Next Article in Journal
Patient-Reported Mobility, Physical Activity, and Bicycle Use after Vulvar Carcinoma Surgery
Previous Article in Journal
The Role of Non-Peripancreatic Lymph Nodes in the Survival of Patients Suffering from Pancreatic Cancer of the Body and Tail: A Systematic Review and Meta-Analysis of High-Quality Studies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Function of NK Cells in Tumor Metastasis and NK Cell-Based Immunotherapy

Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
Cancers 2023, 15(8), 2323; https://doi.org/10.3390/cancers15082323
Submission received: 15 March 2023 / Revised: 9 April 2023 / Accepted: 13 April 2023 / Published: 16 April 2023

Abstract

:

Simple Summary

Innate immune natural killer (NK) cells are capable of killing metastatic cancer cells without activation by antigen-presenting cells beforehand. The cytotoxic effects and immune regulation of NK cells are precisely controlled by an energetic balance of signals produced by a group of activating and inhibitory receptors expressed on NK cells, while metastatic tumor cells with multiple strategies escape immune cells attack. This review focuses on the critical function of NK cells in metastasis and recently developed objectively effective NK cell-based immunotherapies.

Abstract

Metastatic tumors cause the most deaths in cancer patients. Treating metastasis remains the primary goal of current cancer research. Although the immune system prevents and kills the tumor cells, the function of the immune system in metastatic cancer has been unappreciated for decades because tumors are able to develop complex signaling pathways to suppress immune responses, leading them to escape detection and elimination. Studies showed NK cell-based therapies have many advantages and promise for fighting metastatic cancers. We here review the function of the immune system in tumor progression, specifically focusing on the ability of NK cells in antimetastasis, how metastatic tumors escape the NK cell attack, as well as the recent development of effective antimetastatic immunotherapies.

1. Introduction of the General Immune System

The immune system, composed of a complex of organs, immune cells, and molecules in the body, not only defends against foreign pathogens but also eliminates potentially deleterious mutation-bearing cells to protect and maintain normal body functions (Figure 1). Immune cells originate from stem cells in immune organs and become a variety of cell types, including neutrophils, eosinophils, basophils, mast cells, monocytes, macrophages, dendritic cells, natural killer cells (NK cells), B lymphocytes (B cells), and T lymphocytes (T cells). Physiologically, immune cells maintain a homeostatic balance between activating and inhibitory immunity for an adequate immune reaction against pathogens and diseases, including cancer [1]. The immune reaction contains two primary types: innate and adaptive immunity. Innate immunity is predominantly nonspecific, and defense reactions are rapidly generated within foreign antigen encounters. The NK and dendritic cells, as well as macrophage, neutrophils, basophils, and eosinophils, are the primary cell types in innate immunity [2]. Adaptive immunity is based on the clonal selection of T cells and B cells with receptors recognizing particular ‘non-self’ antigens represented by major histocompatibility complex (MHC) molecules on antigen-presenting cells. It is the second line of protection and needs a few days to become activated. After activation and expansion, T cells become cytotoxicity T cells (CD8+ T cell) to recognize and kill the target cells expressing the same antigen through phagocytosis and antigen-specific cytotoxicity [3], while B cells produce the antibody for antigen and are primarily demanded in antibody-mediated immunity [4] (Figure 1B).
Cancer originates from cells harboring genetic mutations [5], which can express the mutant proteins as neoantigens. Neoantigen peptides are presented to immune cells through the MHC class I (MHC-I) molecules on the surface of the cancer cell, distinguishing them from their normal counterparts (nonself) [6]. This presentation leads to the activation of T cells to become specific cytotoxicity CD8+ T cells, which can kill those tumor cells. NK cells can detect and rapidly destroy tumor cells with downregulated MHC-I expression, terming ‘missing self’ recognition [7,8]. Hereafter, NK cells quickly produce cytokines and secret the interferon-γ (IFNγ), tumor necrosis factor-alpha (TNFα), granulocyte-macrophage colony-stimulating factor (GM-CSF), and chemokines for activating other immune cells such as T and B cells to boost adaptive immunity [9]. Moreover, NK cells can be provoked by antibody opsonization for mediating antibody-dependent cellular cytotoxicity (ADCC) [10]. To trigger and develop the entire immune response requires the interaction of tumor cells and immune cells, which needs three signals: (1) first signal—an antigen expressed in cancer cells; (2) second signal—stimulatory molecules in both cancer and immune cells, and (3) third signal—cytokine signaling in immune cells [11,12]. However, cancers can develop multiple strategies to escape immune cell attack [13]. Immune escape is usually associated with loss of stimulating molecules that includes the downregulation of classical HLA molecules (missing self-hypothesis), loss of stimulatory cytokines, and/or gain of suppressing molecules such as expression of nonclassical HLA-G, functional Th2-type activity shift (e.g., decrease in IFNγ and/or increase in TGFb, IL6, and IL10) and elevation of the Fas ligand on cancer cells [14,15,16]. The interactivity of the immune cells and cancer cells can determine the fate of tumors. This can be described as three “E’s”, which are eradication, equilibrium, and escape (Figure 2) [17,18]. This process, called cancer immunoediting, has built a theoretical hypothesis for comprehending our immune protection and sculpting cancer immunogenicity on tumor progression. In early cancer development, tumor cells can be eliminated by the immune system via the responses of innate and adaptive immunity in the eradication phase. However, tumor cells can manage the immune pressure to survive and then may enter the equilibrium phase. Escape is the phase of the tumor progression, where tumors are sculpted immunologically with the ability to inhibit the immune cells attach and establish an immunosuppressive tumor microenvironment (TME), then become clinically relevant as a mass and/or metastasis [13,17,18,19,20] (Figure 2). Boosting immunity and inhibiting immunosuppressive TME are principal strategies for anticancer immune therapy. One of the breakthroughs in antitumor immunotherapy focuses on utilizing adaptive immunity, achieving primary success through inhibiting immune checkpoints [21] or using CAR-T cells to enhance antitumor CD8+ T cell responses [22,23]. Emerging evidence indicates that tumors can develop various strategies to evade CD8+ T cell recognition, and side effects remain in T cell therapies [24]. Although T cells can eliminate most cells within a primary tumor, a fraction can still escape antitumor immunity and survive, ultimately leading to metastatic disease and death of the patient. However, NK cells can preferentially attack these tumors [25]. NK cells have built-in safety characteristics to avoid autoimmunity and have the ability to maintain immune homeostasis to prevent autoimmune disease [26]. Moreover, a subgroup of NK cells can present memory-like recall responses, playing the role of adaptive immunity [27,28]. Thus, these unique features of NK cells make them attractive targets for immunotherapy that can provoke their potent antitumor mechanisms (Table 1). Consequently, a new field of NK cells in metastasis has highlighted the important role of NK cells in immunosurveillance against metastasis [29]. This review focuses on the critical functions of NK cells in metastasis and the recent development of novel and objectively effective cancer immunotherapies.

2. NK Cell Biology and Function

NK cells represent 5–15% of the lymphocytes in human peripheral blood, with the unique ability to rapidly eliminate infected, mutated cells without preactivation [30,31,32]. NK cells originate from CD34+ hematopoietic stem cells in the BM, migrate to blood and other organs, and infiltrate cancer tissue [31,33]. Based on the surface marker profiles, NK cells exhibit different populations with functional heterogeneity at different maturation stages and anatomical locations [34]. Distinguished from myeloid cells, NK cells are defined as CD3CD56+ in peripheral blood [35]. Two major subtypes of NK cells are differentiated by levels of marker CD56 (neural cell adhesion molecule, NCAM) [36]. A total of 5–10% of blood NK cells are immature and express high levels of CD56 named CD56 bright NK cells, while most (90–95%) have downregulation of CD56 and convert into the matured CD56dim subgroup and also begin to express CD16 (FcrRIIIa) [37,38]. NK cells play roles in cytotoxic effects and immune regulation, two significant functions in innate immunity [39]. Without prestimulation, NK cells are able to detect and kill transformed cells by secreting perforin and granzymes [40]. Activated NK cells can also deliver the death ligands of TNFa, FasL, and apoptosis-inducing ligand (TRAIL), starting the apoptotic pathway [41]. In immune regulation, NK cells release various cytokines and chemokines such as IFNγ, IL10, CCL3, CCL4, and CCL5, connecting innate immunity to adaptive immunity [42]. NK cells express diverse activating and inhibitory receptors that transfer positive and negative signals for precisely regulating NK cell activity (Figure 3). The quantity of activating and inhibitory receptor signals contributes to the fate of NK cells’ target cells [43]. NKG2D and DNAM-1 are important receptors for activating NK cells. NKG2D can bind to the ligands of NKG2D (NKG2D), such as MICA/B [44] and UL16-binding proteins (ULBPs) [45,46], extending the signals via the adaptor protein DAP10, thereby activating the PI3K singling pathway to trigger cytotoxicity [47]. DNAM-1 can interact with its ligands, such as CD112 and CD155, in cancer cells [48] (Figure 3). CD16 is another highly efficient activating receptor that matured NK cells express [10]. After binding with the Fc region of antibodies, CD16 induces activation signals and triggers ADCC for lysing antibody-coated cancer cells [10]. Studies showed that several therapeutic antibodies, such as the anti-HER2 antibody trastuzumab not only recognized tumor-specific antigens but also increased NK cell infiltration in tumors to trigger ADCC with a role for NK cells in adaptive immunity [49,50]. Immunoglobulin family receptors, NKp46, NKp30, and NKp44 in NK cells, also play a natural cytotoxicity function when activated by metastatic tumor cells [51,52,53,54,55,56] (Figure 3). The killer immunoglobulin-like receptors (KIRs) [57] are the main NK cell inhibitory receptors that specifically bind to MHC-I molecules. Usually, MHC-I molecules are expressed in the normal cell but are lost in cancer and metastatic cells. By detecting MHC-I molecules, NK cells are inhibited by self-molecules, so they avoid killing normal cells and check “missing self” cancer and metastatic cells using their inhibitory receptors. NKG2A is another inhibitory receptor in NK cells. Upon interacting with nonclassical human leukocyte antigen class I molecule E (HLA-E), NKG2A triggers an inhibitory signal through the immunoreceptor tyrosine-based inhibition motif (ITIM) [58]. Furthermore, the inhibitory leukocyte immunoglobulin-like receptor-1 (LIR-1) could bind to HLA-G and HLA-I to inhibit NK cell activation [59]. Moreover, other inhibitory receptors, TIGIT, CD96, LAG3, PD-1, and Tim-3, express in NK cells for negatively regulating NK cell activation. CD96 and TIGIT can also bind to CD112 and CD155 competitively with DNAM-1 to play checkpoint action for maintaining a balance of activating and inhibitory signals. These receptors also increase as cancer progresses, causing NK cell exhaustion to act as an NK checkpoint, suggesting that these are potential targets in cancer immunotherapy [60] (Figure 3).

3. The Function of NK Cells in Tumor Metastasis

NK cells constitutively express lytic machinery that kills aberrant cells independently from preactivation with safety characteristics and rarely elicits autoimmunity (Table 1). Since their identification, NK cells with unique functional features have been suggested to play an important role in controlling tumor growth and metastasis [61,62,63].

3.1. NK Cell Function Related to Metastasis

NK cells were first found for their capability to eliminate tumor cells without prior stimulation by antigen-presenting cells [63,64]. Studies have reported that patients with a higher cancer incidence had inadequate peripheral NK cell cytotoxicity responses in various types of cancer [65,66,67,68,69]. Additionally, the patients who suffered NK cell dysfunction had an increased rate of malignancies and metastases [70,71,72,73,74,75,76,77]. Using an experimental metastasis assay, we and others found that antibodies mediated depletion of NK function or the use of an NK cell-deficient host increased metastasis, suggesting that NK cells play an essential role in antimetastasis ([78], our unpublished data). An intravital imaging system directly observed NK cells attack disseminated tumor cells leading to cancer cell death in mouse models [78]. These observations indicated that NK cells act as a killer in tumor progression, especially cancer cells spreading from their original site to the bloodstream [29,62]. Clinically, the patients who suffered various cancers with low amounts of peripheral or infiltrating NK cells at tumor sites have higher numbers of metastatic lesions. [74,79,80,81,82,83,84]. In contrast, the patients who suffered an increased metastatic cancer risk with high levels of NK cell activating receptors (NKAR) have good prognoses [77,85,86,87,88,89]. Moreover, a high level of IFNγ production by circulating NK cells and the presence of NKp30 are associated with a positive prediction for long-term survival in patients with breast cancer, gastrointestinal stromal tumor, or melanoma under treatments [69,79,80,90,91,92]. More recently, a study has shown that NK cells could sustain breast cancer dormancy for controlling breast cancer liver metastasis [93]. These facts suggest the notion that NK cells mediate antimetastatic effects in clinical. The notion has also been strongly linked to cancer treatment and preventing tumor metastasis. For example, after successfully removing the tumor by surgery, many patients still developed distant metastasis later; one of the important reasons is that low NK cell cytotoxicity and less IFNγ secretion link impaired NK cell function directly to increased postoperative metastases [94,95,96]. Thus, the functional restoration of NK cells by agents such as arginine prevents metastases [97,98].

3.2. How NK Cells Kill Metastases

It is well appreciated that the amount of positive and negative signals transmitted by NK cell activating or inhibitory receptors determines the fate of the targets [99]. Normal cells typically express low amounts of activating ligands and have higher amounts of MHC-I molecules that interact with inhibitory receptors and transduce more negative signals; therefore, NK cells will not eliminate normal somatic cells. In contrast, in malignant tumor cells, the expression of activating ligands has been increased, and the positive signals will overcome the negative signals, thus ensuring that aberrant cells are destroyed by NK cells [100,101]. The deficiency of NK cell activating receptor NCR1 in a GEM (genetically engineered mouse) model was shown to promote tumor growth [101,102]. Interestingly, when comparing with B16 melanoma growing subcutaneously in either NCR1 heterozygous (het) or homozygous (KO) mice, the tumor size in the two groups had no significant difference. Still, there was a substantial difference in the rate of metastasis between the two groups: NCR1 KO mice appeared to have significantly higher metastatic lesions than NCR1 het mice [102,103]. This indicates that the absence of an activating receptor dramatically affects metastasis development but has no evident effect on the primary tumor, implying that the level of NK cell activating signaling plays a vital role in controlling tumor metastasis. Subsequently, it was found that elevated secretion of IFNγ by NK cells enhanced the level of FN1 in the tumor, resulting in architectural alteration and less aggressive metastasis. In contrast, deficient NCR1 abolished the release of IFNγ [103]. Recent studies have also reported that NK cells are necessary for the selective destruction of circulating single breast tumor cells [104,105]. Extensive studies also support that NK cells control metastasis by activating receptor signaling. For example, NK cells efficiently eliminate metastatic melanoma cells when overexpressing ligands for NKp44, NKp46, and DNAM-1 [74]. In contrast, GEM mice deficient in DNAM-1 [106,107], Tlr3 (regulating NK cell responses to cytokines) [108], Il2rg (ablating NKp46+ NK cells) [109], or T-bet (regulating the differentiation of NK cells) [110,111] are susceptible to metastatic colonization. Interestingly, the metastatic potential could be prevented by transplanting bulk NK cells [110,111,112,113,114,115,116]. Moreover, treating NK cells with activating cytokine IL-15 can restore protection from metastasis in Tbx21-deficient mice [111]. Deletion of a negative regulator Clbl also promotes NK cell-dependent antimetastatic effects in mice [115]. Furthermore, deficient endogenous IL-15 inhibitor of Cish in NK cells (Cish−/−NK cells) leads to NK cell hyperactivation; transplanting the Cish−/−NK cells into mice could robustly abrogate the metastatic phenotype of highly metastatic B16F10 melanoma cells [117,118]. Decrease in DNAM-1 limits NK cytotoxicity and blocks IFNγ production, while the overexpression of DNAM-1 ligand in tumor cells causes a decrease in NKG2D ligands, indicating that NK cell-induced killing is initiated by DNAM-1 or NKG2D signaling pathway [74,117,119]. Moreover, NK cell depletion with antibodies of NK1.1 or asiago-GM markedly enhances the metastasis [116,120,121,122]. Mice with NK cell activating factors, including IFNγ, perforin 1 (PRF1), or TRAIL deficiency by gene knockout or antibody-caused neutralization, were more susceptible to metastatic incidences following challenges with tumor cell inoculation or with carcinogen-induced tumors [123,124,125]. NK cells also produce and release IFNγ and TNFα to function on macrophages and dendritic cells for enhancing the immune response [126]. Interestingly, using an image tracker system, a recent study functionally visualized that NK cells directly contacted metastatic tumor cells rapidly, leading to its ERK activation and metastatic tumor cell apoptosis [78]. They then confirmed metastatic tumor cell death related to DNAM-1 activation in NK cells [78]. Whereas a more recent study has shown that activation of inhibitory receptor NKG2A/HLA-E signaling promoted the distant metastasis of PDAC; blocking this pathway provokes NK cells and inhibits PDAC metastasis [127]. Whether the NK cells destroy metastatic cancer depends on the amounts of signals from activating and inhibitory receptors in the NK cells. Activating receptors interact with molecules on the surface of cancer cells and ‘turn on’ activation of the NK cell. Inhibitory receptors on NK cells check the signals from its ligands on metastatic cancer cells to block the ability of NK cell-mediated killing. Metastatic tumor cells often lose NK cell inhibitory receptor ligands of MHC-I, which reduce the inhibitory signal and leave them vulnerable to NK cell killing. Activating signals through activating receptors in NK cells promote NK proliferation and stimulate the secretion of cytotoxic granules to release perforin and granzymes, leading to metastatic tumor lysis (Figure 1B and Figure 3).

4. Immunosuppression of NK in Tumor Metastasis

The effectiveness of NK cell killing of metastatic tumors depends not only on the immensity of the NK cell response, such as the activation of NK cells, but also on the capacity of cancer cells to evade destruction. Tumor cells can develop a wide range of strategies to elude detection and destruction by NK cells, continue to grow at distant sites, and then form metastases [61]. The process by which tumor cells develop the intrinsic properties to avoid recognition and elimination by NK cells is referred to as “escape” [61]. The efficiency of escape determines the success of disseminated tumor cells in giving rise to distant metastases. Most strategies tumor cells use to evade NK cell killing are described here. First, tumor cells can increase ligands of NK inhibitory receptors such as CD111 [128], PD-L1 [129], HLA-G [130], galectin-9 [131], HMGB-1 [132,133,134], and CEACAM-1 [135], therefore the activation of inhibitory receptor signal pathways [127]. The expression of nonclassical HLA-G is reported in various metastatic cancers, which ties up the inhibitory receptor of LIR-1 to transduce the inhibitory signals to NK cells and inhibit the NK cell proliferation and gene expression (Figure 3) [130]. In fact, cancer cells frequently lose their MHC-1 to escape T cell attack but are vulnerable to NK cell killing, and the metastatic tumor has too many tricks to manage the MHC-1 level for refraining both T cell and NK cell elimination [67], such as genetic and epigenetic modification, and transcriptional and translational regulation [136,137,138]. Second, metastatic cancer cells decrease the ligands of NK cells activating receptors (NKAR) [139,140]. For example, metastatic tumor cells often shed the NKG2D ligand of MICA and MICB proteins [141] through proteolytic proteins such as ADAM10, ADAM17, and MMP14 [142,143], thereby producing soluble variants of ligands, acting as molecular decoys for blocking NK cell activation [144,145]. Indeed, in many different cohorts of cancers, patients with advanced stage tumors often have a high level of soluble NKAR ligands [146,147]. Loss of PVR and nectin 2, a ligand for the NK cell activating receptor DNAM-1, abolished NK cell-mediated destruction of metastatic melanoma B16F10 [78]; the consequence of deletion of DNAM-1 ligand caused due to failure to activation of ERK signal pathway in NK cells [78]. The downregulation of a death receptor FAS in metastatic tumors is another way to turn off NK cells for killing the metastatic cells through the FAS/FASL pathway [148]. Tumor cells can also decrease immunostimulatory factors within the TME. Tumor cells can secrete IL10, CXCL8, and TGFB1, which directly reduce NK cell cytotoxic functions [149,150,151] and/or recruit other immune cells such as Treg cells [152], MDSCs [153], CD11b + Ly6G + neutrophils [122,154], and DC [155], thus inhibiting NK cell functions indirectly. Analysis of the secreted protein profiling of progressive cancers showed little amounts of NK cell-stimulating molecules such as IFN [156] and IL-15 [157]. Tumor cells can rewrite their metabolic programming and release the metabolites that affect the TME to interfere with the antimetastatic functions of NK cells. For example, overexpression of the ectonucleoside triphosphate diphosphohydrolase 1 (ENTPD1) in metastatic tumor cells hydrolyze extracellular ATP into AMP to convert it to adenosine in hypoxia condition caused by HIF-1 [158,159]. Adenosine unleashes powerful immunosuppressive effects on NK cells via adenosine A2a receptor (ADORA2A) signaling [159,160,161,162]. Hypoxia in TME favors metastatic tumor cells for avoiding NK cell elimination by releasing TGFB1 and miRNAs through exosome to target NKG2D [163] and trigger cell autophagy to block tumor cells to GZMB-mediated lysis [164]. In contrast, hyperoxia promotes the destruction of metastases through immune responses of CD8+ CTLs and NK cells [165]. Inhibition of ENTPD1 by small molecular inhibitor polyoxometalate-1 or Entpd1 deletion blocked the metastasis of melanoma and colon cancer cells [166]. In addition, lactate produced by tumors creates a favorable niche for metastasis and modulates the TME preventing NK cell activation [167,168]. Furthermore, tumor-derived stromal inflammation has a condition-dependent effect on controlling metastasis by NK cells [169,170]. Recent studies showed other proteins such as PAEP, pp12, and pp14 upregulated in metastatic tumor cells to hamper NK cell function [171,172]. Last, some reports have shown that metastases exhibited higher stemness features than primary tumors, suggesting that metastatic tumor cells may obtain immune escape abilities from stem cells or dormant cells [173,174] that could shield their proliferation and hide in distant sites for a long time.

5. NK Cell-Based Immunotherapy for Metastasis Therapy

Metastatic tumors are challenging to treat and are the principal driver of cancer-related death [175]. Currently, available immunotherapies show promise for treating metastatic disease in some cancer patients. NK cells were discovered over half a century and are well known for eliminating tumor cells without prior activation; thus, they are in the first line against malignant tumors and metastases. Multiple strategies have been aimed at activating NK function, showing substantial therapeutic effects on metastatic diseases.

5.1. Cytokines to Boost Activation of NK Cells

NK cells can be activated by various factors. The cytokines are the first to use the direct engagement of NKR to boost the immune response of NK cells for treating cancer disease. These include IL2, IL12, IL15, IL21, and type I IFNs but not for all [176,177,178,179,180,181,182]. Following cytokine treatment, NK cells convert (turn into) lymphokine-activated killer (LAK) cells, subsequently producing cytokines and upregulating the factors such as adhesion molecules, perforin, granzymes, FasL, and TRAIL [183,184,185,186,187], thereby enhancing their capability to detect and adhere to cancer cells, triggering various activity to eliminate cancer cells through perforin/granzyme-dependent necrosis [188,189] and/or Fasl/TRAIL-mediated apoptosis [184,185,186,187].
IL2. Immunotherapeutic strategies against cancer started in the early 1980s. At the time, clinicians used IL2 to activate NK cells by injecting NK cell-stimulating doses of IL2 or preactivated NK cells (LAK cells) for treating patients with primary or metastasized tumors [190]. Although treatment of NK-stimulating doses of IL2 or transplantation of LAK showed some positive effects in patients with advanced cancers [190], IL2 treatment was unfortunately associated with side effects of capillary leak syndrome [191]. Moreover, IL2 enhanced the sensitivity of NK cells to apoptosis when they came into contact with vascular endothelium [192], leading to a reduction in tumor NK cell infiltration.
IL15. IL15 could promote NK cell survival and protects NK from activation-induced cell death (AICD). Additionally, it stimulates NK cell expansion more efficiently than IL2 [192,193]. However, relevant high doses of IL15 are required to elicit significant antitumor effects [194].
IL12. Unlike IL2 and IL15, IL12 mainly stimulates NK cell-mediated IFNγ production [195,196] to inhibit tumor angiogenesis and stimulate TRAIL/FasL-induced cell apoptosis in various cancers [197,198]. However, IFN-γ stimulated type 1 immunity may counteract the functions of tumor immunosuppressive type 2 cytokines (TGFβ and IL10) [199,200].
IL21. IL21 is a promising cytokine with the capability to activate NK cell antitumor immunity [201,202]. It promotes the expression of genes associated with type 1 immune reaction and differentiation of the highly cytotoxic CD56dim/CD16+ NK cells for triggering ADCC against tumor cells [202,203]. However, its treatment appears to have no specificity, and its efficacy was not as expected. Moreover, its treatment does not create cross-resistance or overlapping toxicities as other agents [204]; it still uses in cancer treatment in combination with other cytokine-based immunotherapies [205,206].
Other cytokines. The early-acting cytokines such as Flt3L, SCF, and IL7 can also enhance NK cell numbers [207,208]. However, Flt3L did not stimulate a desirable effect for a long time; more studies will be needed to examine the efficacy [209].
TLR. The engagement of TLR3 and TLR9 could lead to NK cell activation [108,210]. Due to viral and bacterial products could stimulate immune activity through TLRS, the study has synthesized the molecules from viral and bacterial products to treat the cancer cells in combination with cytokines [211].
Although the promise of cytokines to boost NK cell activation, the side effects of the toxicity of systemic cytokine usages and induced NK cell apoptosis are two significant limitations of cytokine-related NK cell immunotherapies in cancers. To overcome the limitations, a new strategy of dose pulsing of cytokine combinations keeps synergistic antitumor function and manages their toxicities. For example, using low doses of IL2 to promote NK cell proliferation, following high-dose pulses of IL2 or other cytokines to stimulate NK cells [212,213,214]. Additionally, an alternative approach was reported to use fusion proteins to deliver cytokines to tumor cells by tumor-specific Abs in synergy with NK activating cytokines IL21 [214,215].
To efficiently kill metastases, NK cells require the capability to traffic to tumor sites. Chemokines could regulate NK cell migration and stimulate NK cells to infiltrate tumor sites. For example, NK cells express chemokine receptors to respond to CXCL12 and CXC3L1, leading to migration vigorously [19]. Chemokines also regulate the NK cell interaction with other immune cells, such as DC [216,217], which triggers NK cell-associated antitumor immunity [218]. After directly contacting NK cells, DC promotes NK cell survival, activation, and differentiation through the secretion of IL12, IL1, IL18, and IL15 cytokines [219].

5.2. Immunomodulatory Agents to Modulate the Activation of NK Cells

Recent studies have renewed interest in using molecules that can block inhibitory pathways of NK cells to counter metastatic disease [220]. For example, one study showed that deletion of the gene-encoded E3 ubiquitin ligase Cbl-b or inactivation of its E3 ligase, allows NK cells to destroy metastatic tumors spontaneously [115]. The TAM tyrosine kinase receptors (Tyro3, Axl, and Mer) were identified to be a ubiquitylation substrate of Cbl-b. Treating mice bearing metastatic melanoma and mammary cancer with a small molecule TAM kinase inhibitor remarkedly reduced metastases and enhanced NK cell-mediated antimetastatic activity [221,222]. Moreover, the anticoagulant warfarin was found to exert antimetastatic activity via Cbl-b/TAM receptors through NK cells, suggesting that the TAM/Cbl-b inhibitory pathway may be the druggable target for awakening the NK cells to kill cancer metastases. Others that can mediate antimetastatic potential through NK cells include CD39/Entpd1 inhibitor polyoxomethalate-1 [166], ADORA2A antagonist PFB-709 [223], immunomodulatory drugs lenalidomide [224] and pomalidomide [225], as well as IDO inhibitors indoximod [226] and epacadostat [227] that inhibit IDO mediated suppression of NK cell function.

5.3. Enhancing NK Cell-Mediated Antibody-Dependent Cellular Cytotoxicity

Most matured NK cells express potent activating receptor CD16 that crosslinks with the Fc region of antibodies to initiate ADCC, killing the antibody-coated metastatic cancer cells [228] (Figure 4). Many targeted anticancer antibodies used in the clinic can not only inhibit the targeted signaling pathways for metastatic tumor cell survival and proliferation but also enhance NK cell-induced ADCC [229] (Figure 4), such as FDA-approved cetuximab (antibody for the epidermal growth factor receptor) and trastuzumab (antibody of the erb-b2 receptor tyrosine kinase). The efficacy of these antibodies in treatment depends on NK cells to execute ADCC [230]. For example, studies showed that the therapeutic antibodies for tumor antigens could promote NK cell infiltration to cancer and trigger NK cell-mediated ADCC, suggesting the role of NK cells in adaptive immunity [231]. Another agent ALT-803, a superagonist IL-15 mutant and IL-15Rα-Fc fusion complex, is currently under FDA review, which appears to have the potential to treat patients with advanced solid tumors through activating NK cells and enhancing NK-mediated ADCC [232,233]. For promoting the efficacy of ADCC, substantial progress has been achieved in developing advanced-generation antibodies with ameliorated ADCC potential [234] based on isotype and glycosylation of antibody and NK cell activating receptor CD16 [229]. Furthermore, bispecific and trispecific multifunctional antibodies with Fc recognizing NK cell CD16a and one or more Fc regions for tumor antigens that preserve ADCC potential and physically tie NK cells to malignant cells through bispecific killer engagers (BiKEs) or trispecific killer engagers (TriKEs), therefore ensuing specificity and direct interaction between immune cells and tumors while low systemic toxicity [235,236] (Figure 4). NK cell-engaged BiKEs or TriKEs provide a solution to hurdle the limitations, which are lower cost, less toxicity, and less time-consuming preparation compared to T cell-engaged [63,237]. These agents can create an immunological synapse between NK and tumor cells by simultaneously engaging other NK cell activating receptors (not CD16) with one or two tumor antigens to maximize the lysis of the tumor by NK cells. Several BiKEs and TriKEs have been developed for various targets in preclinical or clinical trials (Table 2) [238,239]. For example, AFM-24, a BiTKe engaged IgG1-scFv fusion antibody of CD16 with epidermal growth factor receptor (EGFR) activating NK cells through CD16 receptor for targeting EGFR, is now in clinical Phase II for treating patients with lung, colorectal cancer, and metastasis [240]. Recently, Vallera and colleagues reported successfully generating a functional TriKE bispecific antibody that could bind CD16 on NK cells, recognize CD33 on myeloid cancer cells, and also contain a modified human IL15 to better induce significant NK cell cytotoxicity and cytokine secretion against CD33+ tumors [241] compared to BiKE only recognize CD16 and CD33 [242]. More recent studies have reported that replacement of “humanized” anti-CD16 single-domain camelid antibody from the anti-CD16 scFv enabled the use of wild-type IL15 instant of modified IL15 to build up cam16-wtIL15–33 TriKE, which exhibited more robust IL15 signaling and NK cell activation leading effective elimination of cancer by NK cells [243]. Thus, this TriKE is a unique NK engager with cytokine signaling, tumor-specific targeting, and a single intramolecular ADCC, which drives specific tumor-killing and induces NK cell proliferation and survival via IL15 fragments.

5.4. Blocking (Shielding) the NK Inhibitory Receptor Signals

NK cells undergo functional equilibrium through activating and inhibitory receptors. Metastatic cells often express high levels of ligands for inhibitory receptors to suppress the immune response. For example, NK cells serve as detectors and killers in tumor progression, typically eliminating cells lacking MHC class molecules. However, metastatic cells often manage to express a certain level of downregulated MHC-I and other critical molecules in the antigen presentation pathway, which may inhibit NK cell activation through its inhibitory receptor and limit T cell response to avoid both NK and T cell-mediated elimination [244]. Thus, blockade of the inhibitory signal is essential to evoke the capability of NK cells against metastatic tumors [245]. Specific antibodies against NK cell inhibitory receptors (NKIR) are common agents for blocking NK inhibition signals and inducing activation signals to unleash the capacity of NK cells in antimetastases [246,247]. Several antibodies specific for KIRs, NKG2A, or LIR-1 inhibitory receptors exert substantial therapeutic effects in preclinical models [248,249,250]. For example, in clinical trials, lirilumab targeted KIR2D1/2/3 has been evaluated in multiple patient cohorts with relapsed/refractory myeloma [251,252]. However, clinical trials did not result in the designed efficacy in monotherapy [246] due to the reason of that it might reduce the expression of KIR2D on NK cells and disrupt the NK cell’s “education” process, resulting in a decrease in NK cell quantity and quality [253]. Despite the unexpected in monotherapy, the KIRs antibodies combined with other agents are showing some promise [117,251]. For instance, the combination of lirilumab and lenalidomide was well tolerated and increased the median progression-free survival of multiple myeloma patients [251]. It was also reported that the anti-NKG2A antibody could restore NK cell activation and promote anticancer immunity in combination with PD-1/PD-L1 antibodies for lymphoma [254] as well as the combination with anti-EGFR antibody for treating squamous cell carcinoma [246]. A combination of such anticancer agents could restore NK cell activation by blocking the inhibition of NKG2A and triggering NK cell-mediated ADCC [125]. The TIGIT, CD96, TIM-3, and PD1 inhibitory receptors expressed in NK cells are potential immune checkpoint therapeutic targets [254,255]. Metastatic cancer cells highly express the ligands of these receptors, which leads to NK cell exhaustion and is often associated with unfavorable prognosis. Thus, inhibition of these receptor signaling averts the exhaustion of NK cells and enhances antimetastasis solid effectiveness. In fact, the studies have shown that the combination of anti-TIGIT antibody tiragolumab and anti-PD-L1 antibody atezolizumab exhibited a significant benefit for treating NSCLC patients with high levels of PD-L1 [256,257,258], leading to US FDA to approve the tiragolumab/atezolizumab as the first-line treatment for metastatic NSCLC patients with PD-L1 positive but no EGFR or ALK mutations recently. Other studies using a combination of tiragolumab and atezolizumab for treating solid tumors have also shown significant effects [259]. These indicate that inhibiting PD-1/PD-L1 signaling could significantly promote the capability of NK cells to antitumor progression, implicating that combined therapy with PD-1/PD-L1 on NK cells is a reasonable approach for NK cell-based therapy [260]. Interestingly, recent studies have demonstrated that alloreactive KIR-mismatched NK cells could induce antitumor effects [261,262]. Additionally, the knock of NKG2A inhibitory receptors in NK cells promoted NK cell-induced killing of metastasis [127,263].

5.5. Engineered NK Cells (CAR-NK) for Metastasis Therapy

Although many studies demonstrated that NK cells could kill tumor cells in animal models, the clinical benefits of NK cells in cancer therapy are limited. Moreover, conventional immunotherapies have several disadvantages, for example, cytokine-induced toxicity, tumor lysis syndrome, other side effects on normal tissues, and genotoxicity, which may cause death. A new generation of cancer immunotherapy has started to genetically engineer T cells and NK cells that express chimeric antigen receptors (CARs), which have been demonstrated to have promise for treating hematologic cancers [264]. CAR-T or -NK cells express engineered IgG to recognize tumor antigens allowing T cells and NK to destroy the tumor cells precisely and effectively [264,265]. Unlike CAR-T cells, CAR-NK cells are designed to avoid therapy-related toxicity and immune-induced side effects [266,267]. Furthermore, NK cells have significant advantages, including their contribution to the graft-vs-leukemia/graft-vs-tumor effect, and are not responsible for the graft-vs-host disease and multiple various sources [266,267,268,269,270] (Figure 5. Like the structure and design of the CAR-T, the CAR-NK is composed of a promoter, a short signal peptide (SP), a tumor antigen binding domain (scFv with a linker between VH and VL) (ectodomain), a hinge region, a transmembrane domain, and intracellular activation signal domain (endodomain) (Figure 6) [271]. The tumor antigen binding domain derives from single chain variable fragments (scFv) of an antibody that recognizes tumor-specific antigens. The hinge region links a tumor antigen scFV to a transmembrane domain for promoting the dimerization of the CAR, increasing cytokine production and cell proliferation of CAR cells, and enhancing persistence and antitumor effects in vivo. The transmembrane domain leads the CAR structure to anchor on the cell NK cell membrane. Once recognizing tumor cells by the tumor antigen binding domain, the CAR triggers the activation domain signal for killing targeted tumor cells. Numerous CAR-NKs have shown promise in mediating antimetastasis activity in preclinical and clinical trials (Figure 5 and Figure 6) [272,273].

5.6. Combination of Multiple Strategies for Metastasis Therapy

Despite the success of CAR-NK cells in some hematologic cancers, their application has been hindered by the high production cost, the time-consuming treatment procedure, and some unexpected side effects. In recent years, a variety of combined approaches have been tested, including NKIR and immune checkpoint blockers, CAR-NK producing Bi-specific scFv and immune checkpoint blocker, CAR-NK with NKIR blocker, CAR-NK with targeted inhibitor, or multifunctional BiKEs and TriKEs (Figure 7) [274,275,276]. The fusion molecules containing one or two NK activating receptors binding scFv or IgG (CD16-binding scFv and NKG2D binding IgG VHH or NKp46 binding Fc) and one or more tumor-specific antigens binding regions have been engineered as the trifunctional or tetrafunctional NK cell engager that targets tumor-specific antigens and activates NK cells for eliminating the tumors significantly with minimized side effects [38,277,278,279]. This approach combined the different strategies for activating NK cells to kill tumors specifically and efficiently, which is particularly innovative.

6. Perspectives

Tumor immunology has progressed impressively in developing innovative therapeutic approaches for fighting cancer and metastasis. The remarkable body of research aimed to boost the immune response by activating positive and/or inhibiting negative signals. Antibodies targeting the immune checkpoint have unlatched a new era for the clinical treatment of cancers. The significant research in the mechanism of antibodies has provided awareness to understand how the Fc region of an antibody affects the immune response. Accordingly, BiKEs or TriKEs have increased specificity and facilitated direct interaction between NK cells and tumors with less systemic toxicity. Recently, new techniques can be used to control when and where CAR-NK cells are active, targeting tumors at specific times and preventing CAR-NK cells from becoming less effective over time during cancer treatment. NK cells possess vigorous cytolytic activity in metastatic cancer and have become an attractive tool for antimetastatic immunotherapy. Therefore, more studies on the interactions between metastatic cancer cells and NK cells will be necessary to clarify the mechanism of NK cell cytolytic activity against metastasis and tumor resistance to NK cell-induced destruction. For example, trogocytosis has been observed between CAR cells and tumor cells, causing antigen reduction and tumor relapse [39,280]. A study of the mechanism of trogocytosis in CAR-NK cell therapy will be needed to develop a more effective strategy for CAR-NK application in antimetastatic diseases. Recently, a study using a dual CAR system with an NK self-recognizing inhibitory CAR and an activating CAR against a tumor antigen prevented the trogocytosis-caused tumor resistance to CAR-NK cells mediated elimination, indicating its practical application moving forward [39,281]. In summary, future studies should focus on better understanding the molecular mechanisms of how metastatic tumors escape immune detection and destruction and developing new agents with higher immune specificity, better tolerability, and fewer side effects for clinics to cure metastatic dead diseases successfully.

Funding

This work was funded in part by the Intramural Research Program at the National Institutes of Health, and a Staff Scientist/Staff Clinician Research Award to Y.Y. from the Center for Cancer Research, National Cancer Institute.

Acknowledgments

Thanks to Glenn Merlino for the critical reading of this manuscript.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Arase, H.; Mocarski, E.S.; Campbell, A.E.; Hill, A.B.; Lanier, L.L. Direct recognition of cytomegalovirus by activating and inhibitory NK cell receptors. Science 2002, 296, 1323–1326. [Google Scholar] [CrossRef]
  2. Bedoui, S.; Gebhardt, T.; Gasteiger, G.; Kastenmuller, W. Parallels and differences between innate and adaptive lymphocytes. Nat. Immunol. 2016, 17, 490–494. [Google Scholar] [CrossRef]
  3. Janeway, C.A., Jr.; Travers, P.; Walport, M.; Shlomchik, M.J. Principles of Innate and Adaptive Immunity. Immunobiology: The Immune System in Health and Disease, 5th ed.; Garland Science: New York, NY, USA, 2001. [Google Scholar]
  4. Cooper, M.D.; Alder, M.N. The evolution of adaptive immune systems. Cell 2006, 124, 815–822. [Google Scholar] [CrossRef] [PubMed]
  5. Tian, L.; Goldstein, A.; Wang, H.; Ching Lo, H.; Sun Kim, I.; Welte, T.; Sheng, K.; Dobrolecki, L.E.; Zhang, X.; Putluri, N.; et al. Mutual regulation of tumour vessel normalization and immunostimulatory reprogramming. Nature 2017, 544, 250–254. [Google Scholar] [CrossRef]
  6. Ljunggren, H.G.; Glas, R.; Sandberg, J.K.; Kärre, K. Reactivity and specificity of CD8+ T cells in mice with defects in the MHC class I antigen-presenting pathway. Immunol. Rev. 1996, 151, 123–148. [Google Scholar] [CrossRef] [PubMed]
  7. Ljunggren, H.G.; Kärre, K. In search of the ‘missing self’: MHC molecules and NK cell recognition. Immunol. Today 1990, 11, 237–244. [Google Scholar] [CrossRef] [PubMed]
  8. Malmberg, K.J.; Sohlberg, E.; Goodridge, J.P.; Ljunggren, H.G. Immune selection during tumor checkpoint inhibition therapy paves way for NK-cell “missing self” recognition. Immunogenetics 2017, 69, 547–556. [Google Scholar] [CrossRef] [PubMed]
  9. Sonnenberg, G.F.; Hepworth, M.R. Functional interactions between innate lymphoid cells and adaptive immunity. Nature Rev. Immunol. 2019, 19, 599–613. [Google Scholar] [CrossRef]
  10. Nimmerjahn, F.; Ravetch, J.V. Fcgamma receptors as regulators of immune responses. Nat. Rev. Immunol. 2008, 8, 34–47. [Google Scholar] [CrossRef]
  11. Driessens, G.; Kline, J.; Gajewski, T.F. Costimulatory and coinhibitory receptors in anti-tumor immunity. Immunol. Rev. 2009, 229, 126–144. [Google Scholar] [CrossRef]
  12. Cronin, S.J.; Penninger, J.M. From T-cell activation signals to signaling control of anti-cancer immunity. Immunol. Rev. 2007, 220, 151–168. [Google Scholar] [CrossRef] [PubMed]
  13. Dunn, G.P.; Old, L.J.; Schreiber, R.D. The immunobiology of cancer immunosurveillance and immunoediting. Immunity 2004, 21, 137–148. [Google Scholar] [CrossRef] [PubMed]
  14. Rouas-Freiss, N.; Khalil-Daher, I.; Riteau, B.; Menier, C.; Paul, P.; Dausset, J.; Carosella, E.D. The immunotolerance role of HLA-G. Semin. Cancer Biol. 1999, 9, 3–12. [Google Scholar] [CrossRef] [PubMed]
  15. Paul, P.; Rouas-Freiss, N.; Khalil-Daher, I.; Moreau, P.; Riteau, B.; Le Gal, F.A.; Avril, M.F.; Dausset, J.; Guillet, J.G.; Carosella, E.D. HLA-G expression in melanoma: A way for tumor cells to escape from immunosurveillance. Proc. Natl. Acad. Sci. USA 1998, 95, 4510–4515. [Google Scholar] [CrossRef] [PubMed]
  16. O’Garra, A.; Barrat, F.J.; Castro, A.G.; Vicari, A.; Hawrylowicz, C. Strategies for use of IL-10 or its antagonists in human disease. Immunol. Rev. 2008, 223, 114–131. [Google Scholar] [CrossRef]
  17. Dunn, G.P.; Bruce, A.T.; Ikeda, H.; Old, L.J.; Schreiber, R.D. Cancer immunoediting: From immunosurveillance to tumor escape. Nat. Immunol. 2002, 3, 991–998. [Google Scholar] [CrossRef]
  18. Dunn, G.P.; Old, L.J.; Schreiber, R.D. The three Es of cancer immunoediting. Annu. Rev. Immunol. 2004, 22, 329–360. [Google Scholar] [CrossRef]
  19. Garner, H.; de Visser, K.E. Immune crosstalk in cancer progression and metastatic spread: A complex conversation. Nat. Rev. Immunol. 2020, 20, 483–497. [Google Scholar] [CrossRef]
  20. Liu, Y.; Cao, X. Characteristics and Significance of the Pre-metastatic Niche. Cancer Cell 2016, 30, 668–681. [Google Scholar] [CrossRef]
  21. Basudan, A.M. The Role of Immune Checkpoint Inhibitors in Cancer Therapy. Clin. Pract. 2022, 13, 22–40. [Google Scholar] [CrossRef]
  22. Hsu, J.; Yang, Y.; Gergis, M.; Bi, X.; Yi, D.; Gergis, U. Chimeric Antigen Receptor T Cell Therapy for Solid Tumors. Hematol. Oncol. Stem Cell Ther. 2022, 15, 94–99. [Google Scholar] [CrossRef]
  23. Rotte, A.; Frigault, M.J.; Ansari, A.; Gliner, B.; Heery, C.; Shah, B. Dose-response correlation for CAR-T cells: A systematic review of clinical studies. J. Immunother. Cancer 2022, 10, e005678. [Google Scholar] [CrossRef]
  24. Beauvais, D.; Danhof, S.; Hayden, P.J.; Einsele, H.; Yakoub-Agha, I. Clinical data, limitations and perspectives on chimeric antigen receptor T-cell therapy in multiple myeloma. Curr. Opin. Oncol. 2020, 32, 418–426. [Google Scholar] [CrossRef] [PubMed]
  25. Bachanova, V.; Miller, J.S. NK cells in therapy of cancer. Crit. Rev. Oncog. 2014, 19, 133–141. [Google Scholar] [CrossRef] [PubMed]
  26. Bahmanyar, M.; Vakil, M.K.; Al-Awsi, G.R.L.; Kouhpayeh, S.A.; Mansoori, Y.; Mansoori, B.; Moravej, A.; Mazarzaei, A.; Ghasemian, A. Anticancer traits of chimeric antigen receptors (CARs)-Natural Killer (NK) cells as novel approaches for melanoma treatment. BMC Cancer 2022, 22, 1220. [Google Scholar] [CrossRef] [PubMed]
  27. Bald, T.; Krummel, M.F.; Smyth, M.J.; Barry, K.C. The NK cell–cancer cycle: Advances and new challenges in NK cell–based immunotherapies. Nat. Immunol. 2020, 21, 835–847. [Google Scholar] [CrossRef]
  28. Freud, A.G.; Mundy-Bosse, B.L.; Yu, J.; Caligiuri, M.A. The broad spectrum of human natural killer cell diversity. Immunity 2017, 47, 820–833. [Google Scholar] [CrossRef]
  29. Gonzalez, H.; Hagerling, C.; Werb, Z. Roles of the immune system in cancer: From tumor initiation to metastatic progression. Genes Dev. 2018, 32, 1267–1284. [Google Scholar] [CrossRef]
  30. Richards, J.O.; Chang, X.; Blaser, B.W.; Caligiuri, M.A.; Zheng, P.; Liu, Y. Tumor growth impedes natural-killer-cell maturation in the bone marrow. Blood 2006, 108, 246–252. [Google Scholar] [CrossRef] [PubMed]
  31. Sun, J.C.; Lanier, L.L. NK cell development, homeostasis and function: Parallels with CD8(+) T cells. Nat. Rev. Immunol. 2011, 11, 645–657. [Google Scholar] [CrossRef]
  32. Herberman, R.B.; Holden, H.T. Natural killer cells as antitumor effector cells. J. Natl. Cancer Inst. 1979, 62, 441–445. [Google Scholar] [CrossRef] [PubMed]
  33. Yu, J.; Freud, A.G.; Caligiuri, M.A. Location and cellular stages of natural killer cell development. Trends Immunol. 2013, 34, 573–582. [Google Scholar] [CrossRef] [PubMed]
  34. Stabile, H.; Fionda, C.; Gismondi, A.; Santoni, A. Role of distinct natural killer cell subsets in anticancer response. Front. Immunol. 2017, 8, 293. [Google Scholar] [CrossRef] [PubMed]
  35. Wu, C.; Li, B.; Lu, R.; Koelle, S.J.; Yang, Y.; Jares, A.; Krouse, A.E.; Metzger, M.; Liang, F.; Lore, K.; et al. Clonal tracking of rhesus macaque hematopoiesis highlights a distinct lineage origin for natural killer cells. Cell Stem Cell 2014, 14, 486–499. [Google Scholar] [CrossRef]
  36. Lotzová, E.; Savary, C.A. Human natural killer cell development from bone marrow progenitors: Analysis of phenotype, cytotoxicity and growth. Nat. Immun. 1993, 12, 209–217. [Google Scholar]
  37. Maskalenko, N.A.; Zhigarev, D.; Campbell, K.S. Harnessing natural killer cells for cancer immunotherapy: Dispatching the first responders. Nat. Rev. Drug Discov. 2022, 21, 559–577. [Google Scholar] [CrossRef]
  38. Laskowski, T.J.; Biederstädt, A.; Rezvani, K. Natural killer cells in antitumour adoptive cell immunotherapy. Nat. Rev. Cancer 2022, 22, 557–575. [Google Scholar] [CrossRef]
  39. Li, Y.; Basar, R.; Wang, G.; Liu, E.; Moyes, J.S.; Li, L.; Kerbauy, L.N.; Uprety, N.; Fathi, M.; Rezvan, A.; et al. KIR-based inhibitory CARs overcome CAR-NK cell trogocytosis-mediated fratricide and tumor escape. Nat. Med. 2022, 28, 2133–2144. [Google Scholar] [CrossRef]
  40. Voskoboinik, I.; Whisstock, J.C.; Trapani, J.A. Perforin and granzymes: Function, dysfunction and human pathology. Nat. Rev. Immunol. 2015, 15, 388–400. [Google Scholar] [CrossRef]
  41. Martinez-Lostao, L.; Anel, A.; Pardo, J. How do cytotoxic lymphocytes kill cancer cells? Clin. Cancer Res. 2015, 21, 5047–5056. [Google Scholar] [CrossRef]
  42. Vivier, E.; Tomasello, E.; Baratin, M.; Walzer, T.; Ugolini, S. Functions of natural killer cells. Nat. Immunol. 2008, 9, 503–510. [Google Scholar] [CrossRef]
  43. Martinet, L.; Smyth, M.J. Balancing natural killer cell activation through paired receptors. Nat. Rev. Immunol. 2015, 15, 243–254. [Google Scholar] [CrossRef]
  44. Bauer, S.; Groh, V.; Wu, J.; Steinle, A.; Phillips, J.H.; Lanier, L.L.; Spies, T. Activation of NK cells and T cells by NKG2D, a receptor for stress-inducible MICA. Science 1999, 285, 727–729. [Google Scholar] [CrossRef] [PubMed]
  45. Kriegeskorte, A.K.; Gebhardt, F.E.; Porcellini, S.; Schiemann, M.; Stemberger, C.; Franz, T.J.; Huster, K.M.; Carayannopoulos, L.N.; Yokoyama, W.M.; Colonna, M.; et al. NKG2D-independent suppression of T cell proliferation by H60 and MICA. Proc. Natl. Acad. Sci. USA 2005, 102, 11805–11810. [Google Scholar] [CrossRef]
  46. Lanier, L.L. NKG2D Receptor and Its Ligands in Host Defense. Cancer Immunol. Res. 2015, 3, 575–582. [Google Scholar] [CrossRef]
  47. Wu, J.; Song, Y.; Bakker, A.B.; Bauer, S.; Spies, T.; Lanier, L.L.; Phillips, J.H. An activating immunoreceptor complex formed by NKG2D and DAP10. Science 1999, 285, 730–732. [Google Scholar] [CrossRef]
  48. Bottino, C.; Castriconi, R.; Pende, D.; Rivera, P.; Nanni, M.; Carnemolla, B.; Cantoni, C.; Grassi, J.; Marcenaro, S.; Reymond, N.; et al. Identification of PVR (CD155) and Nectin-2 (CD112) as cell surface ligands for the human DNAM-1 (CD226) activating molecule. J. Exp. Med. 2003, 198, 557–567. [Google Scholar] [CrossRef]
  49. Arnould, L.; Gelly, M.; Penault-Llorca, F.; Benoit, L.; Bonnetain, F.; Migeon, C.; Cabaret, V.; Fermeaux, V.; Bertheau, P.; Garnier, J.; et al. Trastuzumab-based treatment of HER2-positive breast cancer: An antibody-dependent cellular cytotoxicity mechanism? Br. J. Cancer 2006, 94, 259–267. [Google Scholar] [CrossRef] [PubMed]
  50. Collins, D.M.; O’Donovan, N.; McGowan, P.M.; O’Sullivan, F.; Duffy, M.J.; Crown, J. Trastuzumab induces antibody-dependent cell-mediated cytotoxicity (ADCC) in HER-2-non-amplified breast cancer cell lines. Ann. Oncol. 2012, 23, 1788–1795. [Google Scholar] [CrossRef]
  51. Barrow, A.D.; Martin, C.J.; Colonna, M. The Natural Cytotoxicity Receptors in Health and Disease. Front. Immunol. 2019, 10, 909. [Google Scholar] [CrossRef] [PubMed]
  52. Pessino, A.; Sivori, S.; Bottino, C.; Malaspina, A.; Morelli, L.; Moretta, L.; Biassoni, R.; Moretta, A. Molecular cloning of NKp46, A novel member of the immunoglobulin superfamily involved in triggering of natural cytotoxicity. J. Exp. Med. 1998, 188, 953–960. [Google Scholar] [CrossRef] [PubMed]
  53. Pende, D.; Parolini, S.; Pessino, A.; Sivori, S.; Augugliaro, R.; Morelli, L.; Marcenaro, E.; Accame, L.; Malaspina, A.; Biassoni, R.; et al. Identification and molecular characterization of NKp30, a novel triggering receptor involved in natural cytotoxicity mediated by human natural killer cells. J. Exp. Med. 1999, 190, 1505–1516. [Google Scholar] [CrossRef]
  54. Cantoni, C.; Bottino, C.; Vitale, M.; Pessino, A.; Augugliaro, R.; Malaspina, A.; Parolini, S.; Moretta, L.; Moretta, A.; Biassoni, R. NKp44, a triggering receptor involved in tumor cell lysis by activated human natural killer cells, is a novel member of the immunoglobulin superfamily. J. Exp. Med. 1999, 189, 787–796. [Google Scholar] [CrossRef]
  55. Vitale, M.; Bottino, C.; Sivori, S.; Sanseverino, L.; Castriconi, R.; Marcenaro, E.; Augugliaro, R.; Moretta, L.; Moretta, A. NKp44, a novel triggering surface molecule specifically expressed by activated natural killer cells, is involved in non-major histocompatibility complex-restricted tumor cell lysis. J. Exp. Med. 1998, 187, 2065–2072. [Google Scholar] [CrossRef] [PubMed]
  56. Sen Santara, S.; Lee, D.J.; Crespo, Â.; Hu, J.J.; Walker, C.; Ma, X.; Zhang, Y.; Chowdhury, S.; Meza-Sosa, K.F.; Lewandrowski, M.; et al. The NK cell receptor NKp46 recognizes ecto-calreticulin on ER-stressed cells. Nature 2023. [Google Scholar] [CrossRef]
  57. Raulet, D.H.; Vance, R.E. Self-tolerance of natural killer cells. Nat. Rev. Immunol. 2006, 6, 520–531. [Google Scholar] [CrossRef]
  58. Wieten, L.; Mahaweni, N.M.; Voorter, C.E.; Bos, G.M.; Tilanus, M.G. Clinical and immunological significance of HLA-E in stem cell transplantation and cancer. Tissue Antigens 2014, 84, 523–535. [Google Scholar] [CrossRef] [PubMed]
  59. Navarro, F.; Llano, M.; Bellon, T.; Colonna, M.; Geraghty, D.E.; Lopez-Botet, M. The ILT2(LIR1) and CD94/NKG2A NK cell receptors respectively recognize HLA-G1 and HLA-E molecules co-expressed on target cells. Eur. J. Immunol. 1999, 29, 277–283. [Google Scholar] [CrossRef]
  60. Bi, J.; Tian, Z. NK cell dysfunction and checkpoint immunotherapy. Front. Immunol. 2019, 10, 1999. [Google Scholar] [CrossRef]
  61. Chan, I.S.; Knútsdóttir, H.; Ramakrishnan, G.; Padmanaban, V.; Warrier, M.; Ramirez, J.C.; Dunworth, M.; Zhang, H.; Jaffee, E.M.; Bader, J.S.; et al. Cancer cells educate natural killer cells to a metastasis-promoting cell state. J. Cell Biol. 2020, 219, e202001134. [Google Scholar] [CrossRef]
  62. López-Soto, A.; Gonzalez, S.; Smyth, M.J.; Galluzzi, L. Control of Metastasis by NK Cells. Cancer Cell 2017, 32, 135–154. [Google Scholar] [CrossRef] [PubMed]
  63. Chiossone, L.; Dumas, P.Y.; Vienne, M.; Vivier, E. Natural killer cells and other innate lymphoid cells in cancer. Nat. Rev. Immunol. 2018, 18, 671–688. [Google Scholar] [CrossRef]
  64. Barlozzari, T.; Reynolds, C.W.; Herberman, R.B. In vivo role of natural killer cells: Involvement of large granular lymphocytes in the clearance of tumor cells in anti-asialo GM1-treated rats. J. Immunol. 1983, 131, 1024–1027. [Google Scholar] [CrossRef]
  65. 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, 1795–1799. [Google Scholar] [CrossRef]
  66. Balsamo, M.; Vermi, W.; Parodi, M.; Pietra, G.; Manzini, C.; Queirolo, P.; Lonardi, S.; Augugliaro, R.; Moretta, A.; Facchetti, F.; et al. Melanoma cells become resistant to NK-cell-mediated killing when exposed to NK-cell numbers compatible with NK-cell infiltration in the tumor. Eur. J. Immunol. 2012, 42, 1833–1842. [Google Scholar] [CrossRef]
  67. Sandel, M.H.; Speetjens, F.M.; Menon, A.G.; Albertsson, P.A.; Basse, P.H.; Hokland, M.; Nagelkerke, J.F.; Tollenaar, R.A.; van de Velde, C.J.; Kuppen, P.J. Natural killer cells infiltrating colorectal cancer and MHC class I expression. Mol. Immunol. 2005, 42, 541–546. [Google Scholar] [CrossRef]
  68. Esendagli, G.; Bruderek, K.; Goldmann, T.; Busche, A.; Branscheid, D.; Vollmer, E.; Brandau, S. Malignant and non-malignant lung tissue areas are differentially populated by natural killer cells and regulatory T cells in non-small cell lung cancer. Lung Cancer 2008, 59, 32–40. [Google Scholar] [CrossRef]
  69. Mamessier, E.; Sylvain, A.; Thibult, M.L.; Houvenaeghel, G.; Jacquemier, J.; Castellano, R.; Gonçalves, A.; André, P.; Romagné, F.; Thibault, G.; et al. Human breast cancer cells enhance self tolerance by promoting evasion from NK cell antitumor immunity. J. Clin. Investig. 2011, 121, 3609–3622. [Google Scholar] [CrossRef] [PubMed]
  70. Orange, J.S. Natural killer cell deficiency. J. Allergy Clin. Immunol. 2013, 132, 515–525. [Google Scholar] [CrossRef]
  71. MacFarlane, A.W.; Jillab, M.; Smith, M.R.; Alpaugh, R.K.; Cole, M.E.; Litwin, S.; Millenson, M.M.; Al-Saleem, T.; Cohen, A.D.; Campbell, K.S. NK cell dysfunction in chronic lymphocytic leukemia is associated with loss of the mature cells expressing inhibitory killer cell Ig-like receptors. OncoImmunology 2017, 6, e1330235. [Google Scholar] [CrossRef] [PubMed]
  72. Pazina, T.; MacFarlane, A.W.; Bernabei, L.; Dulaimi, E.; Kotcher, R.; Yam, C.; Bezman, N.A.; Robbins, M.D.; Ross, E.A.; Campbell, K.S.; et al. Alterations of NK Cell Phenotype in the Disease Course of Multiple Myeloma. Cancers 2021, 13, 226. [Google Scholar] [CrossRef]
  73. Vetter, C.S.; Groh, V.; Thor Straten, P.; Spies, T.; Brocker, E.B.; Becker, J.C. Expression of stress-induced MHC class I related chain molecules on human melanoma. J. Investig. Dermatol. 2002, 118, 600–605. [Google Scholar] [CrossRef]
  74. Lakshmikanth, T.; Burke, S.; Ali, T.H.; Kimpfler, S.; Ursini, F.; Ruggeri, L.; Capanni, M.; Umansky, V.; Paschen, A.; Sucker, A.; et al. NCRs and DNAM 1 mediate NK cell recognition and lysis of human and mouse melanoma cell lines in vitro and in vivo. J. Clin. Investig. 2009, 119, 1251–1263. [Google Scholar] [CrossRef] [PubMed]
  75. Erdag, G.; Schaefer, J.T.; Smolkin, M.E.; Deacon, D.H.; Shea, S.M.; Dengel, L.T.; Patterson, J.W.; Slingluff, C.L., Jr. Immuno type and immunohistologic characteristics of tumor-infiltrating immune cells are associated with clinical outcome in metastatic melanoma. Cancer Res. 2012, 72, 1070–1080. [Google Scholar] [CrossRef] [PubMed]
  76. Halama, N.; Braun, M.; Kahlert, C.; Spille, A.; Quack, C.; Rahbari, N.; Koch, M.; Weitz, J.; Kloor, M.; Zoernig, I.; et al. Natural killer cells are scarce in colorectal carcinoma tissue despite high levels of chemokines and cytokines. Clin. Cancer Res. 2011, 17, 678–689. [Google Scholar] [CrossRef]
  77. Remark, R.; Alifano, M.; Cremer, I.; Lupo, A.; Dieu-Nosjean, M.C.; Riquet, M.; Crozet, L.; Ouakrim, H.; Goc, J.; Cazes, A.; et al. Characteristics and clinical impacts of the immune environments in colorectal and renal cell carcinoma lung metastases: Influence of tumor origin. Clin. Cancer Res. 2013, 19, 4079–4091. [Google Scholar] [CrossRef] [PubMed]
  78. Ichise, H.; Tsukamoto, S.; Hirashima, T.; Konishi, Y.; Oki, C.; Tsukiji, S.; Iwano, S.; Miyawaki, A.; Sumiyama, K.; Terai, K.; et al. Functional visualization of NK cell-mediated killing of metastatic single tumor cells. Elife 2022, 11, e76269. [Google Scholar] [CrossRef]
  79. Delahaye, N.F.; Rusakiewicz, S.; Martins, I.; Ménard, C.; Roux, S.; Lyonnet, L.; Paul, P.; Sarabi, M.; Chaput, N.; Semeraro, M.; et al. Circulating CD56bright NK cells inversely correlate with survival of melanoma patients. Sci. Rep. 2019, 9, 4487. [Google Scholar]
  80. Semeraro, M.; Rusakiewicz, S.; Minard-Colin, V.; Delahaye, N.F.; Enot, D.; Vély, F.; Marabelle, A.; Papoular, B.; Piperoglou, C.; Ponzoni, M.; et al. Clinical impact of the NKp30/B7-H6 axis in high-risk neuroblastoma patients. Sci. Transl. Med. 2015, 7, 283ra55. [Google Scholar] [CrossRef]
  81. Ishigami, S.; Natsugoe, S.; Tokuda, K.; Nakajo, A.; Che, X.; Iwashige, H.; Aridome, K.; Hokita, S.; Aikou, T. Prognostic value of intratumoral natural killer cells in gastric carcinoma. Cancer 2000, 88, 577–583. [Google Scholar] [CrossRef]
  82. Coca, S.; Perez-Piqueras, J.; Martinez, D.; Colmenarejo, A.; Saez, M.A.; Vallejo, C.; Martos, J.A.; Moreno, M. The prognostic significance of intratumoral natural killer cells in patients with colorectal carcinoma. Cancer 1997, 79, 2320–2328. [Google Scholar] [CrossRef]
  83. Donskov, F.; von der Maase, H. Impact of immune parameters on long-term survival in metastatic renal cell carcinoma. J. Clin. Oncol. 2006, 24, 1997–2005. [Google Scholar] [CrossRef]
  84. Gannon, P.O.; Poisson, A.O.; Delvoye, N.; Lapointe, R.; Mes-Masson, A.M.; Saad, F. Characterization of the intra-prostatic immune cell infiltration in androgen-deprived prostate cancer patients. J. Immunol. Methods 2009, 348, 9–17. [Google Scholar] [CrossRef] [PubMed]
  85. Rusakiewicz, S.; Semeraro, M.; Sarabi, M.; Desbois, M.; Locher, C.; Mendez, R.; Vimond, N.; Concha, A.; Garrido, F.; Isambert, N.; et al. Immune infil trates are prognostic factors in localized gastrointestinal stromal tumors. Cancer Res. 2013, 73, 3499–3510. [Google Scholar] [CrossRef]
  86. Pasero, C.; Gravis, G.; Granjeaud, S.; Guerin, M.; Thomassin-Piana, J.; Rocchi, P.; Salem, N.; Walz, J.; Moretta, A.; Olive, D. Highly effective NK cells are associated with good prognosis in patients with metastatic prostate cancer. Oncotarget 2015, 6, 14360–14373. [Google Scholar] [CrossRef] [PubMed]
  87. Platonova, S.; Cherfils-Vicini, J.; Damotte, D.; Crozet, L.; Vieillard, V.; Validire, P.; André, P.; Dieu-Nosjean, M.C.; Alifano, M.; Régnard, J.F.; et al. Profound coordinated alterations of intratumoral NK cell phenotype and function in lung carcinoma. Cancer Res. 2011, 71, 5412–5422. [Google Scholar] [CrossRef]
  88. Pasero, C.; Gravis, G.; Guerin, M.; Granjeaud, S.; Thomassin-Piana, J.; Rocchi, P.; Paciencia-Gros, M.; Poizat, F.; Bentobji, M.; Azario-Cheillan, F.; et al. Inherent and tumor-driven immune tolerance in the prostate microenvironment impairs natural killer cell antitumor activity. Cancer Res. 2016, 76, 2153–2165. [Google Scholar] [CrossRef]
  89. de Andrade, L.F.; Lu, Y.; Luoma, A.; Ito, Y.; Pan, D.; Pyrdol, J.W.; Yoon, C.H.; Yuan, G.C.; Wucherpfennig, K.W. Discovery of specialized NK cell populations infiltrating human melanoma metastases. JCI Insight 2019, 4, e133103. [Google Scholar] [CrossRef]
  90. Couanet, D.; Gutierrez, J.C.; Nunès, J.A.; Commo, F.; Bonvalot, S.; Ibrahim, N.; Terrier, P.; Opolon, P.; Bottino, C.; Moretta, A.; et al. Alternatively spliced NKp30 isoforms affect the prognosis of gastrointestinal stromal tumors. Nat. Med. 2011, 17, 700–707. [Google Scholar]
  91. Ménard, C.; Blay, J.Y.; Borg, C.; Michiels, S.; Ghiringhelli, F.; Robert, C.; Nonn, C.; Chaput, N.; Taïeb, J.; Delahaye, N.F.; et al. Natural killer cell IFN-gamma levels predict long-term survival with imatinib mesylate therapy in gastrointestinal stromal tumor-bearing patients. Cancer Res. 2009, 69, 3563–3569. [Google Scholar] [CrossRef] [PubMed]
  92. Messaoudene, M.; Fregni, G.; Enot, D.; Jacquelot, N.; Neves, E.; Germaud, N.; Garchon, H.J.; Boukouaci, W.; Tamouza, R.; Chanal, J.; et al. NKp30 isoforms and NKp46 transcripts in metastatic melanoma patients: Unique NKp30 pattern in rare melanoma patients with favorable evolution. OncoImmunology 2016, 5, e1154251. [Google Scholar] [CrossRef]
  93. Correia, A.L.; Guimaraes, J.C.; Auf der Maur, P.; De Silva, D.; Trefny, M.P.; Okamoto, R.; Bruno, S.; Schmidt, A.; Mertz, K.; Volkmann, K.; et al. Hepatic stellate cells suppress NK cell-sustained breast cancer dormancy. Nature 2021, 594, 566–571. [Google Scholar] [CrossRef]
  94. Angka, L.; Martel, A.B.; Kilgour, M.; Jeong, A.; Sadiq, M.; de Souza, C.T.; Baker, L.; Kennedy, M.A.; Kekre, N.; Auer, R.C. Natural killer cell IFNg secretion is profoundly suppressed following colorectal cancer surgery. Ann. Surg. Oncol. 2018, 25, 3747–3754. [Google Scholar] [CrossRef] [PubMed]
  95. Seth, R.; Tai, L.H.; Falls, T.; de Souza, C.T.; Bell, J.C.; Carrier, M.; Atkins, H.; Boushey, R.; Auer, R.A. Surgical stress promotes the development of cancer metastases by a coagulation-dependent mechanism involving natural killer cells in a murine model. Ann. Surg. 2013, 258, 158–168. [Google Scholar] [CrossRef]
  96. Angka, L.; Khan, S.; Kilgour, M.; Xu, R.; Kennedy, M.; Auer, R. Dysfunctional natural killer cells in the aftermath of cancer surgery. Int. J. Mol. Sci. 2017, 18, 1787. [Google Scholar] [CrossRef]
  97. Tai, L.H.; de Souza, C.T.; Bélanger, S.; Ly, L.; Alkayyal, A.A.; Zhang, J.; Rintoul, J.L.; Ananth, A.A.; Lam, T.; Breitbach, C.J.; et al. Preventing postoperative metastatic disease by inhibiting surgery-induced dysfunction in natural killer cells. Cancer Res. 2013, 73, 97–107. [Google Scholar] [CrossRef] [PubMed]
  98. Angka, L.; Tanese de Souza, C.; Baxter, K.E.; Khan, S.T.; Market, M.; Martel, A.B.; Tai, L.H.; Kennedy, M.A.; Bell, J.C.; Auer, R.C. Perioperative arginine prevents metastases by accelerating natural killer cell recovery after surgery. Mol. Ther. 2022, 30, 3270–3283. [Google Scholar] [CrossRef] [PubMed]
  99. Koch, J.; Steinle, A.; Watzl, C.; Mandelboim, O. Activating natural cytotoxicity receptors of natural killer cells in cancer and infection. Trends Immunol. 2013, 34, 182–191. [Google Scholar] [CrossRef] [PubMed]
  100. Ascierto, M.L.; Idowu, M.O.; Zhao, Y.; Khalak, H.; Payne, K.K.; Wang, X.Y.; Dumur, C.I.; Bedognetti, D.; Tomei, S.; Ascierto, P.A.; et al. Molecular signatures mostly associated with NK cells are predictive of relapse free survival in breast cancer patients. J. Transl. Med. 2013, 11, 145. [Google Scholar] [CrossRef]
  101. Halfteck, G.G.; Elboim, M.; Gur, C.; Achdout, H.; Ghadially, H.; Mandelboim, O. Enhanced in vivo growth of lymphoma tumors in the absence of the NK-activating receptor NKp46/NCR1. J. Immunol. 2009, 182, 2221–2230. [Google Scholar] [CrossRef] [PubMed]
  102. Glasner, A.; Ghadially, H.; Gur, C.; Stanietsky, N.; Tsukerman, P.; Enk, J.; Mandelboim, O. Recognition and prevention of tumor metastasis by the NK receptor NKp46/NCR1. J. Immunol. 2012, 188, 2509–2515. [Google Scholar] [CrossRef]
  103. Glasner, A.; Levi, A.; Enk, J.; Isaacson, B.; Viukov, S.; Orlanski, S.; Scope, A.; Neuman, T.; Enk, C.D.; Hanna, J.H.; et al. NKp46 receptor-mediated interferon-γ production by natural killer cells increases fibronectin 1 to alter tumor architecture and control metastasis. Immunity 2018, 48, 107–119. [Google Scholar] [CrossRef] [PubMed]
  104. Laughney, A.M.; Hu, J.; Campbell, N.R.; Bakhoum, S.F.; Setty, M.; Lavallée, V.P.; Xie, Y.; Masilionis, I.; Carr, A.J.; Kottapalli, S.; et al. Regenerative lineages and immune-mediated pruning in lung cancer metastasis. Nat. Med. 2020, 26, 259–269. [Google Scholar] [CrossRef] [PubMed]
  105. Lo, H.C.; Xu, Z.; Kim, I.S.; Pingel, B.; Aguirre, S.; Kodali, S.; Liu, J.; Zhang, W.; Muscarella, A.M.; Hein, S.M.; et al. Resistance to natural killer cell immunosurveillance confers a selective advantage to polyclonal metastasis. Nat. Cancer 2020, 1, 709–722. [Google Scholar] [CrossRef] [PubMed]
  106. Iguchi-Manaka, A.; Kai, H.; Yamashita, Y.; Shibata, K.; Tahara-Hanaoka, S.; Honda, S.; Yasui, T.; Kikutani, H.; Shibuya, K.; Shibuya, A. Accelerated tumor growth in mice deficient in DNAM-1 receptor. J. Exp. Med. 2008, 205, 2959–2964. [Google Scholar] [CrossRef]
  107. Chan, C.J.; Andrews, D.M.; McLaughlin, N.M.; Yagita, H.; Gilfillan, S.; Colonna, M.; Smyth, M.J. DNAM-1/CD155 interactions promote cytokine and NK cell-mediated suppression of poorly immunogenic melanoma metastases. J. Immunol. 2010, 184, 902–911. [Google Scholar] [CrossRef] [PubMed]
  108. Guillerey, C.; Chow, M.T.; Miles, K.; Olver, S.; Sceneay, J.; Takeda, K.; Möller, A.; Smyth, M.J. Toll-like receptor 3 regulates NK cell responses to cytokines and controls experimental metastasis. OncoImmunology 2015, 4, e1027468. [Google Scholar] [CrossRef]
  109. Merzoug, L.B.; Marie, S.; Satoh-Takayama, N.; Lesjean, S.; Albanesi, M.; Luche, H.; Fehling, H.J.; Di Santo, J.P.; Vosshenrich, C.A. Conditional ablation of NKp46+ cells using a novel Ncr1(greenCre) mouse strain: NK cells are essential for protection against pulmonary B16 metastases. Eur. J. Immunol. 2014, 44, 3380–3391. [Google Scholar] [CrossRef]
  110. Werneck, M.B.; Lugo-Villarino, G.; Hwang, E.S.; Cantor, H.; Glimcher, L.H. T-bet plays a key role in NK-mediated control of melanoma metastatic disease. J. Immunol. 2008, 180, 8004–8010. [Google Scholar] [CrossRef]
  111. Malaisé, M.; Rovira, J.; Renner, P.; Eggenhofer, E.; Sabet-Baktach, M.; Lantow, M.; Lang, S.A.; Koehl, G.E.; Farkas, S.A.; Loss, M.; et al. KLRG1+ NK cells protect T-bet-deficient mice from pulmonary metastatic colorectal carcinoma. J. Immunol. 2014, 192, 1954–1961. [Google Scholar] [CrossRef]
  112. Chan, C.J.; Martinet, L.; Gilfillan, S.; Souza-Fonseca-Guimaraes, F.; Chow, M.T.; Town, L.; Ritchie, D.S.; Colonna, M.; Andrews, D.M.; Smyth, M.J. The receptors CD96 and CD226 oppose each other in the regulation of natural killer cell functions. Nat. Immunol. 2014, 15, 431–438. [Google Scholar] [CrossRef]
  113. Martinet, L.; Ferrari De Andrade, L.; Guillerey, C.; Lee, J.S.; Liu, J.; Souza-Fonseca-Guimaraes, F.; Hutchinson, D.S.; Kolesnik, T.B.; Nicholson, S.E.; Smyth, M.J. DNAM-1 expression marks an alternative program of NK cell maturation. Cell Rep. 2015, 11, 85–97. [Google Scholar] [CrossRef]
  114. de Andrade, L.F.; Ngiow, S.F.; Martinet, L.; Smyth, M.J. Natural Killer cell control of BRAFV600E mutant melanoma during targeted therapy. Oncoimmunology 2015, 4, e998119. [Google Scholar] [CrossRef]
  115. Paolino, M.; Choidas, A.; Wallner, S.; Pranjic, B.; Uribesalgo, I.; Loeser, S.; Jamieson, A.M.; Langdon, W.Y.; Ikeda, F.; Fededa, J.P.; et al. The E3 ligase Cbl-b and TAM receptors regulate cancer metastasis via natural killer cells. Nature 2014, 507, 508–512. [Google Scholar] [CrossRef]
  116. Ferrari de Andrade, L.; Ngiow, S.F.; Stannard, K.; Rusakiewicz, S.; Kalimutho, M.; Khanna, K.K.; Tey, S.K.; Takeda, K.; Zitvogel, L.; Martinet, L.; et al. Natural killer cells are essential for the ability of BRAF inhibitors to control BRAFV600E-mutant metastatic melanoma. Cancer Res. 2014, 74, 7298–7308. [Google Scholar] [CrossRef] [PubMed]
  117. Putz, E.M.; Guillerey, C.; Kos, K.; Stannard, K.; Miles, K.; Delconte, R.B.; Takeda, K.; Nicholson, S.E.; Huntington, N.D.; Smyth, M.J. Targeting cytokine signaling checkpoint CIS activates NK cells to protect from tumor initiation and metastasis. OncoImmunology 2017, 6, e1267892. [Google Scholar] [CrossRef]
  118. Delconte, R.B.; Kolesnik, T.B.; Dagley, L.F.; Rautela, J.; Shi, W.; Putz, E.M.; Stannard, K.; Zhang, J.G.; Teh, C.; Firth, M.; et al. CIS is a potent checkpoint in NK cell-mediated tumor immunity. Nat. Immunol. 2016, 17, 816–824. [Google Scholar] [CrossRef] [PubMed]
  119. Morisaki, T.; Onishi, H.; Katano, M. Cancer immunotherapy using NKG2D and DNAM-1 systems. Anticancer Res. 2012, 32, 2241–2247. [Google Scholar] [PubMed]
  120. Spiegel, A.; Brooks, M.W.; Houshyar, S.; Reinhardt, F.; Ardolino, M.; Fessler, E.; Chen, M.B.; Krall, J.A.; DeCock, J.; Zervantonakis, I.K.; et al. Neutrophils suppress intraluminal NK cell-mediated tumor cell clearance and enhance extravasation of disseminated carcinoma cells. Cancer Discov. 2016, 6, 630–649. [Google Scholar] [CrossRef]
  121. Diefenbach, A.; Jensen, E.R.; Jamieson, A.M.; Raulet, D.H. Rae1 and H60 ligands of the NKG2D receptor stimulate tumour immunity. Nature 2001, 413, 165–171. [Google Scholar] [CrossRef]
  122. Malladi, S.; Macalinao, D.G.; Jin, X.; He, L.; Basnet, H.; Zou, Y.; de Stanchina, E.; Massague, J. Metastatic latency and immune evasion through autocrine inhibition of WNT. Cell 2016, 165, 45–60. [Google Scholar] [CrossRef]
  123. Street, S.E.; Cretney, E.; Smyth, M.J. Perforin and interferon-gamma activities independently control tumor initiation, growth, and metastasis. Blood 2001, 97, 192–197. [Google Scholar] [CrossRef]
  124. Smyth, M.J.; Thia, K.Y.; Cretney, E.; Kelly, J.M.; Snook, M.B.; Forbes, C.A.; Scalzo, A.A. Perforin is a major contributor to NK cell control of tumor metastasis. J. Immunol. 1999, 162, 6658–6662. [Google Scholar] [CrossRef]
  125. Takeda, K.; Hayakawa, Y.; Smyth, M.J.; Kayagaki, N.; Yamaguchi, N.; Kakuta, S.; Iwakura, Y.; Yagita, H.; Okumura, K. Involvement of tumor necrosis factor-related apoptosis-inducing ligand in surveillance of tumor metastasis by liver natural killer cells. Nat. Med. 2001, 7, 94–100. [Google Scholar] [CrossRef] [PubMed]
  126. Prager, I.; Watzl, C. Mechanisms of natural killer cell-mediated cellular cytotoxicity. J. Leukoc. Biol. 2019, 105, 1319–1329. [Google Scholar] [CrossRef] [PubMed]
  127. Liu, X.; Song, J.; Zhang, H.; Liu, X.; Zuo, F.; Zhao, Y.; Zhao, Y.; Yin, X.; Guo, X.; Wu, X.; et al. Immune ch.eckpoint HLA-E:CD94-NKG2A mediates evasion of circulating tumor cells from NK cell surveillance. Cancer Cell 2023, 41, 272–287.e9. [Google Scholar] [CrossRef] [PubMed]
  128. Wang, X.; Xing, Z.; Chen, H.; Yang, H.; Wang, Q.; Xing, T. High expression of nectin-1 indicates a poor prognosis and promotes metastasis in hepatocellular carcinoma. Front. Oncol. 2022, 12, 953529. [Google Scholar] [CrossRef]
  129. Benson, D.M., Jr.; Bakan, C.E.; Mishra, A.; Hofmeister, C.C.; Efebera, Y.; Becknell, B.; Baiocchi, R.A.; Zhang, J.; Yu, J.; Smith, M.K.; 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, 2286–2294. [Google Scholar] [CrossRef]
  130. Carosella, E.D.; Rouas-Freiss, N.; Tronik-Le Roux, D.; Moreau, P.; LeMaoult, J. HLA-G: An immune checkpoint molecule. Adv. Immunol. 2015, 127, 33–144. [Google Scholar]
  131. Girotti, M.R.; Salatino, M.; Dalotto-Moreno, T.; Rabinovich, G.A. Sweetening the hallmarks of cancer: Galectins as multifunctional mediators of tumor progression. J. Exp. Med. 2020, 217, e20182041. [Google Scholar] [CrossRef]
  132. Yan, W.; Chang, Y.; Liang, X.; Cardinal, J.S.; Huang, H.; Thorne, S.H.; Monga, S.P.; Geller, D.A.; Lotze, M.T.; Tsung, A. High-mobility group box 1 activates caspase-1 and promotes hepatocellular carcinoma invasiveness and metastases. Hepatology 2012, 55, 1863–1875. [Google Scholar] [CrossRef] [PubMed]
  133. Moriwaka, Y.; Luo, Y.; Ohmori, H.; Fujii, K.; Tatsumoto, N.; Sasahira, T.; Kuniyasu, H. HMGB1 attenuates antimetastatic defense of the lymph nodes in colorectal cancer. Pathobiology 2010, 77, 17–23. [Google Scholar] [CrossRef]
  134. Chiba, S.; Baghdadi, M.; Akiba, H.; Yoshiyama, H.; Kinoshita, I.; Dosaka-Akita, H.; Fujioka, Y.; Ohba, Y.; Gorman, J.V.; Colgan, J.D.; et al. Tumor-infiltrating DCs suppress nucleic acid-mediated innate immune responses through interactions between the receptor TIM-3 and the alarmin HMGB1. Nat. Immunol. 2012, 13, 832–842. [Google Scholar] [CrossRef]
  135. Thies, A.; Mauer, S.; Fodstad, O.; Schumacher, U. Clinically proven markers of metastasis predict metastatic spread of human melanoma cells engrafted in scid mice. Br. J. Cancer 2007, 96, 609–616. [Google Scholar] [CrossRef]
  136. Cornel, A.M.; Mimpen, I.L.; Nierkens, S. MHC Class I downregulation in cancer: Underlying mechanisms and potential targets for cancer immunotherapy. Cancers 2020, 12, 1760. [Google Scholar] [CrossRef] [PubMed]
  137. Garrido, F.; Aptsiauri, N. Cancer immune escape: MHC expression in primary tumours versus metastases. Immunology 2019, 158, 255–266. [Google Scholar] [CrossRef]
  138. Taylor, B.C.; Balko, J.M. Mechanisms of MHC-I downregulation and role in immunotherapy response. Front. Immunol. 2022, 13, 844866. [Google Scholar] [CrossRef]
  139. Armeanu, S.; Bitzer, M.; Lauer, U.M.; Venturelli, S.; Pathil, A.; Krusch, M.; Kaiser, S.; Jobst, J.; Smirnow, I.; Wagner, A.; et al. Natural killer cell-mediated lysis of hepatoma cells via specific induction of NKG2D ligands by the histone deacetylase inhibitor sodium valproate. Cancer Res. 2005, 65, 6321–6329. [Google Scholar] [CrossRef]
  140. Lopez-Soto, A.; Folgueras, A.R.; Seto, E.; Gonzalez, S. HDAC3 represses the expression of NKG2D ligands ULBPs in epithelial tumour cells: Potential implications for the immunosurveillance of cancer. Oncogene 2009, 28, 2370–2382. [Google Scholar] [CrossRef] [PubMed]
  141. Deng, W.; Gowen, B.G.; Zhang, L.; Wang, L.; Lau, S.; Iannello, A.; Xu, J.; Rovis, T.L.; Xiong, N.; Raulet, D.H. Antitumor immunity. A shed NKG2D ligand that promotes natural killer cell activation and tumor rejection. Science 2015, 348, 136–139. [Google Scholar] [CrossRef]
  142. Chitadze, G.; Lettau, M.; Bhat, J.; Wesch, D.; Steinle, A.; Furst, D.; Mytilineos, J.; Kalthoff, H.; Janssen, O.; Oberg, H.H.; et al. Shedding of endogenous MHC class I-related chain molecules A and B from different human tumor entities: Heterogeneous involvement of the “a disintegrin and metalloproteases” 10 and 17. Int. J. Cancer 2013, 133, 1557–1566. [Google Scholar] [CrossRef] [PubMed]
  143. Liu, G.; Atteridge, C.L.; Wang, X.; Lundgren, A.D.; Wu, J.D. The membrane type matrix metalloproteinase MMP14 mediates constitutive shedding of MHC class I chain-related molecule A independent of A disintegrin and metalloproteinases. J. Immunol. 2010, 184, 3346–3350. [Google Scholar] [CrossRef]
  144. Zhang, J.; Basher, F.; Wu, J.D. NKG2D ligands in tumor immunity: Two sides of a coin. Front. Immunol. 2015, 6, 97. [Google Scholar] [CrossRef]
  145. Schlecker, E.; Fiegler, N.; Arnold, A.; Altevogt, P.; Rose-John, S.; Moldenhauer, G.; Sucker, A.; Paschen, A.; von Strandmann, E.P.; Textor, S.; et al. Metalloprotease-mediated tumor cell shedding of B7-H6, the ligand of the natural killer cell-activating receptor NKp30. Cancer Res. 2014, 74, 3429–3440. [Google Scholar] [CrossRef]
  146. Yamaguchi, K.; Chikumi, H.; Shimizu, A.; Takata, M.; Kinoshita, N.; Hashimoto, K.; Nakamoto, M.; Matsunaga, S.; Kurai, J.; Miyake, N.; et al. Diagnostic and prognostic impact of serum-soluble UL16-binding protein 2 in lung cancer patients. Cancer Sci. 2012, 103, 1405–1413. [Google Scholar] [CrossRef]
  147. Paschen, A.; Sucker, A.; Hill, B.; Moll, I.; Zapatka, M.; Nguyen, X.D.; Sim, G.C.; Gutmann, I.; Hassel, J.; Becker, J.C.; et al. Differential clinical significance of individual NKG2D ligands in melanoma: Soluble ULBP2 as an indicator of poor prognosis superior to S100B. Clin. Cancer Res. 2009, 15, 5208–5215. [Google Scholar] [CrossRef]
  148. Maecker, H.L.; Yun, Z.; Maecker, H.T.; Giaccia, A.J. Epigenetic changes in tumor Fas levels determine immune escape and response to therapy. Cancer Cell 2002, 2, 139–148. [Google Scholar] [CrossRef] [PubMed]
  149. Bruno, A.; Focaccetti, C.; Pagani, A.; Imperatori, A.S.; Spagnoletti, M.; Rotolo, N.; Cantelmo, A.R.; Franzi, F.; Capella, C.; Ferlazzo, G.; et al. The proangiogenic phenotype of natural killer cells in patients with non-small cell lung cancer. Neoplasia 2013, 15, 133–142. [Google Scholar] [CrossRef] [PubMed]
  150. Kopp, H.G.; Placke, T.; Salih, H.R. Platelet-derived transforming growth factor-beta down-regulates NKG2D thereby inhibiting natural killer cell antitumor reactivity. Cancer Res. 2009, 69, 7775–7783. [Google Scholar] [CrossRef] [PubMed]
  151. Tang, P.M.; Zhou, S.; Meng, X.M.; Wang, Q.M.; Li, C.J.; Lian, G.Y.; Huang, X.R.; Tang, Y.J.; Guan, X.Y.; Yan, B.P.; et al. Smad3 promotes cancer progression by inhibiting E4BP4-mediated NK cell development. Nat. Commun. 2017, 8, 14677. [Google Scholar] [CrossRef]
  152. Pedroza-Pacheco, I.; Madrigal, A.; Saudemont, A. Interaction between natural killer cells and regulatory T cells: Perspectives for immunotherapy. Cell Mol. Immunol. 2013, 10, 222–229. [Google Scholar] [CrossRef] [PubMed]
  153. Li, H.; Han, Y.; Guo, Q.; Zhang, M.; Cao, X. Cancer-expanded myeloid-derived suppressor cells induce anergy of NK cells through membrane- bound TGF-beta 1. J. Immunol. 2009, 182, 240–249. [Google Scholar] [CrossRef] [PubMed]
  154. Sceneay, J.; Chow, M.T.; Chen, A.; Halse, H.M.; Wong, C.S.; Andrews, D.M.; Sloan, E.K.; Parker, B.S.; Bowtell, D.D.; Smyth, M.J.; et al. Primary tumor hypoxia recruits CD11b+/Ly6Cmed/Ly6G+ immune suppressor cells and compromises NK cell cytotoxicity in the premetastatic niche. Cancer Res. 2012, 72, 3906–3911. [Google Scholar] [CrossRef] [PubMed]
  155. Della Chiesa, M.; Carlomagno, S.; Frumento, G.; Balsamo, M.; Cantoni, C.; Conte, R.; Moretta, L.; Moretta, A.; Vitale, M. The tryptophan catabolite L-kynurenine inhibits the surface expression of NKp46- and NKG2D-activating receptors and regulates NK-cell function. Blood 2006, 108, 4118–4125. [Google Scholar] [CrossRef]
  156. Bidwell, B.N.; Slaney, C.Y.; Withana, N.P.; Forster, S.; Cao, Y.; Loi, S.; Andrews, D.; Mikeska, T.; Mangan, N.E.; Samarajiwa, S.A.; et al. Silencing of Irf7 pathways in breast cancer cells promotes bone metastasis through immune escape. Nat. Med. 2012, 18, 1224–1231. [Google Scholar] [CrossRef]
  157. Mlecnik, B.; Bindea, G.; Angell, H.K.; Sasso, M.S.; Obenauf, A.C.; Fredriksen, T.; Lafontaine, L.; Bilocq, A.M.; Kirilovsky, A.; Tosolini, M.; et al. Functional network pipeline reveals genetic determinants associated with in situ lymphocyte proliferation and survival of cancer patients. Sci. Transl. Med. 2014, 6, 228ra37. [Google Scholar] [CrossRef]
  158. Whitley, M.J.; Suwanpradid, J.; Lai, C.; Jiang, S.W.; Cook, J.L.; Zelac, D.E.; Rudolph, R.; Corcoran, D.L.; Degan, S.; Spasojevic, I.; et al. ENTPD1 (CD39) expression inhibits UVR-induced DNA damage repair through purinergic signaling and is associated with metastasis in human cutaneous squamous cell carcinoma. J. Investig. Dermatol. 2021, 141, 2509–2520. [Google Scholar] [CrossRef] [PubMed]
  159. Young, A.; Mittal, D.; Stagg, J.; Smyth, M.J. Targeting cancer derived adenosine: New therapeutic approaches. Cancer Discov. 2014, 4, 879–888. [Google Scholar] [CrossRef]
  160. Beavis, P.A.; Divisekera, U.; Paget, C.; Chow, M.T.; John, L.B.; Devaud, C.; Dwyer, K.; Stagg, J.; Smyth, M.J.; Darcy, P.K. Blockade of A2A receptors potently suppresses the metastasis of CD73+ tumors. Proc. Natl. Acad. Sci. USA 2013, 110, 14711–14716. [Google Scholar] [CrossRef]
  161. Cekic, C.; Day, Y.J.; Sag, D.; Linden, J. Myeloid expression of adenosine A2A receptor suppresses T and NK cell responses in the solid tumor microenvironment. Cancer Res. 2014, 74, 7250–7259. [Google Scholar] [CrossRef]
  162. Young, A.; Ngiow, S.F.; Barkauskas, D.S.; Sult, E.; Hay, C.; Blake, S.J.; Huang, Q.; Liu, J.; Takeda, K.; Teng, M.W.; et al. Co-inhibition of CD73 and A2AR adenosine signaling improves anti-tumor immune responses. Cancer Cell 2016, 30, 391–403. [Google Scholar] [CrossRef]
  163. Berchem, G.; Noman, M.Z.; Bosseler, M.; Paggetti, J.; Baconnais, S.; Le Cam, E.; Nanbakhsh, A.; Moussay, E.; Mami-Chouaib, F.; Janji, B.; et al. Hypoxic tumor-derived microvesicles negatively regulate NK cell function by a mechanism involving TGF-beta and miR23a transfer. Oncoimmunology 2016, 5, e1062968. [Google Scholar] [CrossRef] [PubMed]
  164. Baginska, J.; Viry, E.; Berchem, G.; Poli, A.; Noman, M.Z.; van Moer, K.; Medves, S.; Zimmer, J.A.; Niclou, S.P.; Bleackley, R.C.; et al. Granzyme B degradation by autophagy decreases tumor cell susceptibility to natural killer-mediated lysis under hypoxia. Proc. Natl. Acad. Sci. USA 2013, 110, 17450–17455. [Google Scholar] [CrossRef] [PubMed]
  165. Hatfield, S.M.; Kjaergaard, J.; Lukashev, D.; Schreiber, T.H.; Belikoff, B.; Abbott, R.; Sethumadhavan, S.; Philbrook, P.; Ko, K.; Cannici, R.; et al. Immunological mechanisms of the antitumor effects of supplemental oxygenation. Sci. Transl. Med. 2015, 7, 277ra230. [Google Scholar] [CrossRef] [PubMed]
  166. Sun, X.; Wu, Y.; Gao, W.; Enjyoji, K.; Csizmadia, E.; Muller, C.E.; Murakami, T.; Robson, S.C. CD39/ENTPD1 expression by CD4+Foxp3+ regulatory T cells promotes hepatic metastatic tumor growth in mice. Gastroenterology 2010, 139, 1030–1040. [Google Scholar] [CrossRef]
  167. Faubert, B.; Solmonson, A.; DeBerardinis, R.J. Metabolic reprogramming, and cancer progression. Science 2020, 368, eaaw5473. [Google Scholar] [CrossRef]
  168. Sheppard, S.; Santosa, E.K.; Lau, C.M.; Violante, S.; Giovanelli, P.; Kim, H.; Cross, J.R.; Li, M.O.; Sun, J.C. Lactate dehydrogenase A-dependent aerobic glycolysis promotes natural killer cell anti-viral and anti-tumor function. Cell Rep. 2021, 35, 109210. [Google Scholar] [CrossRef]
  169. Chow, M.T.; Sceneay, J.; Paget, C.; Wong, C.S.; Duret, H.; Tschopp, J.; Moller, A.; Smyth, M.J. NLRP3 suppresses NK cell-mediated responses to carcinogen-induced tumors and metastases. Cancer Res. 2012, 72, 5721–5732. [Google Scholar] [CrossRef] [PubMed]
  170. Dupaul-Chicoine, J.; Arabzadeh, A.; Dagenais, M.; Douglas, T.; Champagne, C.; Morizot, A.; Rodrigue-Gervais, I.G.; Breton, V.; Colpitts, S.L.; Beauchemin, N.; et al. The Nlrp3 inflammasome suppresses colorectal cancer metastatic growth in the liver by promoting natural killer cell tumoricidal activity. Immunity 2015, 43, 751–763. [Google Scholar] [CrossRef]
  171. Shabani, N.; Mylonas, I.; Kunert-Keil, C.; Briese, V.; Janni, W.; Gerber, B.; Friese, K.; Jeschke, U. Expression of glycodelin in human breast cancer: Immunohistochemical analysis in mammary carcinoma in situ, invasive carcinomas and their lymph node metastases. Anticancer Res. 2005, 25, 1761–1764. [Google Scholar]
  172. Okamoto, N.; Uchida, A.; Takakura, K.; Kariya, Y.; Kanzaki, H.; Riittinen, L.; Koistinen, R.; Seppälä, M.; Mori, T. Suppression by human placental protein 14 of natural killer cell activity. Am. J. Reprod. Immunol. 1991, 26, 137–142. [Google Scholar] [CrossRef]
  173. Phan, T.G.; Croucher, P.I. The dormant cancer cell life cycle. Nat. Rev. Cancer 2020, 20, 398–411. [Google Scholar] [CrossRef]
  174. Wang, B.; Wang, Q.; Wang, Z.; Jiang, J.; Yu, S.C.; Ping, Y.F.; Yang, J.; Xu, S.L.; Ye, X.Z.; Xu, C.; et al. Metastatic consequences of immune escape from NK cell cytotoxicity by human breast cancer stem cells. Cancer Res. 2014, 74, 5746–5757. [Google Scholar] [CrossRef]
  175. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer statistics, 2022. CA Cancer J. Clin. 2022, 72, 7–33. [Google Scholar] [CrossRef] [PubMed]
  176. Best, S.A.; Hess, J.B.; Souza-Fonseca-Guimaraes, F.; Cursons, J.; Kersbergen, A.; Dong, X.; Rautela, J.; Hyslop, S.R.; Ritchie, M.E.; Davis, M.J.; et al. Harnessing natural killer immunity in metastatic SCLC. J. Thorac. Oncol. 2020, 15, 1507–1521. [Google Scholar] [CrossRef] [PubMed]
  177. Blake, S.J.; Stannard, K.; Liu, J.; Allen, S.; Yong, M.C.; Mittal, D.; Aguilera, A.R.; Miles, J.J.; Lutzky, V.P.; de Andrade, L.F.; et al. Suppression of metastases using a new lymphocyte checkpoint target for cancer immunotherapy. Cancer Discov. 2016, 6, 446–459. [Google Scholar] [CrossRef]
  178. Ni, J.; Miller, M.; Stojanovic, A.; Garbi, N.; Cerwenka, A. Sustained effector function of IL-12/15/18-preactivated NK cells against established tumors. J. Exp. Med. 2012, 209, 2351–2365. [Google Scholar] [CrossRef] [PubMed]
  179. Song, X.; Voronov, E.; Dvorkin, T.; Fima, E.; Cagnano, E.; Benharroch, D.; Shendler, Y.; Bjorkdahl, O.; Segal, S.; Dinarello, C.A.; et al. Differential effects of IL-1 alpha and IL-1 beta on tumorigenicity patterns and invasiveness. J. Immunol. 2003, 171, 6448–6456. [Google Scholar] [CrossRef]
  180. Sun, R.; Gao, D.S.; Shoush, J.; Lu, B. The IL-1 family in tumorigenesis and antitumor immunity. Semin. Cancer Biol. 2022, 86 Pt 2, 280–295. [Google Scholar] [CrossRef]
  181. Robertson, M.J.; Mier, J.W.; Logan, T.; Atkins, M.; Koon, H.; Koch, K.M.; Kathman, S.; Pandite, L.N.; Oei, C.; Kirby, L.C.; et al. Clinical and biological effects of recombinant human interleukin-18 administered by intravenous infusion to patients with advanced cancer. Clin. Cancer Res. 2006, 12 Pt 1, 4265–4273. [Google Scholar] [CrossRef]
  182. Swann, J.B.; Hayakawa, Y.; Zerafa, N.; Sheehan, K.C.; Scott, B.; Schreiber, R.D.; Hertzog, P.; Smyth, M.J. Type I IFN contributes to NK cell homeostasis, activation, and antitumor function. J. Immunol. 2007, 178, 7540–7549. [Google Scholar] [CrossRef]
  183. Rosenberg, S. Lymphokine-activated killer cells: A new approach to immunotherapy of cancer. J. Natl. Cancer Inst. 1985, 75, 595–603. [Google Scholar]
  184. Yamauchi, A.; Taga, K.; Mostowski, H.S.; Bloom, E.T. Target cell-induced apoptosis of interleukin-2-activated human natural killer cells: Roles of cell surface molecules and intracellular events. Blood 1996, 87, 5127–5135. [Google Scholar] [CrossRef]
  185. Woan, K.V.; Miller, J.S. Harnessing natural killer cell antitumor immunity: From the bench to bedside. Cancer Immunol. Res. 2019, 7, 1742–1747. [Google Scholar] [CrossRef]
  186. Escudier, B.; Farace, F.; Angevin, E.; Charpentier, F.; Nitenberg, G.; Triebel, F.; Hercend, T. Immunotherapy with interleukin-2 (IL2) and lymphokine-activated natural killer cells: Improvement of clinical responses in metastatic renal cell carcinoma patients previously treated with IL2. Eur. J. Cancer 1994, 30, 1078–1083. [Google Scholar] [CrossRef]
  187. Schmidt-Wolf, I.; Lefterova, P.; Mehta, B.A.; Fernandez, L.P.; Huhn, D.; Blume, K.G.; Weissman, I.L.; Negrin, R.S. Phenotypic characterization and identification of effector cells involved in tumor cell recognition of cytokine-induced killer cells. Exp. Hematol. 1993, 21, 1673–1679. [Google Scholar] [PubMed]
  188. Grudzien, M.; Rapak, A. Effect of Natural Compounds on NK Cell Activation. J. Immunol. Res. 2018, 2018, 4868417. [Google Scholar] [CrossRef]
  189. Thiery, J.; Keefe, D.; Saffarian, S.; Martinvalet, D.; Walch, M.; Boucrot, E.; Kirchhausen, T.; Lieberman, J. Perforin activates clathrin- and dynamin-dependent endocytosis, which is required for plasma membrane repair and delivery of granzyme B for granzyme-mediated apoptosis. Blood 2010, 115, 1582–1593. [Google Scholar] [CrossRef] [PubMed]
  190. Bachanova, V.; Cooley, S.; Defor, T.E.; Verneris, M.R.; Zhang, B.; McKenna, D.H.; Curtsinger, J.; Panoskaltsis-Mortari, A.; Lewis, D.; Hippen, K.; et al. Clearance of acute myeloid leukemia by haploidentical natural killer cells is improved using IL-2 diphtheria toxin fusion protein. Blood 2014, 123, 3855–3863. [Google Scholar] [CrossRef] [PubMed]
  191. Levin, A.M.; Bates, D.L.; Ring, A.M.; Krieg, C.; Lin, J.T.; Su, L.; Moraga, I.; Raeber, M.E.; Bowman, G.R.; Novick, P.; et al. Exploiting a natural conformational switch to engineer an interleukin-2 ‘superkine’. Nature 2012, 484, 529–533. [Google Scholar] [CrossRef]
  192. Sim, G.C.; Radvanyi, L. The IL-2 cytokine family in cancer immunotherapy. Cytokine Growth Factor Rev. 2014, 25, 377–390. [Google Scholar] [CrossRef]
  193. Carson, W.E.; Giri, J.G.; Lindemann, M.J.; Linett, M.L.; Ahdieh, M.; Paxton, R.; Anderson, D.; Eisenmann, J.; Grabstein, K.; Caligiuri, M.A. Interleukin (IL) 15 is a novel cytokine that activates human natural killer cells via components of the IL-2 receptor. J. Exp. Med. 1994, 180, 1395–1403. [Google Scholar] [CrossRef] [PubMed]
  194. Carson, W.E.; Ross, M.E.; Baiocchi, R.A.; Marien, M.J.; Boiani, N.; Grabstein, K.; Caligiuri, M.A. Endogenous production of interleukin 15 by activated human monocytes is critical for optimal production of interferon-gamma by natural killer cells in vitro. J. Clin. Investig. 1995, 96, 2578–2582. [Google Scholar] [CrossRef]
  195. Vignali, D.A.; Kuchroo, V.K. IL-12 family cytokines: Immunological playmakers. Nat. Immunol. 2012, 13, 722–728. [Google Scholar] [CrossRef] [PubMed]
  196. Gollob, J.A.; Mier, J.W.; Veenstra, K.; McDermott, D.F.; Clancy, D.; Clancy, M.; Atkins, M.B. Phase I trial of twice-weekly intravenous interleukin 12 in patients with metastatic renal cell cancer or malignant melanoma: Ability to maintain IFN-gamma induction is associated with clinical response. Clin. Cancer Res. 2000, 6, 1678–1692. [Google Scholar]
  197. Smyth, M.J.; Taniguchi, M.; Street, S.E. The anti-tumor activity of IL-12, mechanisms of innate immunity that are model and dose dependent. J. Immunol. 2000, 165, 2665–2670. [Google Scholar] [CrossRef] [PubMed]
  198. Mittal, D.; Vijayan, D.; Putz, E.M.; Aguilera, A.R.; Markey, K.A.; Straube, J.; Kazakoff, S.; Nutt, S.L.; Takeda, K.; Hill, G.R.; et al. Interleukin-12 from CD103+ Batf3-dependent dendritic cells required for NK-cell suppression of metastasis. Cancer Immunol. Res. 2017, 5, 1098–1108. [Google Scholar] [CrossRef]
  199. Bajetta, E.; Del Vecchio, M.; Mortarini, R.; Nadeau, R.; Rakhit, A.; Rimassa, L.; Fowst, C.; Borri, A.; Anichini, A.; Parmiani, G. Pilot study of subcutaneous recombinant human interleukin 12 in metastatic melanoma. Clin. Cancer Res. 1998, 4, 75–85. [Google Scholar]
  200. Leonard, J.P.; Sherman, M.L.; Fisher, G.L.; Buchanan, L.J.; Larsen, G.; Atkins, M.B.; Sosman, J.A.; Dutcher, J.P.; Vogelzang, N.J.; Ryan, J.L. Effects of single-dose interleukin-12 exposure on interleukin-12-associated toxicity and interferon-gamma production. Blood 1997, 90, 2541–2548. [Google Scholar]
  201. Lind, H.; Gameiro, S.R.; Jochems, C.; Donahue, R.N.; Strauss, J.; Gulley, J.L.; Palena, C.; Schlom, J. Dual targeting of TGF-β and PD-L1 via a bifunctional anti-PD-L1/TGF-βRII agent: Status of preclinical and clinical advances. J. Immunother. Cancer 2020, 8, e000433. [Google Scholar] [CrossRef]
  202. Davis, I.D.; Skrumsager, B.K.; Cebon, J.; Nicholaou, T.; Barlow, J.W.; Moller, N.P.; Skak, K.; Lundsgaard, D.; Frederiksen, K.S.; Thygesen, P.; et al. An open-label, two arm, phase I trial of recombinant human interleukin-21 in patients with metastatic melanoma. Clin. Cancer Res. 2007, 13, 3630–3636. [Google Scholar] [CrossRef] [PubMed]
  203. Steele, N.; Anthony, A.; Saunders, M.; Esmarck, B.; Ehrnrooth, E.; Kristjansen, P.E.; Nihlen, A.; Hansen, L.T.; Cassidy, J. A phase 1 trial of recombinant human IL-21 in combination with cetuximab in patients with metastatic colorectal cancer. Br. J. Cancer 2012, 106, 793–798. [Google Scholar] [CrossRef]
  204. Davis, L.D.; Brady, B.; Kefford, R.F.; Millward, M.; Cebon, J.; Skrumsager, B.K.; Mouritzen, U.; Hansen, L.T.; Skak, K.; Lundsgaard, D.; et al. Clinical and biological efficacy of recombinant human interleukin-21 in patients with stage IV malignant melanoma without prior treatment: A phase IIa trial. Clin. Cancer Res. 2009, 15, 2123–2129. [Google Scholar] [CrossRef]
  205. Rahimi Kalateh Shah Mohammad, G.; Ghahremanloo, A.; Soltani, A.; Fathi, E.; Hashemy, S.I. Cytokines as potential combination agents with PD-1/PD-L1 blockade for cancer treatment. J. Cell Physiol. 2020, 235, 5449–5460. [Google Scholar] [CrossRef]
  206. Waldmann, T.A. Cytokines in Cancer Immunotherapy. Cold Spring Harb. Perspect. Biol. 2018, 10, a028472. [Google Scholar] [CrossRef]
  207. Shaw, S.G.; Maung, A.A.; Steptoe, R.J.; Thomson, A.W.; Vujanovic, N.L. Expansion of functional NK cells in multiple tissue compartments of mice treated with Flt3-ligand: Implications for anti-cancer and anti-viral therapy. J. Immunol. 1998, 161, 2817–2824. [Google Scholar] [CrossRef] [PubMed]
  208. Shibuya, A.; Nagayoshi, K.; Nakamura, K.; Nakauchi, H. Lymphokine requirement for the generation of natural killer cells from CD34+ hematopoietic progenitor cells. Blood 1995, 85, 3538–3546. [Google Scholar] [CrossRef]
  209. Neal, Z.C.; Sondel, P.M.; Bates, M.K.; Gillies, S.D.; Herweijer, H. Flt3-L gene therapy enhances immunocytokine-mediated antitumor effects and induces long-term memory. Cancer Immunol. Immunother. 2007, 56, 1765–1774. [Google Scholar] [CrossRef]
  210. Long, S.; Gu, Y.; An, Y.; Lin, X.; Chen, X.; Wang, X.; Liao, C.; Ouyang, W.; Wang, N.; He, Z.; et al. Reovirus enhances cytotoxicity of natural killer cells against colorectal cancer via TLR3 pathway. J. Transl. Med. 2021, 19, 185. [Google Scholar] [CrossRef] [PubMed]
  211. Le Noci, V.; Sommariva, M.; Tortoreto, M.; Zaffaroni, N.; Campiglio, M.; Tagliabue, E.; Balsari, A.; Sfondrini, L. Reprogramming the lung microenvironment by inhaled immunotherapy fosters immune destruction of tumor. OncoImmunology 2016, 5, e1234571. [Google Scholar] [CrossRef]
  212. Yang, J.C.; Sherry, R.M.; Steinberg, S.M.; Topalian, S.L.; Schwartzentruber, D.J.; Hwu, P.; Seipp, C.A.; Rogers-Freezer, L.; Morton, K.E.; White, D.E.; et al. Randomized study of high-dose and low-dose interleukin-2 in patients with metastatic renal cancer. J. Clin. Oncol. 2003, 21, 3127–3132. [Google Scholar] [CrossRef] [PubMed]
  213. Yang, Y.; Lundqvist, A. Immunomodulatory effects of IL-2 and IL-15; implications for cancer immunotherapy. Cancers 2020, 12, 3586. [Google Scholar] [CrossRef]
  214. Peng, L.S.; Penichet, M.L.; Morrison, S.L. A single-chain IL-12 IgG3 antibody fusion protein retains antibody specificity and IL-12 bioactivity and demonstrates antitumor activity. J. Immunol. 1999, 163, 250–258. [Google Scholar] [CrossRef]
  215. Young, P.A.; Morrison, S.L.; Timmerman, J.M. Antibody-cytokine fusion proteins for treatment of cancer: Engineering cytokines for improved efficacy and safety. Semin. Oncol. 2014, 41, 623–636. [Google Scholar] [CrossRef]
  216. Robertson, M.J. Role of chemokines in the biology of natural killer cells. J. Leukoc. Biol. 2002, 71, 173–183. [Google Scholar] [CrossRef]
  217. Lucas, M.; Schachterle, W.; Oberle, K.; Aichele, P.; Diefenbach, A. Dendritic cells prime natural killer cells by trans-presenting interleukin 15. Immunity 2007, 26, 503–517. [Google Scholar] [CrossRef] [PubMed]
  218. Kim, J.; Kim, J.S.; Lee, H.K.; Kim, H.S.; Park, E.J.; Choi, J.E.; Choi, Y.J.; Shin, B.R.; Kim, E.Y.; Hong, J.T.; et al. CXCR3-deficient natural killer cells fail to migrate to B16F10 melanoma cells. Int. Immunopharmacol. 2018, 63, 66–73. [Google Scholar] [CrossRef] [PubMed]
  219. Melaiu, O.; Chierici, M.; Lucarini, V.; Jurman, G.; Conti, L.A.; De Vito, R.; Boldrini, R.; Cifaldi, L.; Castellano, A.; Furlanello, C.; et al. Cellular and gene signatures of tumor-infiltrating dendritic cells and natural-killer cells predict prognosis of neuroblastoma. Nat. Commun. 2020, 11, 5992. [Google Scholar] [CrossRef]
  220. Fernandez, N.C.; Lozier, A.; Flament, C.; Ricciardi-Castagnoli, P.; Bellet, D.; Suter, M.; Perricaudet, M.; Tursz, T.; Maraskovsky, E.; Zitvogel, L. Dendritic cells directly trigger NK cell functions: Cross-talk relevant in innate anti-tumor immune responses in vivo. Nat. Med. 1999, 5, 405–411. [Google Scholar] [CrossRef]
  221. Biber, G.; Sabag, B.; Raiff, A.; Ben-Shmuel, A.; Puthenveetil, A.; Benichou, J.I.C.; Jubany, T.; Levy, M.; Killner, S.; Barda-Saad, M. Modulation of intrinsic inhibitory checkpoints using nano-carriers to unleash NK cell activity. EMBO Mol. Med. 2022, 14, e14073. [Google Scholar] [CrossRef] [PubMed]
  222. Yokoyama, Y.; Lew, E.D.; Seelige, R.; Tindall, E.A.; Walsh, C.; Fagan, P.C.; Lee, J.Y.; Nevarez, R.; Oh, J.; Tucker, K.D.; et al. Immuno-oncological efficacy of RXDX-106, a novel TAM (TYRO3, AXL, MER) family small-molecule kinase inhibitor. Cancer Res. 2019, 79, 1996–2008. [Google Scholar] [CrossRef]
  223. Sanchez-Correa, B.; Lopez-Sejas, N.; Duran, E.; Labella, F.; Alonso, C.; Solana, R.; Tarazona, R. Modulation of NK cells with checkpoint inhibitors in the context of cancer immunotherapy. Cancer Immunol. Immunother. 2019, 68, 861–870. [Google Scholar] [CrossRef]
  224. Semeraro, M.; Galluzzi, L. Novel insights into the mechanism of action of lenalidomide. Oncoimmunology 2014, 3, e28386. [Google Scholar] [CrossRef]
  225. Sehgal, K.; Das, R.; Zhang, L.; Verma, R.; Deng, Y.; Kocoglu, M.; Vasquez, J.; Koduru, S.; Ren, Y.; Wang, M.; et al. Clinical and pharmacodynamic analysis of pomalidomide dosing strategies in myeloma: Impact of immune activation and cereblon targets. Blood 2015, 125, 4042–4051. [Google Scholar] [CrossRef]
  226. Lin, D.J.; Ng, J.C.; Huang, L.; Robinson, M.; O’Hara, J.; Wilson, J.A.; Mellor, A.L. The immunotherapeutic role of indoleamine 2,3-dioxygenase in head and neck squamous cell carcinoma: A systematic review. Clin. Otolaryngol. 2021, 46, 919–934. [Google Scholar] [CrossRef]
  227. Lemos, H.; Huang, L.; Prendergast, G.C.; Mellor, A.L. Immune control by amino acid catabolism during tumorigenesis and therapy. Nat. Rev. Cancer 2019, 19, 162–175. [Google Scholar] [CrossRef]
  228. Lim, C.M.; Liou, A.; Poon, M.; Koh, L.P.; Tan, L.K.; Loh, K.S.; Petersson, B.F.; Ting, E.; Campana, D.; Goh, B.C.; et al. Phase I study of expanded natural killer cells in combination with cetuximab for recurrent/metastatic nasopharyngeal carcinoma. Cancer Immunol. Immunother. 2022, 71, 2277–2286. [Google Scholar] [CrossRef]
  229. Weiner, L.M.; Surana, R.; Wang, S. Monoclonal antibodies: Versatile platforms for cancer immunotherapy. Nat. Rev. Immunol. 2010, 10, 317–327. [Google Scholar] [CrossRef]
  230. Collins, D.M.; Madden, S.F.; Gaynor, N.; AlSultan, D.; Le Gal, M.; Eustace, A.J.; Gately, K.A.; Hughes, C.; Davies, A.M.; Mahgoub, T.; et al. Effects of HER family-targeting tyrosine kinase inhibitors on antibody-dependent cell-mediated cytotoxicity in HER2-expressing breast cancer. Clin. Cancer Res. 2021, 27, 807–818. [Google Scholar] [CrossRef]
  231. Cho, H.; Kim, K.H.; Lee, H.; Kim, C.G.; Chung, H.; Choi, Y.S.; Park, S.H.; Cheong, J.W.; Min, Y.H.; Shin, E.C.; et al. Adaptive Natural Killer Cells Facilitate Effector Functions of Daratumumab in Multiple Myeloma. Clin. Cancer Res. 2021, 27, 2947–2958. [Google Scholar] [CrossRef]
  232. Rosario, M.; Liu, B.; Kong, L.; Collins, L.I.; Schneider, S.E.; Chen, X.; Han, K.; Jeng, E.K.; Rhode, P.R.; Leong, J.W.; et al. The IL-15-based ALT-803 complex enhances FcγRIIIa-triggered NK cell responses and in vivo clearance of B cell lymphomas. Clin. Cancer Res. 2016, 22, 596–608. [Google Scholar] [CrossRef]
  233. Margolin, K.; Morishima, C.; Velcheti, V.; Miller, J.S.; Lee, S.M.; Silk, A.W.; Holtan, S.G.; Lacroix, A.M.; Fling, S.P.; Kaiser, J.C.; et al. Phase I trial of ALT-803, a novel recombinant IL15 complex, in patients with advanced solid tumors. Clin. Cancer Res. 2018, 24, 5552–5561. [Google Scholar] [CrossRef]
  234. Junttila, T.T.; 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, 4481–4489. [Google Scholar] [CrossRef]
  235. Au, K.M.; Park, S.I.; Wang, A.Z. Trispecifc natural killer cell nanoengagers for targeted chemoimmunotherapy. Sci. Adv. 2020, 6, eaba8564. [Google Scholar] [CrossRef]
  236. Davis, Z.B.; Vallera, D.A.; Miller, J.S.; 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. [Google Scholar] [CrossRef]
  237. Li, J.; Piskol, R.; Ybarra, R.; Chen, Y.-J.J.; Li, J.; Slaga, D.; Hristopoulos, M.; Clark, R.; Modrusan, Z.; Totpal, K.; et al. CD3 bispecific antibody–induced cytokine release is dispensable for cytotoxic T cell activity. Sci. Trans. Med. 2019, 11, eaax8861. [Google Scholar] [CrossRef]
  238. Felices, M.; Lenvik, T.R.; Davis, Z.B.; Miller, J.S.; Vallera, D.A. Generation of BiKEs and TriKEs to improve NK cell-mediated targeting of tumor cells. Methods Mol. Biol. 2016, 1441, 333–346. [Google Scholar]
  239. Sarhan, D.; Brandt, L.; Felices, M.; Guldevall, K.; Lenvik, T.; Hinderlie, P.; Curtsinger, J.; Warlick, E.; Spellman, S.R.; Blazar, B.R.; et al. 161533 TriKE stimulates NK-cell function to overcome myeloid-derived suppressor cells in MDS. Blood Adv. 2018, 2, 1459–1469. [Google Scholar] [CrossRef]
  240. 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, 1950264. [Google Scholar] [CrossRef]
  241. Vallera, D.A.; Felices, M.; McElmurry, R.; McCullar, V.; Zhou, X.; Schmohl, J.U.; Zhang, B.; Lenvik, A.J.; Panoskaltsis-Mortari, A.; Verneris, M.R.; 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 Res. 2016, 22, 3440–3450. [Google Scholar] [CrossRef]
  242. Reusing, S.B.; Vallera, D.A.; Manser, A.R.; Vatrin, T.; Bhatia, S.; Felices, M.; Miller, J.S.; Uhrberg, M.; Babor, F. CD16xCD33 bispecifc killer cell engager (BiKE) as potential immunotherapeutic in pediatric patients with AML and biphenotypic ALL. Cancer Immunol. Immunother. 2021, 70, 3701–3708. [Google Scholar] [CrossRef]
  243. Arvindam, U.S.; van Hauten, P.M.M.; Schirm, D.; Schaap, N.; Hobo, W.; Blazar, B.R.; Vallera, D.A.; Dolstra, H.; Felices, M.; Miller, J.S. A trispecific killer engager molecule against CLEC12A effectively induces NK-cell mediated killing of AML cells. Leukemia 2021, 35, 1586–1596. [Google Scholar] [CrossRef]
  244. Groh, V.; Wu, J.; Yee, C.; Spies, T. Tumour-derived soluble MIC ligands impair expression of NKG2D and T-cell activation. Nature 2002, 419, 734–738. [Google Scholar] [CrossRef]
  245. Lopez, K.J.; Cross-Najafi, A.A.; Farag, K.; Obando, B.; Thadasina, D.; Isidan, A.; Park, Y.; Zhang, W.; Ekser, B.; Li, P. Strategies to induce natural killer cell tolerance in xenotransplantation. Front. Immunol. 2022, 13, 941880. [Google Scholar] [CrossRef]
  246. Andre, P.; Denis, C.; Soulas, C.; Bourbon-Caillet, C.; Lopez, J.; Arnoux, T.; Bléry, M.; Bonnafous, C.; Gauthier, L.; Morel, A.; et al. Anti-NKG2A mAb is a checkpoint inhibitor that promotes anti-tumor immunity by unleashing both T and NK cells. Cell 2018, 175, 1731–1743. [Google Scholar] [CrossRef]
  247. van Hall, T.; André, P.; Horowitz, A.; Ruan, D.F.; Borst, L.; Zerbib, R.; Narni-Mancinelli, E.; van der Burg, S.H.; Vivier, E. Monalizumab: Inhibiting the novel immune checkpoint NKG2A. J. Immunother. Cancer 2019, 7, 263. [Google Scholar] [CrossRef]
  248. Benson, D.M., Jr.; Bakan, C.E.; Zhang, S.; Collins, S.M.; Liang, J.; Srivastava, S.; Hofmeister, C.C.; Efebera, Y.; Andre, P.; Romagne, F.; et al. IPH2101, a novel anti-inhibitory KIR antibody, and lenalidomide combine to enhance the natural killer cell versus multiple myeloma effect. Blood 2011, 118, 6387–6391. [Google Scholar] [CrossRef]
  249. Kohrt, H.E.; Thielens, A.; Marabelle, A.; Sagiv-Barfi, I.; Sola, C.; Chanuc, F.; Fuseri, N.; Bonnafous, C.; Czerwinski, D.; Rajapaksa, A.; et al. Anti-KIR antibody enhancement of anti-lymphoma activity of natural killer cells as monotherapy and in combination with anti-CD20 antibodies. Blood 2014, 123, 678–686. [Google Scholar] [CrossRef]
  250. Ellwanger, K.; Reusch, U.; Fucek, I.; Wingert, S.; Ross, T.; Müller, 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, 899–918. [Google Scholar] [CrossRef]
  251. Benson, D.M., Jr.; Hofmeister, C.C.; Padmanabhan, S.; Suvannasankha, A.; Jagannath, S.; Abonour, R.; Bakan, C.; Andre, P.; Efebera, Y.; Tiollier, J.; et al. A phase 1 trial of the anti-KIR antibody IPH2101 in patients with relapsed/refractory multiple myeloma. Blood 2012, 120, 4324–4333. [Google Scholar] [CrossRef]
  252. Armand, P.; Lesokhin, A.; Borrello, I.; Timmerman, J.; Gutierrez, M.; Zhu, L.; Popa McKiver, M.; Ansell, S.M. A phase 1b study of dual PD-1 and CTLA-4 or KIR blockade in patients with relapsed/refractory lymphoid malignancies. Leukemia 2021, 35, 777–786. [Google Scholar] [CrossRef]
  253. Benson, D.M., Jr.; Cohen, A.D.; Jagannath, S.; Munshi, N.C.; Spitzer, G.; Hofmeister, C.C.; Efebera, Y.A.; Andre, P.; Zerbib, R.; Caligiuri, M.A. A phase I trial of the anti-KIR antibody IPH2101 and lenalidomide in patients with relapsed/refractory multiple myeloma. Clin. Cancer Res. 2015, 21, 4055–4061. [Google Scholar] [CrossRef]
  254. Esen, F.; Deniz, G.; Aktas, E.C. PD-1, CTLA-4, LAG-3, and TIGIT: The roles of immune checkpoint receptors on the regulation of human NK cell phenotype and functions. Immunol. Lett. 2021, 240, 15–23. [Google Scholar] [CrossRef]
  255. Anderson, A.C.; Joller, N.; Kuchroo, V.K. Lag-3, Tim-3, and TIGIT: Co-inhibitory receptors with specialized functions in immune regulation. Immunity 2016, 44, 989–1004. [Google Scholar] [CrossRef]
  256. Zhang, Q.; Bi, J.; Zheng, X.; Chen, Y.; Wang, H.; Wu, W.; Wang, Z.; Wu, Q.; Peng, H.; Wei, H.; et al. Blockade of the checkpoint receptor TIGIT prevents NK cell exhaustion and elicits potent anti-tumor immunity. Nat. Immunol. 2018, 19, 723–732. [Google Scholar] [CrossRef]
  257. Chauvin, J.M.; Zarour, H.M. TIGIT in cancer immunotherapy. J. Immunother. Cancer 2020, 8, e000957. [Google Scholar] [CrossRef]
  258. Cho, B.C.; Abreu, D.R.; Hussein, M.; Cobo, M.; Patel, A.J.; Secen, N.; Lee, K.H.; Massuti, B.; Hiret, S.; Yang, J.C.H.; et al. Tiragolumab plus atezolizumab versus placebo plus atezolizumab as a first-line treatment for PD-L1-selected non-small-cell lung cancer (CITYSCAPE): Primary and follow-up analyses of a randomised, double-blind, phase 2 study. Lancet Oncol. 2022, 23, 781–792. [Google Scholar] [CrossRef]
  259. Thibaudin, M.; Limagne, E.; Hampe, L.; Ballot, E.; Truntzer, C.; Ghiringhelli, F. Targeting PD-L1 and TIGIT could restore intratumoral CD8 T cell function in human colorectal cancer. Cancer Immunol. Immunother. 2022, 71, 2549–2563. [Google Scholar] [CrossRef]
  260. Hsu, J.; Hodgins, J.J.; Marathe, M.; Nicolai, C.J.; Bourgeois-Daigneault, M.C.; Trevino, T.N.; Azimi, C.S.; Scheer, A.K.; Randolph, H.E.; Thompson, T.W.; et al. Contribution of NK cells to immunotherapy mediated by PD-1/PD-L1 blockade. J. Clin. Investig. 2018, 128, 4654–4668. [Google Scholar] [CrossRef]
  261. Igarashi, T.; Wynberg, J.; Srinivasan, R.; Becknell, B.; McCoy, J.P., Jr.; Takahashi, Y.; Suffredini, D.A.; Linehan, W.M.; Caligiuri, M.A.; Childs, R.W. Enhanced cytotoxicity of allogeneic NK cells with killer immunoglobulin-like receptor ligand incompatibility against melanoma and renal cell carcinoma cells. Blood 2004, 104, 170–177. [Google Scholar] [CrossRef]
  262. Delgado, D.; Webster, D.E.; DeSantes, K.B.; Durkin, E.T.; Shaaban, A.F. KIR receptor-ligand incompatibility predicts killing of osteosarcoma cell lines by allogeneic NK cells. Pediatr. Blood Cancer 2010, 55, 1300–1305. [Google Scholar] [CrossRef]
  263. Grote, S.; Ureña-Bailén, G.; Chan, K.C.; Baden, C.; Mezger, M.; Handgretinger, R.; Schleicher, S. In vitro evaluation of CD276-CAR NK-92 functionality, migration and invasion potential in the presence of immune Inhibitory factors of the tumor microenvironment. Cells 2021, 10, 1020. [Google Scholar] [CrossRef]
  264. Soltantoyeh, T.; Akbari, B.; Karimi, A.; Mahmoodi Chalbatani, G.; Ghahri-Saremi, N.; Hadjati, J.; Hamblin, M.R.; Mirzaei, H.R. Chimeric antigen receptor (CAR) T cell therapy for metastatic melanoma: Challenges and road ahead. Cells 2021, 10, 1450. [Google Scholar] [CrossRef]
  265. Daher, M.; Rezvani, K. Outlook for new CAR-based therapies with a focus on CAR NK cells: What lies beyond CAR-engineered T cells in the race against cancer. Cancer Discov. 2020, 11, 45–58. [Google Scholar] [CrossRef]
  266. Azoulay, E.; Shimabukuro-Vornhagen, A.; Darmon, M.; von Bergwelt-Baildon, M. Critical care management of chimeric antigen receptor T cell–related toxicity. Be aware and prepared. Am. J. Respir. Crit. Care Med. 2019, 200, 20–23. [Google Scholar] [CrossRef]
  267. Daher, M.; Melo Garcia, L.; Li, Y.; Rezvani, K. CAR-NK cells: The next wave of cellular therapy for cancer. Clin. Transl. Immunol. 2021, 10, e1274. [Google Scholar] [CrossRef]
  268. Siegler, E.L.; Zhu, Y.; Wang, P.; Yang, L. Of-the-shelf CAR-NK cells for cancer immunotherapy. Cell Stem Cell 2018, 23, 160–161. [Google Scholar] [CrossRef]
  269. Klingemann, H.; Boissel, L.; Toneguzzo, F. Natural killer cells for immunotherapy–advantages of the NK-92 cell line over blood NK cells. Front. Immunol. 2016, 7, 91. [Google Scholar] [CrossRef]
  270. Li, Y.; Hermanson, D.L.; Moriarity, B.S.; Kaufman, D.S. Human iPSC-derived natural killer cells engineered with chimeric antigen receptors enhance anti-tumor activity. Cell Stem Cell 2018, 23, 181–192.e5. [Google Scholar] [CrossRef]
  271. Gong, Y.; Klein Wolterink, R.G.J.; Wang, J.; Bos, G.M.J.; Germeraad, W.T.V. Chimeric antigen receptor natural killer (CAR-NK) cell design and engineering for cancer therapy. J. Hematol. Oncol. 2021, 14, 73. [Google Scholar] [CrossRef]
  272. Stoiber, S.; Cadilha, B.L.; Benmebarek, M.R.; Lesch, S.; Endres, S.; Kobold, S. Limitations in the design of chimeric antigen receptors for cancer therapy. Cells 2019, 8, 472. [Google Scholar] [CrossRef]
  273. Liu, S.; Galat, V.; Galat, Y.; Lee, Y.K.A.; Wainwright, D.; Wu, J. NK cell-based cancer immunotherapy: From basic biology to clinical development. J. Hematol. Oncol. 2021, 14, 7. [Google Scholar] [CrossRef]
  274. Marof, F.; Abdul-Rasheed, O.F.; Rahman, H.S.; Budi, H.S.; Jalil, A.T.; Yumashev, A.V.; Hassanzadeh, A.; Yazdanifar, M.; Motavalli, R.; Chartrand, M.S.; et al. CAR-NK cell in cancer immunotherapy; A promising frontier. Cancer Sci. 2021, 112, 3427–3436. [Google Scholar] [CrossRef]
  275. Matosevic, S. Viral and nonviral engineering of natural killer cells as emerging adoptive cancer immunotherapies. J. Immunol. Res. 2018, 2018, 4054815. [Google Scholar] [CrossRef]
  276. Rafei, H.; Daher, M.; Rezvani, K. Chimeric antigen receptor (CAR) natural killer (NK)-cell therapy: Leveraging the power of innate immunity. Br. J. Haematol. 2021, 193, 216–230. [Google Scholar] [CrossRef] [PubMed]
  277. Gauthier, L.; Virone-Oddos, A.; Beninga, J.; Rossi, B.; Nicolazzi, C.; Amara, C.; Blanchard-Alvarez, A.; Gourdin, N.; Courta, J.; Basset, A.; et al. Control of acute myeloid leukemia by a trifunctional NKp46-CD16a-NK cell engager targeting CD123. Nat. Biotechnol. 2023. [Google Scholar] [CrossRef]
  278. Yanakieva, D.; Pekar, L.; Evers, A.; Fleischer, M.; Keller, S.; Mueller-Pompalla, D.; Toleikis, L.; Kolmar, H.; Zielonka, S.; Krah, S. Beyond bispecificity: Controlled Fab arm exchange for the generation of antibodies with multiple specificities. mAbs 2022, 14, 2018960. [Google Scholar] [CrossRef]
  279. Schmohl, J.U.; Felices, M.; Todhunter, D.; Taras, E.; Miller, J.S.; Vallera, D.A. Tetraspecific scFv construct provides NK cell mediated ADCC and self-sustaining stimuli via insertion of IL-15 as a cross-linker. Oncotarget 2016, 7, 73830–73844. [Google Scholar] [CrossRef]
  280. Hamieh, M.; Dobrin, A.; Cabriolu, A.; van der Stegen, S.J.C.; Giavridis, T.; Mansilla-Soto, J.; Eyquem, J.; Zhao, Z.; Whitlock, B.M.; Miele, M.M.; et al. CAR T cell trogocytosis and cooperative killing regulate tumour antigen escape. Nature 2019, 568, 112–116. [Google Scholar] [CrossRef]
  281. Li, H.S.; Wong, N.M.; Tague, E.; Ngo, J.T.; Khalil, A.S.; Wong, W.W. High-performance multiplex drug-gated CAR circuits. Cancer Cell 2022, 40, 1294–1305.e4. [Google Scholar] [CrossRef]
Figure 1. The human immune system and its functions. (A) The immune system consists of organs (thymus, tonsils, spleen, lymph, lymphatic tissue of the gut and bone marrow), immune cells (T cells, NK cells, B cells, macrophage, etc.), and molecules (antibodies, cytokines, chemokines, etc.). It protects against foreign pathogens, eliminates cancer cells, and maintains normal body functions. (B) Two primary arms of effector immunity are innate and adaptive immunities for immune response. Innate immunity is nonspecific and refers to defense mechanisms activated rapidly to prevent the spread of foreign antigens, including those produced by cancer cells. The main cell types involved in innate immunity are natural killer cells (NK), dendritic cells (DC), and macrophages. NK cells are activated by cancers through NK cell activating receptor interaction with its ligands in cancer cells, releasing perforin and other killer molecules to trigger cancer cell death. Adaptive (acquired) immunity comprises the second line of defense, responding to particular ‘non-self’ antigens expressed in cancer cells. It is characterized by clonal expansion of T and B lymphocytes (T cell and B cell); upon expansion, these clonal cells express the same antigen receptor and are primed to fight the same pathogen. B lymphocytes are primarily involved in antibody-mediated immunity, while T lymphocytes are mainly associated with cell-mediated immunity. After activation through interaction, T cells and tumor antigens represented by DC, CD8+ T cells become cytotoxicity T cells, and CD4+ T cells release cytokines to stimulate CD8+ T cells to convert to cytotoxicity T cells. Cytotoxicity T cells recognize the cancer cells and mediate cancer cell clearance through direct cytotoxicity or secretion of inflammatory cytokines.
Figure 1. The human immune system and its functions. (A) The immune system consists of organs (thymus, tonsils, spleen, lymph, lymphatic tissue of the gut and bone marrow), immune cells (T cells, NK cells, B cells, macrophage, etc.), and molecules (antibodies, cytokines, chemokines, etc.). It protects against foreign pathogens, eliminates cancer cells, and maintains normal body functions. (B) Two primary arms of effector immunity are innate and adaptive immunities for immune response. Innate immunity is nonspecific and refers to defense mechanisms activated rapidly to prevent the spread of foreign antigens, including those produced by cancer cells. The main cell types involved in innate immunity are natural killer cells (NK), dendritic cells (DC), and macrophages. NK cells are activated by cancers through NK cell activating receptor interaction with its ligands in cancer cells, releasing perforin and other killer molecules to trigger cancer cell death. Adaptive (acquired) immunity comprises the second line of defense, responding to particular ‘non-self’ antigens expressed in cancer cells. It is characterized by clonal expansion of T and B lymphocytes (T cell and B cell); upon expansion, these clonal cells express the same antigen receptor and are primed to fight the same pathogen. B lymphocytes are primarily involved in antibody-mediated immunity, while T lymphocytes are mainly associated with cell-mediated immunity. After activation through interaction, T cells and tumor antigens represented by DC, CD8+ T cells become cytotoxicity T cells, and CD4+ T cells release cytokines to stimulate CD8+ T cells to convert to cytotoxicity T cells. Cytotoxicity T cells recognize the cancer cells and mediate cancer cell clearance through direct cytotoxicity or secretion of inflammatory cytokines.
Cancers 15 02323 g001
Figure 2. Interactions between the immune system and tumor cells can determine the fate of tumors. Interactions between the immune system and tumor cells are like the relationship between a mouse and a cat game. This process is called cancer immunoediting, which consists of three “Es”: eradication, equilibrium, and escape. In eradication, tumor cells are initially destroyed by the immune system by activating both innate and adaptive mechanisms. However, some tumor cells manage to survive immune destruction and enter what could be a lengthy equilibrium phase. Immunologically sculpted tumors grow progressively during the final escape phase, become clinically mass or metastasize, and establish an immunosuppressive TME.
Figure 2. Interactions between the immune system and tumor cells can determine the fate of tumors. Interactions between the immune system and tumor cells are like the relationship between a mouse and a cat game. This process is called cancer immunoediting, which consists of three “Es”: eradication, equilibrium, and escape. In eradication, tumor cells are initially destroyed by the immune system by activating both innate and adaptive mechanisms. However, some tumor cells manage to survive immune destruction and enter what could be a lengthy equilibrium phase. Immunologically sculpted tumors grow progressively during the final escape phase, become clinically mass or metastasize, and establish an immunosuppressive TME.
Cancers 15 02323 g002
Figure 3. NK cell activity is precisely regulated by a dynamic balance of signals transduced by a diverse set of activating and inhibitory receptors expressed on NK cells. NK cells express an array of activating and inhibitory receptors. Both activating and inhibitory receptor signals jointly mediate the outcome of NK cell target metastatic tumor cells. The activating receptors encounter the ligands expressed in metastatic tumor cells, activate the immunoreceptor tyrosine-based activation motif (ITAM), and activate PI3K and PLC through adaptors such as Syk, Vav1, Zap70, Lck, and Fyn, therefore activating transcriptional factors for stimulating gene expression and promote NK proliferation and release killing molecules for cytotoxicity. As another highly efficient activating receptor, CD16 induces activation signals and triggers antibody-dependent cellular cytotoxicity (ADCC) against antibody-coated cancer cells following binding with the Fc region of antibodies. Moreover, NK cells are activated by cytokines through cytokine receptors. The inhibitory receptors contain the immunoreceptor tyrosine-based inhibition motif (ITIM) in the cytoplasmic tail. After binding to the ligands in metastatic tumor cells, the inhibitory receptors trigger an inhibitory signal through the immunoreceptor tyrosine-based inhibition motif (ITIM), requite the SHP-1 and delivering the inhibition signals through adaptors molecules, therefore blocking gene expression and NK cell proliferation and maintaining NK cell self-tolerance.
Figure 3. NK cell activity is precisely regulated by a dynamic balance of signals transduced by a diverse set of activating and inhibitory receptors expressed on NK cells. NK cells express an array of activating and inhibitory receptors. Both activating and inhibitory receptor signals jointly mediate the outcome of NK cell target metastatic tumor cells. The activating receptors encounter the ligands expressed in metastatic tumor cells, activate the immunoreceptor tyrosine-based activation motif (ITAM), and activate PI3K and PLC through adaptors such as Syk, Vav1, Zap70, Lck, and Fyn, therefore activating transcriptional factors for stimulating gene expression and promote NK proliferation and release killing molecules for cytotoxicity. As another highly efficient activating receptor, CD16 induces activation signals and triggers antibody-dependent cellular cytotoxicity (ADCC) against antibody-coated cancer cells following binding with the Fc region of antibodies. Moreover, NK cells are activated by cytokines through cytokine receptors. The inhibitory receptors contain the immunoreceptor tyrosine-based inhibition motif (ITIM) in the cytoplasmic tail. After binding to the ligands in metastatic tumor cells, the inhibitory receptors trigger an inhibitory signal through the immunoreceptor tyrosine-based inhibition motif (ITIM), requite the SHP-1 and delivering the inhibition signals through adaptors molecules, therefore blocking gene expression and NK cell proliferation and maintaining NK cell self-tolerance.
Cancers 15 02323 g003
Figure 4. Enhancement of NK cell cytotoxicity (antibody-dependent cellular cytotoxicity, ADCC). (A) Matured NK cells express activating receptor CD16 (FcγRIII) that crosslinks with the Fc region of antibodies and initiates antibody-dependent cellular cytotoxicity (ADCC), killing the antibody-coated metastatic cancer cells. (B) For promoting the efficacy of ADCC, a novel bispecific IgG1-scFv fusion antibody targeting CD16a on NK cells and specific tumor antigens (bispecific) on metastatic tumor cells has been developed as a second-generation antibody with improved ADCC potential. (C) Multifunctional antibodies combined anti-CD16 scFv with IL15 and antimetastatic tumor scFv (bispecific) or anti-CD16 scFv with two antimetastatic tumor scFvs (trispecific) to amplify NK cells and enhance their mediated killing of antigen-expressed metastatic tumor cells. This approach has also been used to physically bridge malignant cells and immune effectors through bispecific killer engagers (BiKEs) or trispecific killer engagers (TriKEs) to increase specificity and facilitate more direct interaction between immune cells and tumors while decreasing systemic toxicity.
Figure 4. Enhancement of NK cell cytotoxicity (antibody-dependent cellular cytotoxicity, ADCC). (A) Matured NK cells express activating receptor CD16 (FcγRIII) that crosslinks with the Fc region of antibodies and initiates antibody-dependent cellular cytotoxicity (ADCC), killing the antibody-coated metastatic cancer cells. (B) For promoting the efficacy of ADCC, a novel bispecific IgG1-scFv fusion antibody targeting CD16a on NK cells and specific tumor antigens (bispecific) on metastatic tumor cells has been developed as a second-generation antibody with improved ADCC potential. (C) Multifunctional antibodies combined anti-CD16 scFv with IL15 and antimetastatic tumor scFv (bispecific) or anti-CD16 scFv with two antimetastatic tumor scFvs (trispecific) to amplify NK cells and enhance their mediated killing of antigen-expressed metastatic tumor cells. This approach has also been used to physically bridge malignant cells and immune effectors through bispecific killer engagers (BiKEs) or trispecific killer engagers (TriKEs) to increase specificity and facilitate more direct interaction between immune cells and tumors while decreasing systemic toxicity.
Cancers 15 02323 g004
Figure 5. Properties of CAR-NK cells and CAR-T cells. CAR-NK cell-based therapy is superior to CAR-T cell therapy concerning allergenicity, tumor killing, and toxicity. Moreover, CAR-NK cell-based therapy could combine with multiple strategies of various activating receptors.
Figure 5. Properties of CAR-NK cells and CAR-T cells. CAR-NK cell-based therapy is superior to CAR-T cell therapy concerning allergenicity, tumor killing, and toxicity. Moreover, CAR-NK cell-based therapy could combine with multiple strategies of various activating receptors.
Cancers 15 02323 g005
Figure 6. Engineered NK cells (CAR-NK) for metastasis therapy. CARs for NK cells consist of a tumor antigen binding domain (scFv with a linker between VH and VL), a hinge region, a transmembrane domain, and an intracellular activation signal domain. The tumor antigen binding domain contains single chain variable fragments (scFv) derived from antibody recognize tumor-specific antigens. The hinge region links to a transmembrane domain to promote the dimerization of CAR, increase cytokine production and cell proliferation of CAR, and enhance persistence and antitumor effects in vivo. The transmembrane domain allows the CAR structure to anchor on the NK membrane. Recognition of tumor cells by CARs through the tumor antigen binding domain triggers the activation domain signal and leads to tumor cell death. The scFv binds and recognizes the tumor-specific antigen, then activate CAR-NK cells’ intracellular activation domain triggering functional activation and proliferation.
Figure 6. Engineered NK cells (CAR-NK) for metastasis therapy. CARs for NK cells consist of a tumor antigen binding domain (scFv with a linker between VH and VL), a hinge region, a transmembrane domain, and an intracellular activation signal domain. The tumor antigen binding domain contains single chain variable fragments (scFv) derived from antibody recognize tumor-specific antigens. The hinge region links to a transmembrane domain to promote the dimerization of CAR, increase cytokine production and cell proliferation of CAR, and enhance persistence and antitumor effects in vivo. The transmembrane domain allows the CAR structure to anchor on the NK membrane. Recognition of tumor cells by CARs through the tumor antigen binding domain triggers the activation domain signal and leads to tumor cell death. The scFv binds and recognizes the tumor-specific antigen, then activate CAR-NK cells’ intracellular activation domain triggering functional activation and proliferation.
Cancers 15 02323 g006
Figure 7. NK cell-based immunotherapy for metastasis therapy. NK cells are known for killing tumor cells without prior stimulation, thus participating in the first line of defense against malignant transformation and metastasis. Multiple strategies aimed at activating NK cell immunosurveillance have demonstrated substantial therapeutic effects in preclinical models of metastatic dissemination. Most therapeutic strategies currently employed in the clinic stem from restoring NK cell activation-dependent immune responses against metastatic cancers.
Figure 7. NK cell-based immunotherapy for metastasis therapy. NK cells are known for killing tumor cells without prior stimulation, thus participating in the first line of defense against malignant transformation and metastasis. Multiple strategies aimed at activating NK cell immunosurveillance have demonstrated substantial therapeutic effects in preclinical models of metastatic dissemination. Most therapeutic strategies currently employed in the clinic stem from restoring NK cell activation-dependent immune responses against metastatic cancers.
Cancers 15 02323 g007
Table 1. Properties of NK cells and T cells in antitumor activity.
Table 1. Properties of NK cells and T cells in antitumor activity.
PropertyNK CellT Cells
Type of immune responseInnate immunity, Adaptive ImmunityAdaptive immunity
Cell markerCD56+CD3-CD16+CD3+CD4+ or CD3+CD8+
Development Differentiate in the BMDifferentiate in the thymus
pre-stimulationNo antigen primingAntigen priming required
LocationPrimarily in blood and tissue resident Antigen specific tumor site
Side effect of overactivationNo/low risk of GVHDAllogeneic T cells induce GVHD
Mechanism of activationRecognition of abnormal or missing self-molecules on the surface of cancer cells through the complex array of receptorsAntigen-specific recognition of of cancer cells through TCR
MHC dependenceDo not require MHC matching for antigen recognition and activationRequire MHC matching for antigen recognition and activation
CytotoxicityDirectly kill cancer cells through the release of cytotoxic granules, greater cytotoxicityDirectly kill cancer cells through the release of cytotoxic granules
Cytokine secretionSecrete cytokines to stimulate other immune cells to attack cancer cellsSecrete cytokines that stimulate other immune cells to attack cancer cells
TargetTarget various cancer cells that have lost MHC expression or express stress-induced moleculesTarget cancer cells that express specific antigens on their surface
ADCCmediated antibody-dependent cellular cytotoxicity of cancer cellsNo
Anti-metastatic activityHighly effective against dissimilating tumor cells and distant site tumor cellsHave limited efficacy against metastatic tumor cells
Table 2. Progress of multiple-specific functional (bispecific, trispecific, and tetraspeicific) antibodies in NK cell-based therapy.
Table 2. Progress of multiple-specific functional (bispecific, trispecific, and tetraspeicific) antibodies in NK cell-based therapy.
Name TargetFormatMechanismDisease Status
AFM-13CD30/CD16 scFv-scFv (BiKE)CD30 inhibitor CD16 regulatorrelapsed or refractory MMphase II
AFM-24EGFR/CD16 scFv-scFv (BiKE)EGFR blockers CD16 regulatoradvanced solid tumorsphase II
AFM26BCMA/CD16 scFv-scFv (BiKE)BCMA blocker CD16 regulatorrelapsed or refractory MMphase I/II
6MW3411PD-L1/CD16 scFv-scFv (BiKE)PD-L1 inhibitors CD16 NK cell recruitment agentsolid tumorpre-clinical
HRS-3/A9 or Anti-CD16/CD30 BiMABCD16/CD30 scFv-scFv (BiKE)CD30 regulator CD16 regulatorhodgkin’s diseasepre-clinical
NKp46 NKCENCR1/CD16 scFv-scFv (BiKE)NCR1 inhibitor CD16 NK cell recruitment agenttumorpre-clinical
161533/GTB3550/OXS3550CD16/IL15/CD33 scFv-IL15-scFv (TriKE)fusion protein trifunctional, NK cell stimulant, CD16 regulator, CD33 inhibitorHigh -risk MDS, relapsed or refractory AMLphase II
cam161533 TriKECD16/IL15/CD33 scFv-IL15-scFv (TriKE)CD33 regulator IL15 regulator CD16 regulatorHigh -risk MDS, relapsed or refractory AMLpre-clinical
GTB-3650 (humanized CD16scFv)CD16/IL-15/CD33 scFv-IL15-scFv (TriKE)fusion protein trifunctional, NK cell stimulant, CD16 regulator, CD33 inhibitorAML, MDSpre-clinical
CD16-IL15-CLEC12A CD16/IL15/CLEC12A scFv-IL15-scFv (TriKE)NK cell stimulant ADCC effectacute myeloid leukemia, Leukemic stem cellspre-clinical
triplebodyNKG2D/CD19/CD33 ULPB2-scFv-scFv (TriKE)activating NK cell, CD19 and CD33 inhibitorsmixed lineage leukemia (MLL)pre-clinical
triplebodyCD33/CD16/CD19scFv-scFv-scFv (TriKE)CD16 regulator, CD19 and CD33 inhibitors ADCCMLLpre-clinical
sctbCD123/CD16/CD33scFv-scFv-scFv (TriKE)CD16 regulator, CD123 and CD133 inhibitors, ADCCAMLpre-clinical
SPM-2CD33/CD16/CD123scFv-scFv-scFv (TriKE)CD16 regulator, CD19 and CD133 inhibitors, ADCCAMLpre-clinical
TriKECD16/CD22/CD19scFv-scFv-scFv (TriKE)CD16 regulator, CD19 and CD22 inhibitors, ADCCB-ALL, B-CLL, AMLpre-clinical
ATriFlexBCMA/CD200/CD16AscFv-diabody-scFv (TriKE)CD16 regulator, CD200 and BCMA inhibitors, ADCCMMpre-clinical
SAR443579 (ANKET)CD123/CD16/NKp46NKp46-Fc-CD123 (TriKE)fusion protein trifunctional, NK cell stimulant, CD16 regulator, CD123 inhibitorAML, MDSpre-clinical
1615EpCAMCD16/IL-15/EpCAMscFv-IL15-scFv (TriKE)fusion protein trifunctional, NK cell stimulant, CD16 regulator, EpCAM inhibitorVarious carcinomaspre-clinical
1615133CD16/IL-15/CD133scFv-IL15-scFv (TriKE)fusion protein trifunctional, NK cell stimulant, CD16 regulator, CD133 inhibitorCancer stem cellspre-clinical
161519CD16/IL-15/CD19scFv-IL15-scFv (TriKE)fusion protein trifunctional, NK cell stimulant, CD16 regulator, CD19 inhibitorB-CLLpre-clinical
cam1615B7H3CD16/IL-15/B7H3VHH-IL15-scFv (TriKE)fusion protein trifunctional, NK cell stimulant, CD16 regulator, B7H3 inhibitorOvarian cancerpre-clinical
cam1615HER2CD16/IL-15/HER2VHH-IL15-scFv (TriKE)fusion protein trifunctional, NK cell stimulant, CD16 regulator, HER2 inhibitorOvarian cancerpre-clinical
cam1615TEM8CD16/IL-15/TEM8VHH-IL15-scFv (TriKE)fusion protein trifunctional, NK cell stimulant, CD16 regulator, TEM8 inhibitorNSCLC, solid tumorpre-clinical
TetraKE (TtsAb)CD16/IL-5/EpCAM/CD133scFv-IL15-scFv-scFv (tetraKE)fusion protein trifunctional, NK cell stimulant, CD16 regulator, CD133 and EpCAM inhibitorCRCpre-clinical
SEEDbodyEGFR/HER2/NKG2DIgG-like VHH-based (NKCE)activating NK cell, EGFR and HER2inhibitorsBreast cancerpre-clinical
TsAbEGFR/CD16a/PD-L1Bs IgG-Fab (NKCE)EGFR blocker, PD-L1 blocker and CD16 regulator, ADCCEpidermoid carcinomapre-clinical
ANKETNKp46/CD16/CD19-CD20-EGFRFab-Fc-Fab (NKCE)activating NK cell, CD16 regulator CD19 and CD20 EGFR inhibitorsLNHpre-clinical
ANKETNKp46-NKp30/CD16/CD19-CD20Fab-Fc-Fab(NKCE)activating NK cell, CD16 regulator CD19 and CD20 inhibitorsB-ALLpre-clinical
ANKET4NKp46/CD16/CD20/IL-2vFab-Fc-Fab (NKCE)activating NK cell, CD16 regulator CD20 inhibitorsB-ALLpre-clinical
HLE-nano-BiKECD38/CD16/HSAVHH-VHH-VHH (nano)fusion protein trifunctional, NK cell stimulant, CD16 regulator, CD38 inhibitormultiple myelomapre-clinical
DuoBody (DB)-VHH (TtsAb)HER2/cMET/EGFR-IL6R-NKG2Dbs IgG-VHH-VHH (NKCE)fusion protein trifunctional, NK cell stimulant, HER2, cMet, EGFR inhibitorBreast cancerpre-clinical
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Yu, Y. The Function of NK Cells in Tumor Metastasis and NK Cell-Based Immunotherapy. Cancers 2023, 15, 2323. https://doi.org/10.3390/cancers15082323

AMA Style

Yu Y. The Function of NK Cells in Tumor Metastasis and NK Cell-Based Immunotherapy. Cancers. 2023; 15(8):2323. https://doi.org/10.3390/cancers15082323

Chicago/Turabian Style

Yu, Yanlin. 2023. "The Function of NK Cells in Tumor Metastasis and NK Cell-Based Immunotherapy" Cancers 15, no. 8: 2323. https://doi.org/10.3390/cancers15082323

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop