs)-engineered NK cells and their clinical applications. Lower panel: Description of chimeric-activating receptors-engineered NK cells and their clinical applications. The ClinicalTrials.gov identification numbers for each clinical study are reported. scFv, single-chain fragment variable; NKG2D, Natural Killer receptor group 2, member D; DNAM-1, DNAX-accessory molecule-1; NCR, natural cytotoxic receptor.
NK cells have been engineered with many different CARs containing a scFv, and these cells have been evaluated in 65 clinical trials to date (ClinicalTrials.gov, Fig. 1). Anti-CD19 CAR-NK cells were the first to be developed with supporting data from 24 clinical trials [11]. Other CAR-NK cells targeting CD5, CD7, CD22, CD33, CD70, CD123, CD276, DLL3, 5T4, Claudin6, PD-L1, BCMA, PSMA, mesothelin, TROP2, ErbB2/HER2, ROBO1 and MUC1 have been evaluated in 41 clinical trials to date. In contrast, NK cells engineered with chimeric NKG2D-activating receptors have been evaluated in only 8 clinical trials [1] (ClinicalTrials.gov, Fig. 1). In addition, the need to overcome the toxicity that often results from targeting molecules expressed not only by tumor cells but also by normal cells, necessitates the search for less toxic, more efficient and tumor-specific chimeric molecules, such as those represented by chimeric-activating receptors, for use in arming NK cells.
The use of NK cells engineered with the chimeric activating receptor NKG2D has
been extended to several types of cancers (Fig. 1). NK cells expressing the
chimeric activating receptor NKG2D were engineered to express full-length NKG2D
in frame with the CD3
Therefore, NK cells engineered with chimeric NKG2D have rapidly gained prominence as one of the most promising types of nontoxic chimeric molecule-engineered NK cells with activity against tumors expressing ligands recognized by NKG2D. The successful use of NK cells engineered with chimeric NKG2D is also due to their potential to restore the often compromised immune response [2]. Indeed, chimeric NKG2D receptor-engineered NK cells have been reported to bind ligands expressed by tumor-infiltrating myeloid-derived suppressor cells (MDSCs) in the TME, thus neutralizing the immunosuppressive functions of these cells [12].
The use of NKG2D chimeric activating receptor-engineered NK cells, based on the
high expression of ligands recognized by NKG2D on both hematological and solid
tumor cells, has also broadened their application prospects for the treatment of
solid tumors, as compared with hematological malignancies (27 for solid tumors
compared with 38 for Acute Myeloid Leukemia (AML), Multiple Myeloma (MM) and B-lymphoma; Fig. 1). On the basis of this need
and considering that many solid tumor cells exhibit high expression of ligands
recognized mainly by DNAM-1, such as PVR and Nectin-2, we recently demonstrated
the effective cytotoxicity of DNAM-1 chimeric activating receptor-engineered NK
cells in neuroblastoma cell lines [10, 13, 14]. We demonstrated the cytotoxic
potential of primary human NK cells engineered with chimeric DNAM-1-CD3
Very recently, NK cells engineered with NCRs such as NKp30, NKp46 and especially NKp44 depending on DAP12, conjugated to an extracellular anti-HER2 scFv, were reported to augment NK cell activities, such as tumor lysis and cytokine production, against ovarian cancer cells [15].
Overall, NK cells engineered with chimeric activating receptors have shown promising results, as reported in preclinical studies and clinical trials. However, further efforts are needed to generate engineered NK cells with greater antitumor efficacy and less toxicity. In addition, improved methods should be employed to make engineered NK cells more stable, easier to apply clinically, and easier to cryopreserve for immediate use against a broad spectrum of tumors, mainly solid tumors.
LC, ML, RB wrote the editorial. All authors read and approved the final manuscript. All authors have participated sufficiently in the work and agreed to be accountable for all aspects of the work.
Not applicable.
We gratefully acknowledge the assistance in Fig. 1 preparation from Dr. Raffaele Carrano recipient of the Tor Vergata PhD program in Tissue Engineering and Remodeling Biotechnologies for Body Functions.
This research was funded by Ministero dell’Università e della Ricerca, PRIN 2022 grants CUP: E53D23001190006 to Loredana Cifaldi.
The authors declare no conflict of interest. Given Roberto Bei’s role as the Editorial Board Member, he had no involvement in the peer-review of this article and has no access to information regarding its peer review. Full responsibility for the editorial process for this article was delegated to Graham Pawelec.
References
Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

