Abstract
Natural killer (NK) cells can elicit an immune response against malignantly transformed cells without recognizing antigens, and they also exhibit cytotoxic effects and immune surveillance functions in tumor immunotherapy. Although several studies have shown the promising antitumor effects of NK cells in immunotherapy, their function is often limited in the tumor microenvironment because tumor cells can easily escape NK cell-induced death. Thus, for efficient tumor immunotherapy, the mechanism by which tumor cells escape NK cell-induced cytotoxicity must be fully understood. Various novel molecules and checkpoint receptors that mediate the disruption of NK cells in the tumor microenvironment have been discovered. In this review, we analyze and detail the major activating and inhibitory receptors on the surface of NK cells to delineate the mechanism by which tumor cells suppress NKG2D ligand expression and increase tumor receptor and inhibitory receptor expression [NKG2A, programmed cell death 1 (PD-1), and T-cell immunoglobulin and immunoreceptor tyrosine inhibitory motif (TIGIT)] on the NK cell surface, and thus inhibit NK cell activity. We also reviewed the current status of treatments based on these surface molecules. By comparing the therapeutic effects related to the treatment status and bypass mechanisms, we attempt to identify optimal single or combined treatments to suggest new treatment strategies for tumor immunotherapy.
Keywords: Immune evasion, natural killer cell, NKG2D, PD-1, combination therapy
Introduction
Tumor immunotherapy has revolutionized the field of tumor treatment. The use of immune checkpoint blockade (ICB) has been a major breakthrough in tumor immunotherapy; however, it has achieved success in cancer treatment for only a fraction of patients (1). Moreover, its application is limited in clinical practice, thus hindering the broad use of immunotherapy. Immune evasion contributes significantly to tumor progression and metastasis (2). The mechanisms of immune evasion must be investigated to identify novel targets, provide a deeper understanding of the mechanisms underlying ICB resistance, and help develop therapeutic strategies for overcoming the current immunotherapy bottleneck.
Chimeric antigen receptor (CAR) T-cell therapies are cutting edge immunotherapy techniques; however, they have been associated with adverse events, such as neurotoxicity and cytokine release syndrome (3). Moreover, CAR-T cells are expensive and complex to produce because they are manufactured specifically for each patient. Natural killer (NK) cells have similar biological properties to T-cells but are not truly antigen-specific and respond early to tumor-related threats; therefore, research interest in NK cells as candidates for immunotherapy has grown exponentially (4). NK cells, which are innate immune cells, kill virus-infected and malignant cells without recognizing antigens, and they present immune surveillance and cytotoxic functions in tumor immunotherapy (5). Hence CAR-NK cell therapies have rapidly attracted the attention of immunologists. Clinical trials using cord blood-derived CAR-NK cells have demonstrated clinical efficacy (6). Moreover, the use of CAR-NK cells in vivo does not induce the development of adverse events associated with CAR-T cells (6). These encouraging results have laid a foundation for the application of NK cells in future immunotherapy strategies.
Tumors present several mechanisms of evading the immune system’s surveillance, including activation of immune checkpoints that can restrict NK cell function. Although many NK cell-based clinical trials are ongoing, the results have not always been satisfactory. These antitumor strategies need to overcome the challenge of decline in NK cell activity associated with mechanisms that bypass the immune system (7). The activation of NK cells is relatively straightforward, and the overall mechanism of immune escape might not involve the complex antigen presentation of T-cells (1). Therefore, summarizing the mechanism of NK cell-based immune evasion might provide insights for personalized treatment and more reasonable and effective combined immunotherapy. Currently, tumor immunotherapy approaches include administration of single or combination target drugs or adoptive immunotherapy. Therefore, an analysis from the perspective of NK cell-based immune evasion can facilitate the development of improved treatment methods.
The balance between the various activating and inhibitory receptor-mediated signals in NK cells determines the suppression or induction of NK cell function (8). NKG2D and NKG2A are representative NK cell activating and inhibitor receptors, respectively. Jhunjhunwala et al. conducted an in-depth analysis on immune evasion and suggested that NK cell-based immune evasion is represented by downregulated NK cell activity accompanied by changes in histocompatibility leukocyte antigen E (HLA-E) expression, NKG2D downregulation by tumor cell shedding of major histocompatibility complex class I (MHC I) polypeptide-related sequence A and B (MICA/B), and immune checkpoint upregulation, such as programmed cell death 1 (PD-1) and T-cell immunoglobulin and immunoreceptor tyrosine inhibitory motif (TIGIT) (1). Here, we provide an overview of the mechanisms underlying immune evasion and describe the current status of therapies and treatment options based on these mechanisms.
Mechanism of NK cell-based immune evasion
Mechanism based on NKG2D-NKG2DL axis
NKG2D is an essential activating receptor expressed on the surface of NK cells and many T-cells. The YXXM (Tyr.X.X.Meth) sequence in the cytoplasmic domain of DAP10 recruits growth factor receptor binding protein 2 (GRB2) and phosphatidylinositol 3-kinase (PI3K) (9). Activation of these two signaling pathways leads to an increased concentration of intracellular calcium and restructuring of the actin cytoskeleton in NK cells. Eventually, immune synapses form among tumor cells and NK cells (10), with NK cells acting via several key pathways that mediate the death of target cells.
Although NKG2D ligands are overexpressed in tumor cells, they are generally not expressed in normal tissues (11); however, they are upregulated following DNA damage, inflammation, and malignant transformation. Therefore, NKG2D ligands are promising targets for adoptive immunotherapy using NK cells and cytotoxic T-cells, such as NKG2D-based CARs (12). In addition, NKG2D-expressing CAR-NK cells exhibit significantly increased cytotoxicity, suggesting their clinical therapeutic potential (13,14). In a phase I clinical trial, the safety of NKG2D-expressing CAR-T cells has also been demonstrated (15).
Soluble MIC leads to endocytosis and degradation of NKG2D
Tumors present multiple strategies to subvert the biological function of NKG2D and evade the immune system. The first is the proteolytic shedding of the ligand [Figure 1 shows protein (16-18) functioning on this site], which results in the downregulation or degradation of NKG2D (Figure 2). After proteolytic shedding, MICA is released into the microenvironment. The released soluble MICA (sMICA) binds to NKG2D, leading to its endocytosis and degradation (19). Some tumor cells release large amounts of sMICA, which impairs NK cell functions and leads to tumor progression (20).
Figure 1.
Mechanism of immune evasion through NKG2D-NKG2DL axis and therapeutic strategies. Mechanism of immune evasion (left panel): Proteolytic release of MICA/B of tumor cells leads to the downregulation or degradation of NKG2D. Its release is regulated by MMPs, members of the ADAM family, and ERp5, and the site of proteolytic shedding is located in the extracellular α3-domain of MICA. sMICs present in tumor-secreted exosomes might also lead to the downregulation and degradation of the ligand. TGF-β in the TME inhibits NK cell functions by downregulating NKG2D expression. MICA/MICB polymorphisms can also impair the recognition and binding of NKG2D. Therapeutic strategy (right panel): 1) Specific antibodies targeting the MICA α3 domain can decrease the loss of MICA/B on the surface of tumor cells; 2) Antibodies induced by a new tumor vaccine targeting the MICA/B protein can increase the numbers of MICA/B by inhibiting protein hydrolysis and shedding; 3) A CAR subpopulation, which contains the transmembrane domain of NKG2D, can increase the expression of NKG2D and mediate strong antigen-specific NK cell signaling; and 4) The soluble ligands can be removed from the plasma of patients to recover NK cell functions. MICA/B, major histocompatibility complex class I (MHC I) polypeptide-related sequence A and B; sMIC, soluble MIC; TGF, transforming growth factor; TME, tumor microenvironment; NK, natural killer.
Figure 2.
Primary mechanism of immune evasion of tumors based on NK cells. (A) NKG2D is an activating receptor of NK cells. Proteolytic shedding of MICA/B from the tumor cell surface leads to the downregulation or degradation of NKG2D. TGF-β in the TME also inhibits NK cell functions by downregulating NKG2D expression; (B) TGF-β in the TME inhibits NK cell functions by upregulating the expression of NKG2A. Tumor cells, cDCs, and tumor macrophages send a negative signal to NK cells by overexpressing HLA-E; (C) NK cells obtain PD-1 from tumor cells by trogocytosis. Glucocorticoids together with IL-12, IL-15, and IL-18 upregulate the transcription of PDCD1 in NK cells, thereby increasing PD-1 expression. PD-L1 expression is upregulated in response to multiple inflammatory stimuli in the TME, of which interferon-γ is the most potent. IFN-γ upregulates the expression of the ligand on tumor cells and increases the expression of the ligand on myeloid cells, thereby transmitting inhibitory signals to NK cells; (D) TIGIT ligand CD155 is highly expressed in tumor cells and inhibits NK cell functions by binding to TIGIT. MDSCs in the TME produce ROS, which upregulate the expression of CD155 on MDSCs. Interaction of the faap2 protein on the surface of Fusobacterium nucleatum with TIGIT might also inhibit the cytotoxicity of NK cells. NK, natural killer; MICA/B, major histocompatibility complex class I (MHC I) polypeptide-related sequence A and B; TGF, transforming growth factor; TME, tumor microenvironment; cDC, conventional dendritic cell; HLA-E, histocompatibility leukocyte antigen E; PD-1, programmed cell death 1; IL, interleukin; IFN, interferon; TIGIT, T-cell immunoglobulin and immunoreceptor tyrosine inhibitory motif; MDSC, myeloid-derived suppressor cell.
sMIC negatively reprograms the expression of genes relevant to the homeostatic survival and proliferation of NK cells (21). Ligands present in tumor-secreted exosomes (22) also downregulate NKG2D expression. The accumulation of vesicle-derived NKG2D ligands in the tumor microenvironment (TME) suppresses the activity of NK cells either by downregulating NKG2D expression or inducing the fratricide of NK cells dressed with EV-derived NKG2D ligands (Figure 1) (23). These processes impede tumor recognition and cytotoxicity of a large number of NK cells, leading to immune evasion. Moreover, some tumor stem cells are unable to express the NKG2D ligand (24). In acute myeloid leukemia (AML), the stemness of chemotherapy-resistant leukemic stem cells (LSCs) was believed to be related to immune evasion mediated by the NKG2D ligand because the absence of the NKG2D ligand defines LSCs, further indicating the importance of immune evasion mediated by the NKG2D ligand (25).
Downregulated expression of NKG2D
Downregulation of NKG2D receptor expression is a major mechanism of immune evasion by tumor cells. NKG2D expression decreases in NK cells in the TME due to several factors (26,27), such as transforming growth factor (TGF)-β (28,29) (Figure 2). Platelet occlusion also causes NKG2D ligand shedding (30). Mechanistically, platelet-derived TGF-β may be the main factor inhibiting the cytotoxicity of NK cells (31). IDO1 was also suggested to cause dysregulate NK cell activity via downregulating NKG2D expression in NK cells (32).
Polymorphisms of MICA/B
The effects of MICA/MICB ligand polymorphisms have been studied (33), and approximately 100 and 40 alleles have been reported for these ligands, respectively. MICA/B polymorphisms can also impair the recognition and binding of NKG2D, thereby reducing the cytotoxicity and recognition ability of NKG2D-expressing NK cells against tumor cells and causing NKG2D-based immune evasion.
High expression of NKG2A and HLA-E
NK cells express inhibitory receptors, including CD94/NKG2A heterodimer that recognize MHC class I molecules. These inhibitory receptors belong to the C-type lectin family (34). The cytoplasmic tail of NKG2A contains sequences of the immunoreceptor tyrosine-based inhibition motif (ITIM), which forms a heterodimer with the CD94 chain and identifies HLA-E as its ligand. After binding to the NKG2A/CD94 heterodimer, the function of T and NK cells is inhibited by recruiting SHP-1 tyrosine phosphatase to the ITIM of NKG2A (35). This axis is characterized by limited polymorphisms, and HLA-E has been proven to be highly expressed in a wide array of tumors (36,37), indicating that this axis may play an immunosuppressive role in the TME (36). Exploring the immunoregulatory mechanism of the NKG2A-HLA-E axis can help in developing improved and more efficient tumor targeting drug combinations.
Cytokines such as TGF-β can upregulate the expression of CD94/NKG2A in the TME (Figure 2) (38). Tumor-derived interferon (IFN)-γ inhibits the activity of cytotoxic cells via NKG2A/CD94 induced by ICB (39). Moreover, the expression of NKG2A in NK cells was dominant in the TME, while its expression was higher in breast cancer-infiltrating NK cells than in circulating NK cells (26). Although the mechanism is still unknown, this finding suggests that NKG2A contributes to immune evasion not only in tumor tissues but also in lymph nodes. Thus, even though the potential of NKG2A as a therapeutic target for immune evasion has been explored, its contribution to tumor metastasis remains to be elucidated.
Several tumor cells highly express HLA-E to escape the cytotoxicity of NK cells (36,37). The accumulation of HLA-E in cancer tissues originates from epithelial-derived cancer cells, CD141+ conventional dendritic cells (cDCs), and tumor macrophages (Figure 2) (40). Further studies are warranted to shed light on this mechanism of immunosuppression.
Mechanism based on PD-1/programmed cell death 1 ligand (PD-L1) axis
PD-1 is encoded by PDCD1, which is a member of the immunoglobulin gene superfamily. PD-1 is an inhibitory receptor primarily expressed on T-cells and plays a vital physiological role in the maintenance of peripheral tolerance, and it is also expressed in NK cells (41). Although CD8+ T-cells may have difficulty detecting some common tumors with low neoantigen load and HLA loss (42,43), PD-1/PD-L1 blocking immunotherapy was still effective in such patients (44), and NK cells played a vital role. These findings demonstrate the notable contribution of NK cells to PD-1/PD-L1 blocking immunotherapy (44), thus highlighting the importance of PD-1 in NK cells as an inhibitory checkpoint.
Induced expression of PD-1 on NK cells
The expression of PD-1 in NK cells is upregulated in many patients with cancer (45,46); however, the effect of this upregulated expression remains controversial (47). Most studies exploring whether or not PD-1 is expressed by NK cells still focus on the phenotype. However, many patients develop resistance to immunotherapy. Therefore, confirming the molecular mechanisms that control the expression of PD-1 and PD-L1 is important, so that strategies can be developed to enhance the efficacy of blockers of the PD-1/PD-L1 axis. Here we summarize recent studies investigating these mechanisms.
Studies on viruses have shown that endogenous glucocorticoids can induce the selective and tissue-specific expression of the checkpoint receptor PD-1 in NK cells (48). Mechanistically, glucocorticoids, together with interleukin (IL)-12, IL-15, and IL-18, not only upregulate PDCD1 transcription but also promote a transcriptional program that leads to increased protein translation and enhances the surface expression of the protein (Figure 2) (49). Moreover, this regulatory mechanism of human NK cells is not transferable to mouse NK cells, indicating that species specificity should be considered in studies exploring the mechanisms underlying NK cells.
In a study of leukemia, NK cells were shown to acquire PD-1 entirely from leukemia cells via trogocytosis in a SLAM receptor-dependent manner (Figure 2) (50). This should also be considered in the development of future therapies.
Highly expressed PD-L1 in TME
The PD-L1 protein is highly expressed in the TME. Notably, tumor cells activate local immune tolerance by overexpressing PD-L1 to keep NK cells and T-cells under control. PD-L1 is expressed in various kinds of cancer and myeloid cells in the TME, and its expression is upregulated in response to various inflammatory stimuli (51-53), of which IFN-γ is the most potent (Figure 2) (54). These inflammatory mediators induce PD-L1 and PD-L2 expression not only in cancer cells but also in other cell types in the TME (including macrophages, dendritic, and stromal cells) (51,52). The expression of PD-L1 in antigen-presenting cells (APCs) (but not tumor cells) has been suggested to play an essential role in checkpoint blockade therapy (53). These results provided insights into the mechanistic details of this treatment (53). In another study, a previously unknown strategy mediated via a shift toward an exhausted PD-1-enriched CD3-CD56hiCD16-ve NK cell phenotype facilitated immune evasion (55). Through this mechanism, NK cells are indirectly suppressed by PD-L1/PD-L2-expressing tumor-associated macrophages (TAMs) as well as directly inhibited by malignant B cells (55).
Upregulated expression of TIGIT and CD155
TIGIT, also known as WUCAM, Vstm3, and VSIG9, is a receptor of the Ig superfamily that is expressed mainly in NK cells and T-cells. Researchers have revealed that the CD155/TIGIT axis is necessary and sufficient to promote and maintain immune evasion in pancreatic adenocarcinoma (PDAC) (56). Recent studies emphasized the specificity and importance of TIGIT in NK cells, including high expression of TIGIT in NK cells present in the TME (57) and its specificity in NK cell exhaustion, but not that of the other checkpoint molecules CTLA-4 and PD-1 (57). Anti-TIGIT monotherapy or in combination with PD-1 blockade was shown to mainly affect NK cells, not only by promoting NK cell-dependent tumor immunity but also by enhancing NK cell-dependent tumor-specific T-cell immunity (57). These findings demonstrated the role of TIGIT in the NK cell-based mechanism of immune evasion of tumor cells.
The expression of TIGIT on mouse NK cells is upregulated during tumor progression. In patients with colorectal tumors, the constitutive expression of TIGIT in NK cells is reportedly further upregulated in the tumor compared with that in the peritumoral area (57). Likewise, the expression of TIGIT in NK cells from ascites of patients with ovarian cancer was higher than that in NK cells from healthy donors (58). TIGIT/CD155 binding suppresses IFN-γ production via the NF-κB pathway (59). Specifically, a novel adaptor β-arrestin 2, which associates with phosphorylated TIGIT to recruit SH2-containing inositol phosphatase 1 (SHIP1) via the ITT-like motif, impairs the autoubiquitination of TNF receptor-associated factor 6 (TRAF6), thus abrogating the activation of NF-κB, and in turn leading to suppressed production of IFN-γ in NK cells (59).
CD155 is highly expressed in many tumor tissues (60,61), although its mechanism of function in tumor immunity is relatively complex. CD155 can inhibit NK cell functions via PI3K and MAPK signaling after binding to TIGIT (62). Its intrinsic roles in tumor cells include promotion of tumor migration (63), shortening of the cell cycle of tumor cells, and promotion of tumor cell proliferation (64). In addition, CD155 affects the cytotoxicity of NK cells by downregulating the expression of CD226 on their surface (65).
Bacteria present in the TME may also inhibit NK cell functions through TIGIT. Mechanistically, TIGIT binds directly to the fap2 protein of Fusobacterium nucleatum (a bacterium commonly found in the TME), inhibiting the cytotoxicity of NK cells (Figure 2) (66). Similarly, myeloid-derived suppressor cells (MDSCs) in the TME inhibit NK cell functions through TIGIT. Interestingly, TIGIT signaling in NK cells lead to a decrease in the phosphorylation of ZAP70/Syk and ERK1/2 after coculture with MDSCs, which in turn produced reactive oxygen species (ROS) that upregulated the expression of TIGIT ligand CD155 on their surface (Figure 2) (67).
Therapeutic status and opportunities based on above mechanism
Therapeutic status and opportunities related to NKG2D
Antibody-mediated inhibition of MICA/B shedding
Numerous studies have explored methods of improving the cytotoxicity of NK cells by targeting NKG2D-NKG2DL axis-mediated immune evasion mechanisms. The clinical application of α3 domain-targeted MICA/B antibodies may offer a new strategy for cancer immunotherapy (Figure 1) (68). Antibodies targeting tumor-derived sMIC not only reprogrammed the homeostatic survival and function of NK cells but also enhanced the response to PD-L1 blockade therapy in melanoma (21). These findings highlight the importance of antibodies targeting sMIC to improve the effect of anti-PD1/PD-L1 antibody-based treatment in patients with MIC/sMIC+ metastatic melanoma (21).
Anti-MICA/B and tumor vaccine
Anti-MICA/B antibodies have been used to enhance the response of immunotherapy applied to human colorectal tumor spheroids by increasing the activation and infiltration of NK cells (69). Interestingly, tumor cells responded to immunotherapy by upregulating the expression of HLA-E, which is the ligand of NKG2A expressed by NK and CD8+ T-cells (69). This demonstrated the antitumor potential of combination immunotherapy targeting MICA/B and NKG2A (69); however, the specific mechanism remains unclear.
In a preclinical study of osteosarcoma, acetylation of the NKG2D ligand protected RAE-1 from shedding, thereby revealing potential new targets for immunotherapy based on NKG2D (70). Recently, a new tumor vaccine targeting the MICA/B protein was reported. This vaccine induced the generation of antibodies that increased the levels of MICA/B by restraining protein hydrolysis and shedding, thereby facilitating a stronger cytotoxic function of NK cells (Figure 1) (71). Moreover, the design of this vaccine enabled protective immunity even target tumors with common escape mutations (71). Researchers have also attempted to remove these soluble ligands from the plasma of patients to recover NK cell functions (Figure 1). They proposed the adsorption apheresis of these soluble ligands as a future preprocessing method to increase the efficacy of autologous and adoptively transferred immune cells in cancer immunotherapy (72). The mechanism underlying therapeutic potential of chemotherapy-resistant LSCs has also been investigated, and poly-ADP-ribose polymerase 1 (PARP1) was found to repress NKG2D ligand expression. The genetic or pharmacological inhibition of PARP1 can thus induce NKG2DL expression on the LSC surface, thereby providing strong evidence for combining PARP1 inhibition and NK cell functions (25).
NKG2D-CAR-NK cells
Researchers have identified a CAR cell subpopulation that contains the transmembrane domain of NKG2D and can mediate potent antigen-specific NK cell signaling (Figure 1) (73). It remarkably prolonged survival and reduced tumor burden in xenograft models of ovarian cancer, showed similar activity in vivo to that of CAR-T cells, and was less toxic (73). Autologous NKG2D-CAR-NK cells showed enhanced antimyeloma activity in both in-vivo and in-vitro experiments (14), supporting the potential of NKG2D-CAR-NK cells in the treatment of multiplemyeloma (MM). By fusing the extracellular domain of NKG2D to DAP12, a NKG2D RNA CAR was engineered to improve antitumor response of NK cells. Its expression remarkably enhanced the cytotoxicity of NK cells to several kinds of tumor cell lines in preclinical models (13) and led to decrease in ascites and tumor cells in patients with metastatic colorectal cancer (13). The combination of immune metabolism and NKG2D-CAR-NK cells can thus result in remarkable antitumor responses in vivo within the hypoxic TME of solid tumors (74).
However, the therapeutic effect of NKG2D-CAR-NK cells might be limited if the immune evasion mechanism associated with the ligand is not resolved. Therefore, researchers developed a homologous recombinant antibody called NKAB-ErbB2, which enables tumor-specific NKG2D-expressing effector cells to be independent of membrane-anchored NKG2D ligands (75). This molecule carries an NKG2D-based CAR, which enables its relocalization to ERBB2+ tumor cells and increases tumor antigen specificity and NKG2D-CAR and NKG2D-mediated therapeutic efficacy (75). This was shown to be an alternative method of exploiting the molecular properties of NKG2D to avoid immune evasion. Therefore, NKG2D-CAR-NK therapy that simultaneously targets sMICA might be a new strategy to enhance NK cell functions.
NKG2A-related monotherapy and combination therapy
Anti-NKG2A monotherapy
Monalizumab has high affinity for CD94/NKG2A and blocks inhibitory HLA-E signaling (76). Preclinical studies have suggested that blocking NKG2A on NK cells in chronic lymphocytic leukemia (CLL) is sufficient to directly restore the cytotoxicity of NK cells against HLA-E+ targets, without affecting NK cell-mediated antibody-dependent cellular cytotoxicity (ADCC) (37). The low efficacy of monotherapy might be attributed to the fact that only 50% of NK cells in peripheral blood express NKG2A, which may limit the potential of NKG2A therapy (77,78). Nevertheless, the importance of anti-NKG2A antibodies in combination therapy has been well recognized by researchers.
Importance of anti-NKG2A in combination therapy
The expression of NKG2A and PD-1 in NK cells in tumor-draining lymph nodes (TD-LNs) supports their potential for antitumor immunotherapies using anti-NKG2A, anti-PD-1, or both antibodies, and it also suggests that anti-NKG2A and anti-PD-1 combination therapy might be a preferred adjuvant therapy for breast cancer (79). Monalizumab in combination with PD-1/PD-L1 axis blockade enhanced NK cell functions against a variety of tumor cells, improved survival, and rescued the cytotoxic T-cell-dependent response (76). In preclinical mouse tumor models, the combination of NKG2A and PD-L1 blocking therapy (monalizumab and durvalumab) has shown better therapeutic efficacy and synergistic effects (76). In a phase II clinical trial called COAST, the therapeutic efficacy of durvalumab alone or in combination with monalizumab or oleclumab (the anti-CD73 antibody) as consolidation therapy was assessed in patients with unresectable stage III non-small cell lung cancer (80). The confirmed objective response rate (ORR) was reported to be higher and progression-free survival (PFS) was prolonged in patients treated with durvalumab combined with monalizumab as compared to those treated with durvalumab. In addition, adverse events of grades ≥3 occurred at the lowest frequency (27.9%) in patients treated with durvalumab combined with monalizumab but at a higher frequency (39.4%) in patients treated with durvalumab alone (80). This highlights the role of anti-NKG2A therapy, demonstrates the potential of combination therapy with anti-NKG2A and anti-PD-1, and raises expectations for future phase III clinical trials. Table 1 summarizes ongoing clinical trials of NKG2A monotherapy and combination therapy.
Table 1. Ongoing clinical trials involving anti-NKG2A alone or in combination with other therapies.
Trial identifier | Phase | Participants | Drugs | Condition or diseases | Status |
PD-(L)1, programmed cell death 1/programmed cell death 1 ligand. | |||||
NCT04307329 | Phase 2 | 38 | Monalizumab, trastuzumab | Breast cancer | Active, not recruiting |
NCT04590963 | Phase 3 | 369 | Monalizumab, cetuximab | Squamous cell carcinoma of the head and neck | Active, not recruiting |
NCT05221840 | Phase 3 | 999 | Durvalumab with monalizumab or oleclumab | Non-small cell lung cancer | Recruiting |
NCT05414032 | Phase 2 | 200 | Monalizumab, cetuximab | Locoregionally advanced head and neck squamous cell carcinoma | Not yet recruiting |
NCT02643550 | Phase 1, Phase 2 |
143 | Monalizumab and cetuximab with or without anti-PD-(L)1 | Head and neck neoplasms | Active, not recruiting |
NCT03822351 | Phase 2 | 188 | Durvalumab alone or with monalizumab or oleclumab | Stage III non-small cell lung cancer, unresectable | Active, not recruiting |
NCT05061550 | Phase 2 | 210 | Durvalumab with oleclumab or monalizumab | Non-small cell lung cancer | Recruiting |
NCT03088059 | Phase 2 | 340 | Monalizumab alone or with durvalumab and other interventions | Carcinoma, squamous cell of head and neck | Recruiting |
NCT03833440 | Phase 2 | 120 | Durvalumab with monalizumab or oleclumab or other interventions | Non-small cell lung cancer | Recruiting |
Loss of the function of Qa-1b (HLA-E in humans) in mice improved the efficacy of PD-1 immunotherapy (81) in preclinical studies. In addition, disruption of the NKG2A/Qa-1b axis significantly enhanced the effect of therapeutic vaccines, even in PD-1 refractory mouse models (82). Of note, in bladder tumors with limited PD-1 efficacy, NKG2A was associated with improved survival and PD-L1 blocking of immunotherapy reactivity (83). These data support the strategy of combining monoclonal antibodies that block PD-1/PD-L1 with NKG2A/HLA-E in clinical applications.
Preliminary results in patients with squamous cell carcinoma of the head and neck (SCCHN) treated with monalizumab in combination with cetuximab (an anti-EGFR antibody) revealed improved overall response rates, even in refractory patients. Importantly, blockade of NKG2A showed little toxicity and no evidence of autoimmunity (76). In-vitro experiments revealed that monalizumab promoted NK cell-mediated ADCC against SCCHN tumor cells coated with cetuximab. This action was likely mediated by NK cells expressing NKG2A rather than activated cytotoxic T-cells (76). These clinical data suggested that NKG2A is a promising checkpoint inhibitor for combination therapy.
Highly functional NK and ML NK cells
To bypass HLA-E-mediated inhibition, researchers have attempted to develop a method to generate highly functional NK cells that lack NKG2A expression on the surface (84). These NKG2A protein expression blockers (PEBLs) were generated by linking a single-chain variable fragment derived from an anti-NKG2A antibody to endoplasmic reticulum retention domains. Interestingly, PEBLs eliminated the expression of NKG2A, bypassed HLA-E cancer immunity checkpoints, and enhanced the function of NK cell infusions (84).
A group studied the phenotypic characteristics of cytokine-induced memory-like (ML) NK cells and demonstrated that NKG2A is a dominant checkpoint (85). The potent cytotoxic therapeutic effect of ML NK cells relies on the activating receptors NKG2D and NKp46 (86). When anti-MICA/B was used to treat colon tumors, tumor cells responded to the increased cytotoxicity by upregulating the expression of HLA-E (69). Therefore, combined therapy based on targeting the mechanisms of immune evasion through NKG2A and NKG2D may overcome the obstacles of NK cell therapy in the future.
Targeting of PD-1/PD-L1 pathways for improving NK cell functions
CAR-NK cells targeting PD-L1 have an enhanced effect against solid tumors in vitro and in vivo. Combined treatment with CAR-NK cells and nivolumab resulted in a synergistic response against solid tumors (87). Fabian et al. developed a new NK cell line, PD-L1-targeting high-affinity natural killers (t-haNKs), from NK-92 cells that express high-affinity CD16, endoplasmic reticulum-retained interleukin-2, and a PD-L1 specific CAR (88). They demonstrated that PD-L1 t-haNK cells had good antitumor activity and preferentially lysed the myeloid-derived suppressor cell population but not other immune cell types when cocultured with human peripheral blood mononuclear cells (PBMCs) (88). In another study, NK cells were chemically equipped with a TLS11a aptamer targeting HepG2 cells and PD-L1-specific aptamer without genetic alteration. This improved the specificity of NK cells, remarkably upregulated the expression of PD-L1 in HepG2 cells, and enhanced checkpoint blockade. This holds significant potential for improved adoptive immunotherapy against solid tumors (89).
In some tumors deficient in the expression of MHC class I proteins, the efficacy of PD-1 and PD-L1 blockers depends on the activity of NK cells (44). In the model of PD-1-resistant B16-F10 lung metastasis, anti-PD-1 monotherapy failed to achieve an antitumor effect, whereas a lipid nanoparticle containing a stimulator of an interferon gene (STING) and an agonist (STING-LNP) achieved synergistic antitumor effects when combined with anti-PD-1 (90). Mechanistically, NK cells are activated by stimulating the STING pathway to produce IFN-γ, leading to an increase in the expression of PD-L1 on tumor cells, resulting in a synergistic antitumor effect when anti-PD-1 was administered (90). NKTR214 (bempegaldesin) is a novel agonist of the IL2 pathway that provides sustained signaling via heterodimeric IL2 receptor βγ to drive the increased proliferation and activation of CD8+ T-cells and NK cells (91). A phase I trial of the combination of NKTR214 and nivolumab (anti-PD-1 mAb) was conducted in patients with advanced solid tumors (NCT02983045) (92), in which the combination was shown to be safe, with an ORR of 59.5% (92). Based on the assumption of activating NK cells while targeting PD-1/PD-L1, researchers also developed N-809, which is fused with the IL-15 superagonist N-803 in 2 αPD-L1 domains and stimulates the expression of IL-15 and blocks PD-L1 (93). N-809 had a stronger antitumor effect and immunomodulatory effects in the TME compared with the N-803+αPD-L1 combination (94).
Preclinical studies have suggested that blocking PD-L1 enhances the cytotoxicity of NK cells against tumor cells (95). Mechanistically, NK cells secrete a large amount of IFN-γ, which stimulates further expression of PD -L1 on tumor cells (95); conversely, anti-PD-L1 mAbs act directly on PD-L1+ NK cells against PD-L1- tumors via the p38 signaling pathway (96). The endogenous glucocorticoids can induce the selective and tissue-specific expression of the checkpoint receptor PD-1 in NK cells (48). In a study of leukemia, NK cells were shown to acquire PD-1 entirely from leukemia cells via trogocytosis in a SLAM receptor-dependent manner (50). These studies support the combination of NK cells and PD-1 axis therapy.
The combination therapy of anti-PD-1/PD-L1 and NK cells is considered to have good clinical prospect. In non-small cell lung cancer (NSCLC) clinical trials, the anti-PD-1 antibody pembrolizumab in combination with allogeneic NK cells resulted in a higher ORR and significant improvement in survival in patients with advanced NSCLC and treated with PD-L1+ (97). In addition, the combination of highly activated allogeneic NK cells (“SMT-NK”) with pembrolizumab did not result in any serious adverse events directly related to the drug combination in patients with advanced biliary tract cancer (98). This combination therapy also exerted enhanced efficacy compared with monotherapy with pembrolizumab (98). Table 2 summarizes ongoing clinical trials of PD-1/PD-L1 combination therapy with NK-based treatment.
Table 2. Ongoing clinical trials involving anti-PD-1/PD-L1 in combination with NK cell-based therapy.
Trial identifier | Phase | Participants | Intervention/treatment | Condition or diseases | Status |
PD-1, programmed cell death 1; PD-L1, programmed cell death 1 ligand; IL, interleukin; DC, dendritic cell; NK, natural killer; CAR, chimeric antigen receptor. | |||||
NCT05334329 | Phase 1 | 21 | Umbilical cord blood natural killer cells (CB-NK) expressing soluble IL-15 (sIL-15) and PD-L1 +/− atezolizumab | Advanced/metastatic/recurrent/refractory non-small cell lung carcinoma | Recruiting |
NCT05461235 | Phase 2 | / | Anti-PD-1 antibody combined with autologous DC or NK cells | Digestive carcinoma | Not yet recruiting |
NCT03383978 | Phase 1 | 42 | Intracranial injection of NK-92/5.28.z cells in combination with the anti-PD-1 antibody ezabenlimab (BI 754091) | Glioblastoma | Recruiting |
NCT04847466 | Phase 2 | 55 | Irradiated PD-L1 CAR-NK cells plus pembrolizumab plus N-803 | Recurrent/metastatic gastric or head and neck cancer | Recruiting |
NCT04050709 | Phase 1 | 16 | PD-L1 t-haNK | Locally advanced or metastatic solid cancers | Active, not recruiting |
NCT04927884 | Phase 1, Phase 2 |
79 | Sacituzumab plus cyclophosphamide, N-803, and PD-L1 t-haNK | Advanced triple negative breast cancer | Active, not recruiting |
NCT04390399 | Phase 2 | 328 | Standard-of-care chemotherapy versus standard-of-care chemotherapy in combination with aldoxorubicin HCl, N-803, and PD-L1 t-haNK | Pancreatic cancer | Recruiting |
NCT03228667 | Phase 2 | 145 | PD-1/PD-L1 checkpoint inhibitor, N-803, and PD-L1 t-haNK cellular therapy | Patients who have previously received treatment with PD-1/PD-L1 immune checkpoint inhibitors | Active, not recruiting |
TIGIT in NK-cell-based therapy
TIGIT is considered a previously neglected checkpoint in NK cells, and studies have recently highlighted its crucial role in NK cells. For instance, blockade of TIGIT reversed the exhaustion of tumor-infiltrated NK cells and promoted NK cell-dependent tumor immunity (57). Blockade of TIGIT led to NK cell-dependent tumor-specific T-cell immunity and enhanced the efficacy of anti-PD-L1 therapy, which in combination with other checkpoint receptors is a promising therapeutic strategy against cancer (57). Administration of IL-15 in combination with TIGIT blockers increased NK cell-mediated melanoma cytotoxicity in vitro and reduced tumor metastasis in mouse melanoma models. These results support the novel combinatorial immunotherapy with IL15 and TIGIT blockade for promoting NK cell-mediated destruction of MHC class I-deficient melanoma that was refractory to CD8+ T-cell-mediated immunity (99). Current research on the role of TIGIT in NK cells is still in the preclinical stage. However, based on the reported findings of the effects of TIGIT on NK cells, clinical trials on combined treatment with NK cells are expected in the future.
TIGIT is highly expressed in tumor-infiltrating T-cells of humans and mice; hence, most studies were initially focused on the function of TIGIT in T-cells. Combined blockade of TIGIT and administration of PD-L1 antibodies synergistically and specifically improved the effector function of CD8+ T-cells (100). Further, the expression of TIGIT was upregulated in T-cells and in dysfunctional PD-1+TIM-3+ tumor antigen-specific (TA-specific) CD8+ T-cells upon PD-1 blockade (101).
Collectively, these findings suggest that TIGIT might play a complementary role in PD-1/PD-L1-based therapies and support the possibility of combination therapy of anti-PD-1 and anti-TIGIT. In a phase I clinical trial, the anti-TIGIT (T-cell immunoglobulin and ITIM domain) antibody vibostolimab in combination with pembrolizumab was well tolerated and demonstrated antitumor activity in patients with advanced solid tumors, including advanced NSCLC (102). Phase II studies of anti-TIGIT in combination with anti-PD-L1 have also shown a clinically significant improvement in ORR and PFS compared with a placebo plus atezolizumab in patients with NSCLC (103). In addition, a combination of the two drugs was reported to be well tolerated, and its safety profile was similar to that of atezolizumab alone (103). These results suggest the importance of TIGIT in future immunotherapy.
Conclusions and perspectives
NK cells belong to a family of innate immune cells, and they kill virus-infected cells and malignantly transformed cells without recognizing specific antigens, thereby exhibiting immune surveillance activity and cytotoxic functions in tumor immunotherapy (5). Moreover, NK cells exhibit effective cytotoxicity, do not require the involvement of specific antigens, do not rely on MHC molecules to perform allogeneic adoptive transfer, do not cause cytokine release syndrome, and do not cause graft-versus-host disease (6). Because of these properties, NK cells are attractive targets for tumor immunotherapy and have received increasing attention from clinicians and researchers. The effector function of NK cells is based on the integration of activating and inhibitory signals. Here, we elaborated on the mechanism of immune evasion associated with the activating receptor NKG2D and the current status of related treatments, and proposed that activation of NKG2D and simultaneous inhibition of MICA shedding might enhance the activity of NK cells. The inhibitory molecule NKG2A on the surface of NK cells exerts low toxic effects when used in combination therapy, making it a better partner for future combination therapies. Although anti-PD-1/PD-L1 therapies have played a prominent role in the immunotherapy phase, they provide benefits to only a small number of patients. When combined with NK cells with enhanced functions, such as increased activity or specificity, these combination therapies might not only overcome immune evasion but also achieve a significant therapeutic outcome through NK cell-mediated ADCC. Future studies should aim at sustaining and enhancing the immune infiltration of NK cells. The combination of these functions of NK cells with PD-1/PD-L1 targets is expected to result in optimal therapeutic effects. The function and therapeutic benefits of TIGIT in NK cells require data from preclinical studies and clinical trials. Future studies should focus on the combination of anti-TIGIT and anti-PD-1 treatments as well as treatment combinations that enhance NK cell functions, such as CAR-NK cells.
Acknowledgements
None.
Contributor Information
Songling Zhang, Email: slzhang@jlu.edu.cn.
Yufeng Wang, Email: Yufeng_Wang@jlu.edu.cn.
References
- 1.Jhunjhunwala S, Hammer C, Delamarre L Antigen presentation in cancer: insights into tumour immunogenicity and immune evasion. Nat Rev Cancer. 2021;21:298–312. doi: 10.1038/s41568-021-00339-z. [DOI] [PubMed] [Google Scholar]
- 2.Wu SY, Fu T, Jiang YZ, et al Natural killer cells in cancer biology and therapy. Mol Cancer. 2020;19:120. doi: 10.1186/s12943-020-01238-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Neelapu SS, Tummala S, Kebriaei P, et al Toxicity management after chimeric antigen receptor T cell therapy: one size does not fit “ALL”. Nat Rev Clin Oncol. 2018;15:218. doi: 10.1038/nrclinonc.2018.20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Ranti D, Bieber C, Wang YS, et al Natural killer cells: unlocking new treatments for bladder cancer. Trends Cancer. 2022;8:698–710. doi: 10.1016/j.trecan.2022.03.007. [DOI] [PubMed] [Google Scholar]
- 5.Morvan M, Lanier L NK cells and cancer: you can teach innate cells new tricks. Nat Rev Cancer. 2016;16:7–19. doi: 10.1038/nrc.2015.5. [DOI] [PubMed] [Google Scholar]
- 6.Liu E, Marin D, Banerjee P, et al. Use of CAR-transduced natural killer cells in CD19-positive lymphoid tumors. N Engl J Med 2020;382:545-53.
- 7.Fuertes MB, Domaica CI, Zwirner NW Leveraging NKG2D ligands in immuno-oncology. Front Immunol. 2021;12:713158. doi: 10.3389/fimmu.2021.713158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Sivori S, Vacca P, Del Zotto G. Human NK cells: surface receptors, inhibitory checkpoints, and translational applications. Cell Mol Immunol 2019;16:430-41.
- 9.Upshaw JL, Arneson LN, Schoon RA, et al NKG2D-mediated signaling requires a DAP10-bound Grb2-Vav1 intermediate and phosphatidylinositol-3-kinase in human natural killer cells. Nat Immunol. 2006;7:524–32. doi: 10.1038/ni1325. [DOI] [PubMed] [Google Scholar]
- 10.Duan S, Guo W, Xu Z, et al Natural killer group 2D receptor and its ligands in cancer immune escape. Mol Cancer. 2019;18:29. doi: 10.1186/s12943-019-0956-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Lanier LL NKG2D receptor and its ligands in host defense. Cancer Immunol Res. 2015;3:575–82. doi: 10.1158/2326-6066.cir-15-0098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Chang YH, Connolly J, Shimasaki N, et al A chimeric receptor with NKG2D specificity enhances natural killer cell activation and killing of tumor cells. Cancer Res. 2013;73:1777–86. doi: 10.1158/0008-5472.can-12-3558. [DOI] [PubMed] [Google Scholar]
- 13.Xiao L, Cen D, Gan H, et al Adoptive transfer of NKG2D CAR mRNA-engineered natural killer cells in colorectal cancer patients. Mol Ther. 2019;27:1114–25. doi: 10.1016/j.ymthe.2019.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Leivas A, Valeri A, Córdoba L, et al NKG2D-CAR-transduced natural killer cells efficiently target multiple myeloma. Blood Cancer J. 2021;11:146. doi: 10.1038/s41408-021-00537-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Baumeister SH, Murad J, Werner L, et al Phase I trial of autologous CAR T cells targeting NKG2D ligands in patients with AML/MDS and multiple myeloma. Cancer Immunol Res. 2019;7:100–12. doi: 10.1158/2326-6066.cir-18-0307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Salih HR, Rammensee HG, Steinle A Cutting edge: down-regulation of MICA on human tumors by proteolytic shedding. J Immunol. 2002;169:4098–102. doi: 10.4049/jimmunol.169.8.4098. [DOI] [PubMed] [Google Scholar]
- 17.Waldhauer I, Goehlsdorf D, Gieseke F, et al Tumor-associated MICA is shed by ADAM proteases. Cancer Res. 2008;68:6368–76. doi: 10.1158/0008-5472.can-07-6768. [DOI] [PubMed] [Google Scholar]
- 18.Kaiser BK, Yim D, Chow IT, et al Disulphide-isomerase-enabled shedding of tumour-associated NKG2D ligands. Nature. 2007;447:482–6. doi: 10.1038/nature05768. [DOI] [PubMed] [Google Scholar]
- 19.Groh V, Wu J, Yee C, et al Tumour-derived soluble MIC ligands impair expression of NKG2D and T-cell activation. Nature. 2002;419:734–8. doi: 10.1038/nature01112. [DOI] [PubMed] [Google Scholar]
- 20.Jinushi M, Vanneman M, Munshi NC, et al MHC class I chain-related protein A antibodies and shedding are associated with the progression of multiple myeloma. Proc Natl Acad Sci U S A. 2008;105:1285–90. doi: 10.1073/pnas.0711293105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Basher F, Dhar P, Wang X, et al Antibody targeting tumor-derived soluble NKG2D ligand sMIC reprograms NK cell homeostatic survival and function and enhances melanoma response to PDL1 blockade therapy. J Hematol Oncol. 2020;13:74. doi: 10.1186/s13045-020-00896-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Hong CS, Funk S, Muller L, et al Isolation of biologically active and morphologically intact exosomes from plasma of patients with cancer. J Extracell Vesicles. 2016;5:29289. doi: 10.3402/jev.v5.29289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Vulpis E, Loconte L, Peri A, et al Impact on NK cell functions of acute versus chronic exposure to extracellular vesicle-associated MICA: Dual role in cancer immunosurveillance. J Extracell Vesicles. 2022;11:e12176. doi: 10.1002/jev2.12176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Di Tomaso T, Mazzoleni S, Wang E, et al Immunobiological characterization of cancer stem cells isolated from glioblastoma patients. Clin Cancer Res. 2010;16:800–13. doi: 10.1158/1078-0432.ccr-09-2730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Paczulla AM, Rothfelder K, Raffel S, et al Absence of NKG2D ligands defines leukaemia stem cells and mediates their immune evasion. Nature. 2019;572:254–9. doi: 10.1038/s41586-019-1410-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Mamessier E, Sylvain A, Thibult ML, et al Human breast cancer cells enhance self tolerance by promoting evasion from NK cell antitumor immunity. J Clin Invest. 2011;121:3609–22. doi: 10.1172/jci45816. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Platonova S, Cherfils-Vicini J, Damotte D, et al Profound coordinated alterations of intratumoral NK cell phenotype and function in lung carcinoma. Cancer Res. 2011;71:5412–22. doi: 10.1158/0008-5472.can-10-4179. [DOI] [PubMed] [Google Scholar]
- 28.Castriconi R, Cantoni C, Della Chiesa M, et al Transforming growth factor beta 1 inhibits expression of NKp30 and NKG2D receptors: consequences for the NK-mediated killing of dendritic cells. Proc Natl Acad Sci U S A. 2003;100:4120–5. doi: 10.1073/pnas.0730640100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Lee JC, Lee KM, Kim DW, et al Elevated TGF-beta1 secretion and down-modulation of NKG2D underlies impaired NK cytotoxicity in cancer patients. J Immunol. 2004;172:7335–40. doi: 10.4049/jimmunol.172.12.7335. [DOI] [PubMed] [Google Scholar]
- 30.Maurer S, Kropp KN, Klein G, et al Platelet-mediated shedding of NKG2D ligands impairs NK cell immune-surveillance of tumor cells. Oncoimmunology. 2018;7:e1364827. doi: 10.1080/2162402x.2017.1364827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Kopp HG, Placke T, Salih HR Platelet-derived transforming growth factor-beta down-regulates NKG2D thereby inhibiting natural killer cell antitumor reactivity. Cancer Res. 2009;69:7775–83. doi: 10.1158/0008-5472.can-09-2123. [DOI] [PubMed] [Google Scholar]
- 32.Fang X, Guo L, Xing Z, et al IDO1 can impair NK cells function against non-small cell lung cancer by downregulation of NKG2D Ligand via ADAM10. Pharmacol Res. 2022;177:106132. doi: 10.1016/j.phrs.2022.106132. [DOI] [PubMed] [Google Scholar]
- 33.Toledo-Stuardo K, Ribeiro CH, Canals A, et al Major histocompatibility complex class I-related Chain A (MICA) allelic variants associate with susceptibility and prognosis of gastric cancer. Front Immunol. 2021;12:645528. doi: 10.3389/fimmu.2021.645528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Cózar B, Greppi M, Carpentier S, et al. Tumor-infiltrating natural killer cells. Cancer Discov 2021;11:34-44.
- 35.Bi J, Tian Z NK cell dysfunction and checkpoint immunotherapy. Front Immunol. 2019;10:1999. doi: 10.3389/fimmu.2019.01999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Borst L, van der Burg SH, van Hall T The NKG2A-HLA-E axis as a novel checkpoint in the tumor microenvironment. Clin Cancer Res. 2020;26:5549–56. doi: 10.1158/1078-0432.ccr-19-2095. [DOI] [PubMed] [Google Scholar]
- 37.McWilliams EM, Mele JM, Cheney C, et al Therapeutic CD94/NKG2A blockade improves natural killer cell dysfunction in chronic lymphocytic leukemia. Oncoimmunology. 2016;5:e1226720. doi: 10.1080/2162402x.2016.1226720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Bertone S, Schiavetti F, Bellomo R, et al Transforming growth factor-beta-induced expression of CD94/NKG2A inhibitory receptors in human T lymphocytes. Eur J Immunol. 1999;29:23–9. doi: 10.1002/(sici)1521-4141(199901)29:01<23::aid-immu23>3.0.co;2-y. [DOI] [PubMed] [Google Scholar]
- 39.Dubrot J, Du PP, Lane-Reticker SK, et al. In vivo CRISPR screens reveal the landscape of immune evasion pathways across cancer. Nat Immunol 2022;23:1495-506.
- 40.Abd Hamid M, Wang RZ, Yao X, et al. Enriched HLA-E and CD94/NKG2A interaction limits antitumor CD8(+) tumor-infiltrating T lymphocyte responses. Cancer Immunol Res 2019;7:1293-306.
- 41.Pesce S, Greppi M, Tabellini G, et al Identification of a subset of human natural killer cells expressing high levels of programmed death 1: A phenotypic and functional characterization. J Allergy Clin Immunol. 2017;139:335–46.e3. doi: 10.1016/j.jaci.2016.04.025. [DOI] [PubMed] [Google Scholar]
- 42.McGranahan N, Rosenthal R, Hiley CT, et al Allele-specific HLA loss and immune escape in lung cancer evolution. Cell. 2017;171:1259–71.e11. doi: 10.1016/j.cell.2017.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Roemer MG, Advani RH, Redd RA, et al Classical Hodgkin lymphoma with reduced β2M/MHC class I expression is associated with inferior outcome independent of 9p24. 1 status. Cancer Immunol Res. 2016;4:910–6. doi: 10.1158/2326-6066.cir-16-0201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Hsu J, Hodgins JJ, Marathe M, et al Contribution of NK cells to immunotherapy mediated by PD-1/PD-L1 blockade. J Clin Invest. 2018;128:4654–68. doi: 10.1172/jci99317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Quatrini L, Mariotti FR, Munari E, et al. The immune checkpoint PD-1 in natural killer cells: expression, function and targeting in tumour immunotherapy. 2020;12:3285.
- 46.Khan M, Arooj S, Wang H NK cell-based immune checkpoint inhibition. Front Immunol. 2020;11:167. doi: 10.3389/fimmu.2020.00167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Judge SJ, Dunai C, Aguilar EG, et al Minimal PD-1 expression in mouse and human NK cells under diverse conditions. J Clin Invest. 2020;130:3051–68. doi: 10.1172/jci133353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Quatrini L, Wieduwild E, Escaliere B, et al Endogenous glucocorticoids control host resistance to viral infection through the tissue-specific regulation of PD-1 expression on NK cells. Nat Immunol. 2018;19:954–62. doi: 10.1038/s41590-018-0185-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Quatrini L, Vacca P, Tumino N, et al Glucocorticoids and the cytokines IL-12, IL-15, and IL-18 present in the tumor microenvironment induce PD-1 expression on human natural killer cells. J Allergy Clin Immunol. 2021;147:349–60. doi: 10.1016/j.jaci.2020.04.044. [DOI] [PubMed] [Google Scholar]
- 50.Hasim MS, Marotel M, Hodgins JJ, et al When killers become thieves: Trogocytosed PD-1 inhibits NK cells in cancer. Sci Adv. 2022;8:eabj3286. doi: 10.1126/sciadv.abj3286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Benci JL, Xu B, Qiu Y, et al Tumor interferon signaling regulates a multigenic resistance program to immune checkpoint blockade. Cell. 2016;167:1540–54.e12. doi: 10.1016/j.cell.2016.11.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Nguyen LT, Ohashi PS Clinical blockade of PD1 and LAG3-potential mechanisms of action. Nat Rev Immunol. 2015;15:45–56. doi: 10.1038/nri3790. [DOI] [PubMed] [Google Scholar]
- 53.Tang H, Liang Y, Anders RA, et al PD-L1 on host cells is essential for PD-L1 blockade-mediated tumor regression. J Clin Invest. 2018;128:580–8. doi: 10.1172/jci96061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Boussiotis VA Molecular and biochemical aspects of the PD-1 checkpoint pathway. N Engl J Med. 2016;375:1767–78. doi: 10.1056/NEJMra1514296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Vari F, Arpon D, Keane C, et al Immune evasion via PD-1/PD-L1 on NK cells and monocyte/macrophages is more prominent in Hodgkin lymphoma than DLBCL. Blood. 2018;131:1809–19. doi: 10.1182/blood-2017-07-796342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Freed-Pastor WA, Lambert LJ, Ely ZA, et al The CD155/TIGIT axis promotes and maintains immune evasion in neoantigen-expressing pancreatic cancer. Cancer Cell. 2021;39:1342–60.e14. doi: 10.1016/j.ccell.2021.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Zhang Q, Bi J, Zheng X, et al Blockade of the checkpoint receptor TIGIT prevents NK cell exhaustion and elicits potent anti-tumor immunity. Nat Immunol. 2018;19:723–32. doi: 10.1038/s41590-018-0132-0. [DOI] [PubMed] [Google Scholar]
- 58.Maas RJ, Hoogstad-van Evert JS, Van der Meer JM, et al TIGIT blockade enhances functionality of peritoneal NK cells with altered expression of DNAM-1/TIGIT/CD96 checkpoint molecules in ovarian cancer. Oncoimmunology. 2020;9:1843247. doi: 10.1080/2162402x.2020.1843247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Li M, Xia P, Du Y, et al T-cell immunoglobulin and ITIM domain (TIGIT) receptor/poliovirus receptor (PVR) ligand engagement suppresses interferon-γ production of natural killer cells via β-arrestin 2-mediated negative signaling. J Biol Chem. 2014;289:17647–57. doi: 10.1074/jbc.M114.572420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Masson D, Jarry A, Baury B, et al Overexpression of the CD155 gene in human colorectal carcinoma. Gut. 2001;49:236–40. doi: 10.1136/gut.49.2.236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Nakai R, Maniwa Y, Tanaka Y, et al Overexpression of Necl-5 correlates with unfavorable prognosis in patients with lung adenocarcinoma. Cancer Sci. 2010;101:1326–30. doi: 10.1111/j.1349-7006.2010.01530.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Liu S, Zhang H, Li M, et al Recruitment of Grb2 and SHIP1 by the ITT-like motif of TIGIT suppresses granule polarization and cytotoxicity of NK cells. Cell Death Differ. 2013;20:456–64. doi: 10.1038/cdd.2012.141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Sloan KE, Eustace BK, Stewart JK, et al CD155/PVR plays a key role in cell motility during tumor cell invasion and migration. BMC Cancer. 2004;4:73. doi: 10.1186/1471-2407-4-73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Kono T, Imai Y, Yasuda S, et al The CD155/poliovirus receptor enhances the proliferation of ras-mutated cells. Int J Cancer. 2008;122:317–24. doi: 10.1002/ijc.23080. [DOI] [PubMed] [Google Scholar]
- 65.Carlsten M, Norell H, Bryceson YT, et al Primary human tumor cells expressing CD155 impair tumor targeting by down-regulating DNAM-1 on NK cells. J Immunol. 2009;183:4921–30. doi: 10.4049/jimmunol.0901226. [DOI] [PubMed] [Google Scholar]
- 66.Gur C, Ibrahim Y, Isaacson B, et al Binding of the Fap2 protein of Fusobacterium nucleatum to human inhibitory receptor TIGIT protects tumors from immune cell attack. Immunity. 2015;42:344–55. doi: 10.1016/j.immuni.2015.01.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Sarhan D, Cichocki F, Zhang B, et al Adaptive NK cells with low TIGIT expression are inherently resistant to myeloid-derived suppressor cells. Cancer Res. 2016;76:5696–706. doi: 10.1158/0008-5472.can-16-0839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Du C, Bevers J 3rd, Cook R, et al. MICA immune complex formed with alpha 3 domain-specific antibody activates human NK cells in a Fc-dependent manner. J Immunother Cancer 2019;7:207.
- 69.Courau T, Bonnereau J, Chicoteau J, et al Cocultures of human colorectal tumor spheroids with immune cells reveal the therapeutic potential of MICA/B and NKG2A targeting for cancer treatment. J Immunother Cancer. 2019;7:74. doi: 10.1186/s40425-019-0553-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Hu J, Xia X, Zhao Q, et al Lysine acetylation of NKG2D ligand Rae-1 stabilizes the protein and sensitizes tumor cells to NKG2D immune surveillance. Cancer Lett. 2021;502:143–53. doi: 10.1016/j.canlet.2020.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Badrinath S, Dellacherie M, Li A, et al A vaccine targeting resistant tumours by dual T cell plus NK cell attack. Nature. 2022;606:992–8. doi: 10.1038/s41586-022-04772-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Weil S, Memmer S, Lechner A, et al Natural killer group 2D ligand depletion reconstitutes natural killer cell immunosurveillance of head and neck squamous cell carcinoma. Front Immunol. 2017;8:387. doi: 10.3389/fimmu.2017.00387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Li Y, Hermanson DL, Moriarity BS, et al Human iPSC-derived natural killer cells engineered with chimeric antigen receptors enhance anti-tumor activity. Cell Stem Cell. 2018;23:181–92.e5. doi: 10.1016/j.stem.2018.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Wang J, Lupo K, Chambers A, et al Purinergic targeting enhances immunotherapy of CD73+ solid tumors with piggyBac-engineered chimeric antigen receptor natural killer cells. J Immunother Cancer. 2018;6:136. doi: 10.1186/s40425-018-0441-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Zhang C, Röder J, Scherer A, et al Bispecific antibody-mediated redirection of NKG2D-CAR natural killer cells facilitates dual targeting and enhances antitumor activity. J Immunother Cancer. 2021;9:e002980. doi: 10.1136/jitc-2021-002980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.André P, Denis C, Soulas C, et al Anti-NKG2A mAb is a checkpoint inhibitor that promotes anti-tumor immunity by unleashing both T and NK cells. Cell. 2018;175:1731–43.e13. doi: 10.1016/j.cell.2018.10.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Tinker AV, Hirte HW, Provencher D, et al Dose-ranging and cohort-expansion study of monalizumab (IPH2201) in patients with advanced gynecologic malignancies: a trial of the Canadian Cancer Trials Group (CCTG): IND221. Clin Cancer Res. 2019;25:6052–60. doi: 10.1158/1078-0432.ccr-19-0298. [DOI] [PubMed] [Google Scholar]
- 78.Galot R, Le Tourneau C, Saada-Bouzid E, et al A phase II study of monalizumab in patients with recurrent/metastatic squamous cell carcinoma of the head and neck: The I1 cohort of the EORTC-HNCG-1559 UPSTREAM trial. Eur J Cancer. 2021;158:17–26. doi: 10.1016/j.ejca.2021.09.003. [DOI] [PubMed] [Google Scholar]
- 79.Frazao A, Messaoudene M, Nunez N, et al CD16+NKG2Ahigh natural killer cells infiltrate breast cancer-draining lymph nodes. Cancer Immunol Res. 2019;7:208–18. doi: 10.1158/2326-6066.cir-18-0085. [DOI] [PubMed] [Google Scholar]
- 80.Herbst RS, Majem M, Barlesi F, et al COAST: an open-label, phase II, multidrug platform study of durvalumab alone or in combination with oleclumab or monalizumab in patients with unresectable, stage III non-small-cell lung cancer. J Clin Oncol. 2022;40:3383–93. doi: 10.1200/jco.22.00227. [DOI] [PubMed] [Google Scholar]
- 81.Manguso RT, Pope HW, Zimmer MD, et al In vivo CRISPR screening identifies Ptpn2 as a cancer immunotherapy target. Nature. 2017;547:413–8. doi: 10.1038/nature23270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.van Montfoort N, Borst L, Korrer MJ, et al NKG2A blockade potentiates CD8 T cell immunity induced by cancer vaccines. Cell. 2018;175:1744–55.e15. doi: 10.1016/j.cell.2018.10.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Salomé B, Sfakianos JP, Ranti D, et al NKG2A and HLA-E define an alternative immune checkpoint axis in bladder cancer. Cancer Cell. 2022;40:1027–43.e9. doi: 10.1016/j.ccell.2022.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Kamiya T, Seow SV, Wong D, et al Blocking expression of inhibitory receptor NKG2A overcomes tumor resistance to NK cells. J Clin Invest. 2019;129:2094–106. doi: 10.1172/jci123955. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Berrien-Elliott MM, Cashen AF, Cubitt CC, et al Multidimensional analyses of donor memory-like NK cells reveal new associations with response after adoptive immunotherapy for leukemia. Cancer Discov. 2020;10:1854–71. doi: 10.1158/2159-8290.cd-20-0312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Marin ND, Krasnick BA, Becker-Hapak M, et al Memory-like differentiation enhances NK cell responses to melanoma. Clin Cancer Res. 2021;27:4859–69. doi: 10.1158/1078-0432.ccr-21-0851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Liu WN, So WY, Harden SL, et al Successful targeting of PD-1/PD-L1 with chimeric antigen receptor-natural killer cells and nivolumab in a humanized mouse cancer model. Sci Adv. 2022;8:eadd1187. doi: 10.1126/sciadv.add1187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Fabian KP, Padget MR, Donahue RN, et al PD-L1 targeting high-affinity NK (t-haNK) cells induce direct antitumor effects and target suppressive MDSC populations. J Immunother Cancer. 2020;8:e000450. doi: 10.1136/jitc-2019-000450. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Zhang D, Zheng Y, Lin Z, et al Equipping natural killer cells with specific targeting and checkpoint blocking aptamers for enhanced adoptive immunotherapy in solid tumors. Angewandte Chemie (International ed in English) 2020;59:12022–8. doi: 10.1002/anie.202002145. [DOI] [PubMed] [Google Scholar]
- 90.Nakamura T, Sato T, Endo R, et al STING agonist loaded lipid nanoparticles overcome anti-PD-1 resistance in melanoma lung metastasis via NK cell activation. J Immunother Cancer. 2021;9:e002852. doi: 10.1136/jitc-2021-002852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Charych DH, Hoch U, Langowski JL, et al NKTR-214, an engineered cytokine with biased IL2 receptor binding, increased tumor exposure, and marked efficacy in mouse tumor models. Clin Cancer Res. 2016;22:680–90. doi: 10.1158/1078-0432.ccr-15-1631. [DOI] [PubMed] [Google Scholar]
- 92.Diab A, Tannir NM, Bentebibel SE, et al Bempegaldesleukin (NKTR-214) plus nivolumab in patients with advanced solid tumors: Phase I dose-escalation study of safety, efficacy, and immune activation (PIVOT-02) Cancer Discov. 2020;10:1158–73. doi: 10.1158/2159-8290.cd-19-1510. [DOI] [PubMed] [Google Scholar]
- 93.Jochems C, Tritsch SR, Knudson KM, et al The multi-functionality of N-809, a novel fusion protein encompassing anti-PD-L1 and the IL-15 superagonist fusion complex. Oncoimmunology. 2019;8:e1532764. doi: 10.1080/2162402x.2018.1532764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Knudson KM, Hicks KC, Ozawa Y, et al Functional and mechanistic advantage of the use of a bifunctional anti-PD-L1/IL-15 superagonist. J Immunother Cancer. 2020;8:e000493. doi: 10.1136/jitc-2019-000493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Oyer JL, Gitto SB, Altomare DA, et al PD-L1 blockade enhances anti-tumor efficacy of NK cells. Oncoimmunology. 2018;7:e1509819. doi: 10.1080/2162402x.2018.1509819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Dong W, Wu X, Ma S, et al The mechanism of anti-PD-L1 antibody efficacy against PD-L1-negative tumors identifies NK cells expressing PD-L1 as a cytolytic effector. Cancer Discov. 2019;9:1422–37. doi: 10.1158/2159-8290.cd-18-1259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Lin M, Luo H, Liang S, et al Pembrolizumab plus allogeneic NK cells in advanced non-small cell lung cancer patients. J Clin Invest. 2020;130:2560–9. doi: 10.1172/jci132712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Leem G, Jang SI, Cho JH, et al Safety and efficacy of allogeneic natural killer cells in combination with pembrolizumab in patients with chemotherapy-refractory biliary tract cancer: A multicenter open-label phase 1/2a trial. Cancer (Basel) 2022;14:4229. doi: 10.3390/cancers14174229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Chauvin JM, Ka M, Pagliano O, et al IL15 stimulation with TIGIT blockade reverses CD155-mediated NK-cell dysfunction in melanoma. Clin Cancer Res. 2020;26:5520–33. doi: 10.1158/1078-0432.ccr-20-0575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Johnston RJ, Comps-Agrar L, Hackney J, et al The immunoreceptor TIGIT regulates antitumor and antiviral CD8(+) T cell effector function. Cancer Cell. 2014;26:923–37. doi: 10.1016/j.ccell.2014.10.018. [DOI] [PubMed] [Google Scholar]
- 101.Chauvin JM, Pagliano O, Fourcade J, et al TIGIT and PD-1 impair tumor antigen-specific CD8+ T cells in melanoma patients. J Clin Invest. 2015;125:2046–58. doi: 10.1172/jci80445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Niu J, Maurice-Dror C, Lee DH, et al First-in-human phase 1 study of the anti-TIGIT antibody vibostolimab as monotherapy or with pembrolizumab for advanced solid tumors, including non-small-cell lung cancer☆. Ann Oncol. 2022;33:169–80. doi: 10.1016/j.annonc.2021.11.002. [DOI] [PubMed] [Google Scholar]
- 103.Cho BC, Abreu DR, Hussein M, 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;23:781-92.