Skip to main content
Cellular and Molecular Immunology logoLink to Cellular and Molecular Immunology
. 2019 Feb 18;16(5):430–441. doi: 10.1038/s41423-019-0206-4

Human NK cells: surface receptors, inhibitory checkpoints, and translational applications

Simona Sivori 1, Paola Vacca 2, Genny Del Zotto 3, Enrico Munari 4,5, Maria Cristina Mingari 1,6, Lorenzo Moretta 2,
PMCID: PMC6474200  PMID: 30778167

Abstract

NK cells play important roles in innate defenses against viruses and in the control of tumor growth and metastasis. The regulation/induction of NK cell function is mediated by an array of activating or inhibitory surface receptors. In humans, major activating receptors involved in target cell killing are the natural cytotoxicity receptors (NCRs) and NKG2D. Activating receptors recognize ligands that are overexpressed or expressed de novo upon cell stress, viral infection, or tumor transformation. The HLA-class I-specific inhibitory receptors, including KIRs recognizing HLA-class I allotypic determinants and CD94/NKG2A recognizing the class-Ib HLA-E, constitute a fail-safe mechanism to avoid unwanted NK-mediated damage to healthy cells. Other receptors such as PD-1, primarily expressed by activated T lymphocytes, are important inhibitory checkpoints of immune responses that ensure T-cell tolerance. PD-1 also may be expressed by NK cells in cancer patients. Since PD-1 ligand (PD-L1) may be expressed by different tumors, PD-1/PD-L1 interactions inactivate both T and NK cells. Thus, the reliable evaluation of PD-L1 expression in tumors has become a major issue to select patients who may benefit from therapy with mAbs disrupting PD-1/PD-L1 interactions. Recently, NKG2A was revealed to be an important checkpoint controlling both NK and T-cell activation. Since most tumors express HLA-E, mAbs targeting NKG2A has been used alone or in combination with other therapeutic mAbs targeting PD-1 or tumor antigens (e.g., EGFR), with encouraging results. The translational value of NK cells and their receptors is evidenced by the extraordinary therapeutic success of haploidentical HSCT to cure otherwise fatal high-risk leukemias.

Keywords: Human NK cells, NK receptors, Inhibitory checkpoints, Immunotherapy

Subject terms: Innate lymphoid cells, Cancer, Innate immunity

Introduction and general overview of human NK cells

Natural killer (NK) cells play major roles in first-line innate defenses against viral infections, tumor growth, and metastatic spread. Both the constitutive expression of efficient, ready-to-function lytic machinery, and the rapid release of interferon-gamma (IFN-γ) and tumor necrosis factor-alfa (TNF-α) following cell activation allow prompt intervention by NK cells, resulting in target cell killing and the initiation of an inflammatory response.

NK cells were discovered in the mid-70s;1,2 however, the mechanism by which they discriminate between tumor or virus-infected cells and healthy cells remained a mystery for many years. The missing self-hypothesis, proposed in the late 80s by Karre and Ljunggren,3 represented a true milestone, inspiring subsequent groundbreaking discoveries in the early 90s. This hypothesis was based on the finding that murine NK cells could kill a lymphoma cell line that had lost major histocompatibility complex-class I (MHC-cl I) molecules, while parental MHC-cl I+ cells were resistant to lysis. Thus, NK cells appear to sense the absence of MHC-cl I on cells (i.e., the missing self).4 Research aimed to identify the molecular mechanism(s) involved in this phenomenon, leading to the discovery of inhibitory receptors specific for MHC-cl I molecules in both humans and in mice. Remarkably, while the first identified human receptors specific for human leukocyte antigen (HLA)-cl I molecules belong to the immunoglobulin (Ig) superfamily (and were subsequently named killer-cell immunoglobulin-like receptors, KIRs), the murine receptors (Ly49) are members of the lectin family. Thus, humans and mice developed two molecularly different receptors fulfilling the same function, i.e., the recognition of MHC-cl I molecules and the delivery of inhibitory signals to NK cells, resulting in NK cell inactivation.5 The discovery of KIRs was made possible by the availability of two important technologies, namely, monoclonal antibody technology and a high-efficiency lymphocyte cloning system. The latter technology allowed the clonal expansion of virtually 100% of human T lymphocytes.6 Thus, it has been possible to analyze the frequency of cells endowed with a given function (e.g., cytolytic activity) and to establish correlations between the surface phenotype and function of any T-cell population.7 After suitable adaptation of this cloning technique, NK cells were also revealed to be clonogenic and suitable for precise functional studies. NK cell clones were used for mouse immunization and the production of monoclonal antibodies (mAbs) that were selected for their ability to either inhibit or induce NK cell cytolytic activity or cytokine production. Two selected mAbs, originally defined as p58.1 and p58.2810, recognized two highly homologous molecules expressed by partially overlapping NK cell subsets. These mAbs represented prototypes of HLA-cl I-specific inhibitory receptors, and were found to recognize allotypic determinants shared by the two main groups of HLA-C alleles.1013

Other HLA-cl I-specific receptors were identified (see below). Of relevance, the HLA-E-specific NKG2A,14 expressed earlier than KIR during NK cell maturation, was recently found to be of great potential interest as a target of checkpoint inhibitors in tumor immunotherapy15,16(see below).

Remarkably, the pool of mature NK cells is equipped with at least one inhibitory receptor for self HLA-cl I antigens (whether KIR or NKG2A). Only very few peripheral blood (PB) NK cells lack such receptors and are anergic. The repertoire of inhibitory NK receptors is shaped during NK cell maturation and is the result of a process of selection termed NK cell “licensing” or “education”.1719 Accordingly, only NK cells expressing receptors for self HLA-cl I molecules acquire full functional potential, while the remaining are either deleted or anergic.

“Off” signals are necessary to prevent NK-mediated autoreactivity, implying the existence of “on” signals responsible for NK cell activation. Indeed, in the absence of inhibitory interactions, NK cells kill target cells and produce cytokines. Again, the use of NK cell clones and the selection of mAbs were fundamental for the discovery of prototypes that activate NK receptors, namely, NKp46,2022 NKp4423 and NKp30,24 collectively termed “natural cytotoxicity receptors” (NCRs).25 mAbs directed to these surface molecules were selected based on their ability to modulate NK clone-mediated cytotoxicity against selected tumor targets. It became evident that NK cell activation is not due to a master receptor (such as TCR and BCR in T and B cells, respectively) but, in most instances, is the result of the combined action of several receptors and coreceptors (see below). Triggering of the activating NK receptors requires the expression of appropriate ligands on potential target cells. Such ligands are overexpressed or expressed de novo in stressed healthy cells and, more importantly, in cells that have undergone tumor transformation or viral infection.26 Remarkably, tumors and certain viruses may induce the downregulation of activating NK receptors and/or their ligands on target cells, thus favoring escape from the NK-mediated control. This is evident in a relevant percentage of tumors, particularly neuroblastoma, in which not only HLA-cl I molecules but also ligands for most activating NK receptors may be downregulated.27 This mechanism of tumor escape underscores the potential importance not only of T cells but also of NK cells in the immune response against neuroblastoma. Taken together, the expression of inhibitory and activating receptors in NK cells and their ligands on target cells revealed that NK cell activation/inactivation is controlled by different checkpoints.28 This notion is important for translational applications that exploit NK cell function in therapy for leukemia or solid tumors (see below).

Human NK cells have been divided into two major subsets identified according to the surface density of the CD56 antigen.29 CD56bright cells represent only ~10% of PB NK cells, but they predominate in tissues. These cells express NKG2A, are poorly cytolytic but secrete cytokines, primarily IFN-γ and TNF-α, and efficiently proliferate in response to interleukin (IL)-2 or IL-15. CD56dim cells largely predominate in PB (~90%), expressing NKG2A or KIR or both, and rapidly display potent cytolytic activity and cytokine production upon activation. Based on the expression of receptors/markers, CD56dim NK cells are further subdivided into different stages of differentiation, characterized by a progressive decrease in proliferative capacity paralleled by an increase in cytolytic activity.30 The most mature NK cells are KIR+ NKG2Aneg and express CD57 and (in individuals seropositive for cytomegalovirus, CMV) NKG2C, an HLA-E-specific activating receptor. Recent studies revealed that NKG2C+ NK cells display adaptive features, sensing different HLA-E-bound CMV peptides and undergo selective proliferation only in response to given HLA-E-peptide combinations.31 Such specific expansion, together with a number of adaptive characteristics, is compatible with the memory-like function of these cells, which can allow for prompt control of CMV reactivation.32

While NK cells have been known for over 45 years, other cells that are developmentally related to NK cells were identified only 10 years ago and collectively termed innate lymphoid cells (ILCs).33,34 Similar to NK cells, ILCs derive from CD34+ hematopoietic precursors and share with NK cells a common ID2+ lymphoid precursor.35 Absent or infrequent in PB from healthy individuals, ILCs reside in mucosal tissues, skin, and lymphoid organs. Unlike NK cells, the other ILCs (termed “helper ILCs”) are noncytolytic and secrete typical sets of cytokines that differ for each subset, including IFN-γ (ILC1), IL-5, IL-13, and small amounts of IL-4 (ILC2), IL-22 (NCR+ ILC3), IL-17, and TNF-α (NCRneg ILC3 or LTi-like cells).36 Interestingly, helper ILCs mimic corresponding CD4+ T-cell subsets in terms of the type of cytokines produced. Therefore, these cytokines can shape not only adaptive but also innate immune responses, although with different timing. In general, ILCs contribute to innate defenses against various pathogens and are involved in tissue repair and homeostasis and in lymphoid organogenesis, particularly during fetal life. While they will not be discussed further in this review, which is devoted to NK cells, it is worth noting that ILCs may share some markers and the NCR activating receptors with NK cells.37

NK cell traffic in the circulation and towards tissues and lymphoid organs is regulated by chemokines and chemokine receptors, directing the given NK cell subsets to specific sites.38 Thus, while the expression of CD62L, CXCR3, and CCR7 will direct CD56bright NK cells to secondary lymphoid organs (in response to CCL19 and CCL21), the expression of Chemerin R, CXCR1, CXCR2, and CX3CR1 directs CD56dim NK cells to inflammatory peripheral tissues in response to their corresponding chemokine ligands (Chemerin, IL-8, and Fraktalkine). Since CD34+ cells capable of generating NK cells have been detected in different tissues, including thymus,39 tonsils,40,41 decidua42, and liver,43 it is conceivable that at least some tissue-resident NK cells may derive from these precursors, migrate from BM, and acquire unique properties as a consequence of their particular tissue microenvironment.44

The following paragraphs will analyze in more detail the various NK receptors, recently identified inhibitory checkpoints, and their implications in the immunotherapy of tumors. In addition, we will outline the fundamental role of NK receptors in the cure of high-risk leukemias in allogeneic hemopoietic stem cell transplantation (HSCT).

Inhibitory and activating NK receptors and their roles in tumor cell killing

HLA-specific NK receptors

As mentioned above, important regulation of NK cell function is provided by inhibitory receptors specific for HLA-cl I molecules. Indeed, HLA-cl I molecule recognition on healthy cells by these inhibitory NK receptors prevents NK cell-mediated attack.

Human NK cells express two different classes of HLA-class I-specific inhibitory receptors: members of the KIR/CD158 family and the CD94/NKG2A (CD94/CD159a) heterodimer.5,45 KIRs are type I transmembrane receptors specific for polymorphic HLA-A, B and C molecules,810,12,13 whereas NKG2A is a type II transmembrane receptor that recognizes HLA-E, a non-classical HLA molecule characterized by limited polymorphism.14 To transduce inhibitory signals, both types of inhibitory receptors contain ITIM motifs in their cytoplasmic tail. In addition, activating forms of KIRs have been identified.46 Different from inhibitory KIRs, activating KIRs lack ITIM motifs in their cytoplasmic tail and have a transmembrane domain carrying a charged amino acid residue47 that mediates the association with the ITAM-bearing molecule KARAP/DAP12. The role of activating KIRs in the immune response is only partially known. Specificity for HLA-cl I molecules has been demonstrated for only a few of them.46,4854

Inhibitory KIRs are characterized by long cytoplasmic tails, whereas activating KIRs have short cytoplasmic tails (“L” or “S” in the nomenclature, respectively). Regarding Ig domain content, each KIR displays two or three extracellular Ig domains (“KIR2D” or “KIR3D” in the nomenclature, respectively). Two types of KIR2D can be determined according to their extracellular Ig domain content. KIR2Ds of the first type are composed of D1 and D2 domains and include the majority of KIRs (KIR2DL1/L2/L3 and KIR2DS1/S2/S3/S4/S5), whereas KIR2Ds of the second type are composed of D0 and D2 domains and include KIR2DL4/L5.55 Notably, not only HLA class I but also KIRs are characterized by high levels of polymorphism, which may affect given KIR/HLA interactions. In addition, certain KIR/HLA combinations have been shown to correlate with protection or susceptibility to infectious, autoimmune, and reproductive disorders.

Another HLA-specific inhibitory receptor is represented by LIR-1/ILT2/CD85j.56 LIR-1 is a type I transmembrane protein belonging to the Ig-like receptor superfamily that can interact with classical (HLA-A, HLA-B, HLA-C) and non-classical (HLA-G) HLA-cl I molecules.5759 It can also bind UL18, a cytomegalovirus-encoded HLA-cl I homolog that is expressed on CMV-infected cells.56 Notably, high LIR-1 expression correlates with the acquisition of NK cell memory in CMV+ donors.60

Another HLA-specific activating receptor is represented by NKG2C, a receptor that, similar to NKG2A, binds HLA-E but with lower affinity.61,62 Altogether with inhibitory KIRs, CD94/NKG2A prevents the response against cells with normal expression of HLA-I molecules, whereas CD94/NKG2C is involved in the response to human HCMV. Notably, NKG2A is primarily expressed by PB immature NK cells, whereas NKG2C is expressed only at late stages of NK cell maturation.63 The terms “adaptive” or “memory-like” are currently employed to designate the human differentiated NKG2Cbright NK cell subset that is characterized by the CD56dim CD57+ KIR+ NKG2Aneg phenotype and that is expanded in HCMV+ donors.32,64,65 Similar to activating KIRs, NKG2C is coupled to the ITAM-bearing molecule KARAP/DAP12.

Activating NK receptors and coreceptors involved in tumor cell killing and their ligands

Human NK cells express several receptors that can trigger their function upon interaction with specific ligands on the surface of transformed, virus-infected, or stressed cells (Table 1).

Table. 1.

Human NK cell receptors and their ligands

Molecule Ligand CD56bright CD56dim
Non-HLA-specific receptors
 Coreceptors
  CD59 LFA-2 (CD2) All PB NK cells
  NTB-A (CD352) NTB-A (CD352) All PB NK cells
  NKp80 AICL All PB NK cells
  DNAM-1 (CD226) Nectin-2 (CD112), PVR (CD155) All PB NK cells
  2B4 (CD244) CD48 All PB NK cells
 Inhibitory
  PD-1 (CD279) PD-L1 (CD274), PD-L2 (CD273)  − Subsets
  Siglec-7 (CD328) Ganglioside DSGb5 Most of PB NK cells
  IRP60 (CD300a) α-Herpes virus, Pseudorabide virus, Phosphatidylserine, Phosphatidylethnolamine All PB NKs
  Tactile (CD96) PVR (CD155) All PB NK cells
  IL1R8 IL-37 PB NK cells
  TIGIT PVR (CD155) PB NK cells
  TIM-3 Gal-9, PtdSer, HMGB1, CEACAM1 Subset of NK cells
 Activating
  NKp30 (CD337) B7-H6, BAG6/BAT3  ++  +
  NKp44 (CD336) 21spe-MLL5 - Nidogen-1 Activated NK cells
  NKp46 (CD335) CFP (properdin), viral HA and HN, PfEMP1  ++  +
  NKG2D (CD314) MIC-A, MIC-B, ULPBs All PB NK cells
  FcγRIII (CD16) IgG  −/+  +/++
HLA-specific receptors
 Inhibitory
  NKG2A/KLRD1 (CD159a/CD94) HLA-E  + Subsets
  KIR2DL1 (CD158a) HLA-C2  − Subsets
  KIR2DL2/3 (CD158b) HLA-C1, few HLA-Bb  − Subsets
  (CD158d)a HLA-G  − Subsets
  KIR2DL5 (CD158f) ???  − Subsets
  KIR3DL1 (CD158e1) HLA-A-Bw4, HLA-B-Bw4  − Subsets
  KIR3DL2 (CD158k) HLA-A*03 and *11  − Subsets
  ILT2/LIR-1 (CD85J) Different MHC-I alleles . Subsets
  LAG-3 (CD223) MHC-II Activated NK cells
 Activating
  KIR2DS1 (CD158h) HLA-C2  − Subsets
  KIR2DS2/3 (CD158j) ???  − Subsets
  KIR2DL4 (CD158d)a HLA-G  − Subsets
  KIR2DS4 (CD158i) HLA-A*11 and some HLA-C  − Subsets
  KIR2DS5 (CD158f) ???  _ Subsets
  KIR3DS1 (CD158e1) HLA-Bw4, HLA-F  − Subsets
  NKG2C (CD159a) HLA-E  − Subsets
Homing receptors
 2° lymphoid tissues
  CCR7 (CD197) CCL19, CCL21  +  −
  CXCR3 (CD183) CXCL9, CXCL10, CXCL11  ++ Subsets
  L-Selectin (CD62L) GLyCAM-1, MadCAM-1  ++ Subsets
 Inflammation sites
  CXCR1 (CD181) CXCL8 (IL-8)  −  +
  CXCR2 (CD182) IL-8-RB  −  +
  CX3CR1 Fraktalkine  −  +
  ChemR23 Chemerin  −  +
 Others
  CXCR4 (CD184) CXCL2 Subsets of NK cells
  CCR5 (CD195) RANTES, MIP1α and MIP1β Subsets of NK cells
  S1P5 S1P  −  +
  c-Kit (CD117) SCF (KL)  +  −

aKIR2DL4 has been shown to have both inhibitory and activating functions164

bKIR2DL2/L3 weakly recognize also HLA-C2 alleles and few HLA-B alleles that bear the HLA-C1 epitope (e.g., HLA-B*4601 and HLA-B*7301)

The NCRs25 are among the major activating NK receptors and consist of three elements, called NKp46/NCR1/CD335,2022 NKp44/NCR2/CD33623, and NKp30/NCR3/CD337.24 These molecules were classically described as germline-encoded receptors and are important for inducing NK cell cytotoxic function against tumors and infected cells.

NKp46 and NKp30 are expressed on nearly all resting human NK cells, upregulated on activated NK cells and downregulated on “adaptive” NK cells that are found in CMV+ individuals. Unlike NKp46 and NKp30, NKp44 is constitutively expressed only on CD56bright NK cells, but it is acquired by essentially all NK cells after activation by cytokines.

NCRs are type I transmembrane molecules belonging to the immunoglobulin-like family.

These receptors were named in accordance with their molecular weights according to sodium dodecyl sulfate polyacrylamide gel electrophoresis (NKp46, NKp30, and NKp44). Their transmembrane domains contain a positively charged amino acid that allows their association with the transmembrane regions of the TCR-ζ and/or FcεRI-γ (for NKp30 and NKp46) or KARAP/DAP1266 (for NKp44) adaptor proteins. In this regard, it is important to underline that the physical association between NKp44 and KARAP/DAP12 is essential for NKp44 surface expression67 and that the decreased surface density of NKp30 and NKp46 on “adaptive” NK cells is associated with the lack of FcεRI-γ expression in these cells.68

Different molecules can interact with the extracellular domains of NCRs. Some of them are virus-derived molecules that can activate NK cell function against infected cells (such as influenza virus-derived HA recognized by NKp46 or NKp4469) or induce inhibitory signals (such as the HCMV-encoded pp65 protein recognized by NKp30.70) Other NCR ligands (such as BAT3/BAG6, MLL5, and PCNA) are represented by intracellularly localized proteins that may reach the cell surface in response to stress or during tumor transformation. For example, BAT3/BAG6 is typically located in the nucleus;71 however, in cells exposed to heat shock, it can move to the plasma membrane and be secreted in exosomes by tumors and stressed cells.71,72 Then, BAT3/BAG6-expressing exosomes can stimulate cytokine release by NK cells upon interaction with NKp30. MLL5 is expressed in the nuclei of normal cells, but 21spe-MLL5, an MLL5 isoform that functions as an NKp44 ligand, is located in the cytoplasm and at the cell surface.73 Finally, the nuclear protein PCNA can function also as an NCR ligand. Indeed, in normal cells, PCNA is involved in the processes of DNA replication/repair and cell cycle control, but in tumor cells, it can be shuttled to the tumor cell surface and expressed on tumor-derived exosomes, functioning as an NKp44 ligand.74

B7-H6, another NKp30 ligand,75 is not expressed on healthy cells, but it is frequently present on the cell surface of many tumor types through a Myc-mediated mechanism.76 Interestingly, its expression may also be acquired by normal cells upon TLR-mediated stimulation and in the presence of pro-inflammatory cytokines.77

Recently, NKp46 and NKp44 have also been shown to recognize extracellular ligands. Indeed, NKp46 can bind to a soluble plasma glycoprotein called complement factor P/properdin, revealing cross-talk between two partners in innate immunity in the response to Neisseria meningitidis infections.78 Moreover, NKp44 has also been shown to recognize an extracellular ligand called Nidogen-1 (NID1, also known as Entactin).79 The NKp44/NID1 interaction results in reduced NKp44-mediated cytokine release by NK cells and induces relevant changes in the NK cell proteomic profile, suggesting an effect on different biological processes.

Importantly, it has been shown that tumors can orchestrate different mechanisms to impair NCR function. Thus, hypoxia or various soluble factors produced by tumor/tumor-associated cells (such as indoleamine 2,3 dioxygenase [IDO], tumor growth factor-beta [TGF-β], prostaglandin E2 [PGE2]), or inhibitory NCR ligands (such as the soluble form of BAT3 or B7-H6)72,80 can induce decreases in NCR expression and function.81 Indeed, NCRlow NK cells can be detected in PB and particularly in the tumor site in patients affected by solid and hematologic tumors. Notably, reduced expression/function of NCRs can also be detected in NK cells from HIV-infected patients.82

Another important activating NK receptor is NKG2D, a type II transmembrane and C-type lectin-like receptor, which may be expressed on cytotoxic T cells. NKG2D ligands are represented by ULBPs and MICA/B,83 which are HLA-cl I structural homologs that are upregulated in infected, stressed, and tumor cells.84,85 Notably, shedding of NKG2D ligands by tumor cells may represent a mechanism of tumor escape.

Other molecules, including 2B4,86 NTB-A,87 DNAM-1,88 CD59,89 and NKp80,90 function primarily as coreceptors; indeed, they are capable of amplifying the NK cell triggering induced by NCRs or NKG2D. In addition, NK cells may express toll-like receptors (TLRs) that, after interaction with bacterial or viral products and in the presence of pro-inflammatory cytokines, induce potent NK cell activation.9194

Finally, the Fcγ receptor CD16, recognizing the Fc portion of IgG antibodies specific for unhealthy cells, can trigger antibody-dependent cell-mediated cytotoxicity (ADCC).95 CD16bright expression is restricted to mature CD56dim KIR+ NK cells.

The molecular basis of NK cell function

NK cell function is regulated by the activating and inhibitory receptors illustrated above. The fine balance of signaling that occurs through them determines if NK cells kill their target cells or remain inactive. As discussed, autoreactivity in human NK cells is controlled by KIRs and NKG2A inhibitory receptors specific for self HLA-cl I molecules. KIRs and NKG2A are clonally expressed or coexpressed in NK cell subsets, thus creating repertoires of NK cells with different phenotypic and functional characteristics that are capable of responding to different types of virally infected and tumor-transformed self cells. Under normal conditions, the interactions between these inhibitory receptors and their specific HLA-cl I ligands inactivate NK cells, thus preventing cytolytic activity against healthy cells. During cancer progression, the transformed cells decrease or even lose the expression of HLA-I on their surface. In addition, they express ligands for activating NK receptors: two events necessary for the induction of antitumor NK cell responses. Indeed, ligands for the activating NK receptors are generally absent or expressed in small amounts in normal cells, while they are expressed de novo or upregulated at the cell surface in stressed normal cells and, in particular, in virus-infected or tumor cells. Therefore, signaling through activating receptors can overcome the signaling mediated by inhibitory receptors on diseased cells.25,96

NK inhibitory checkpoints and their ligands in solid tumors

Inhibitory checkpoints in NK cells

In addition to the HLA-cl I-specific inhibitory receptors that regulate NK cell function and prevent NK-mediated damage to healthy tissues, additional inhibitory checkpoints, responsible for maintaining immune cell homeostasis, have been described in NK cells, including PD-1, TIGIT, CD96, TIM-3, etc. Under pathological conditions, some of these checkpoint regulators, which are absent on resting NK cells, can be induced de novo and affect antitumor NK cell function upon interaction with their specific ligands that are frequently expressed on the tumor cell surface and facilitate tumor immune escape.9799

PD-1, a member of the immunoglobulin superfamily, is a major checkpoint regulating T-cell activation and ensuring peripheral tolerance.100,101 However, when binding to its ligands (PD-L1 and PD-L2), which may be expressed on tumor cells, PD-1 may compromise the antitumor effector function favoring tumor immune escape. Notably, in mice, the blockade of PD-1 resulted in restoration of T-cell responses.102

Although the expression of PD-1 has been described on T, B, and myeloid cells, recent studies have shown that, under pathological conditions, it is also expressed by NK cells.103,104 PD-1+NK cells that can inactivate NK cell function were found in patients with CMV infections and cancer, including Kaposi sarcoma, ovarian carcinoma, and Hodgkin lymphoma.63,104107

A recent study by our group revealed the presence of a pool of PD-1 proteins and mRNA in the cytoplasm of healthy donor NK cells. The molecular mechanisms regulating the expression of PD-1 on human NK cells have not been defined so far. However, it is conceivable that signals delivered by cells and/or soluble factors in the tumor microenvironment may play a major role (Mariotti et al. in press).

In mice, PD-1 expression may be induced by glucocorticoids, underscoring the importance of specific signaling for its regulation.108 Several murine tumor models showed that PD-1/PD-L1 interactions can strongly suppress NK cell-mediated antitumor immunity and that PD-1+ NK cells present at the tumor site display an exhausted phenotype.109

In humans, in addition to PD-1, other inhibitory checkpoints are expressed not only by T lymphocytes but also by NK cells. They include the T-cell Ig and ITIM domain (TIGIT) and CD96 (tactile). These receptors are members of a group of Ig superfamily receptors that also include the activating receptor DNAM-1. Their corresponding ligands, usually upregulated in tumor cells, are the poliovirus receptor (CD155), nectin-2 (CD112), and nectin-like molecules.110,111 TIGIT expression has been reported in different malignancies in peritumoral lymphocytes, often together with PD-1 and TIM-3, and is associated with CD8+ T-cell and NK cell suppression.112 For example, TIGIT was upregulated in tumor-associated NK cells of patients with colon-rectal cancer. TIGIT knockout mice or the use of mAbs that block TIGIT could induce/potentiate NK and T-cell antitumor activity, prevent NK cell exhaustion and lead to the control of tumor growth. A recent study provided evidence that TIGIT targeting with specific mAbs may represent a new promising tool that can be used alone or in combination with other checkpoints.113 Thus, it has been shown that TIGIT blockade may induce antitumor immune activity in preclinical models, and its combination with PD-1/PD-L1 inhibitors is being explored.114

KLRG1, another inhibitory C-type lectin-like receptor containing one ITIM, is expressed by activated NK cells, ILC2s, T cells, mast cells, basophils, and eosinophils.115 The interaction between KLRG1 and its corresponding ligand E-cadherin has been shown to inhibit human ILC2 function in vitro; however, its actual effect in vivo remains to be established.116118 In non-small cell lung cancers (NSCLC) and colon-rectal carcinoma (CRC), KLRG1 expression was detected in tumor-associated ILC2s.119,120

Other inhibitory receptors expressed by NK cells include LAG-3 (CD223) and TIM-3. LAG-3 is a negative costimulatory receptor homologous to CD4 that binds MHC-cl I molecules on antigen presenting cells (APC) with very high affinity. LAG-3 expression is associated with an exhausted profile of tumor-infiltrating-lymphocytes (TIL), and its blockade could restore antitumor immune function.121123 While its inhibitory activity in T-cell activation, proliferation and homeostasis has been defined, the effect of LAG-3 on NK cells remains poorly explored.124 TIM-3, also known as Hepatitis A virus cellular receptor 2 (HAVCR2), is a type 1 glycoprotein expressed by subsets of T lymphocytes (Th17 and regulatory T cells), dendritic cells and mature NK cells. The main ligand of TIM-3 is Galectin-9,125 but other ligands have been identified, such as phosphatidylserine (PtdSer), high mobility group protein 1 (HMGB1), and carcinoembryonic antigen related cell adhesion molecule 1 (CEACAM1).126128 Previous studies have shown that in TILs of lung,129 gastric,130 head and neck carcinomas131, and in melanoma,132 TIM-3 expression is upregulated. Moreover, the increased TIM-3 expression on NK cells in melanoma or lung adenocarcinoma was shown to be associated with impaired NK cell effector function.133,134 TIM-3 is often coexpressed with PD-1 and is associated with T-cell exhaustion during chronic infection and cancer. Blockade of TIM-3 could restore T-cell effector function in preclinical models and result in increased NK cytotoxicity.132,135

Another important inhibitory receptor, IL1R8 (also known as TIR8 or SIGIRR), is a member of the IL1 receptor family and a component of the human IL-37 receptor.136,137 It has been shown to play a physiologic role in the regulation of responses to pathogens by dampening excessive inflammatory responses and tissue damage. A recent study has shown that IL1R8 is highly expressed in a subset of NK cells, in which it exerts major inhibitory control of cell activation and function. In a murine model, NK cells lacking IL1R8 could prevent the growth of a carcinogen-induced hepatocellular carcinoma. This study provided evidence that IL1R8 functions as an important inhibitory checkpoint in NK cell activation and function. Accordingly, its blocking can unleash NK cells and promote efficient antitumor activity.138

PD-L1 and other checkpoint ligands

As illustrated above, PD-1 negatively regulates immune responses; under physiological conditions, these interactions lead to peripheral T-cell tolerance, while in cancer patients they may impair T-cell responses against tumor cells. In this context, immunotherapy with checkpoint inhibitors that disrupt the PD-1/PD-L1 interaction has proven to be highly effective in different tumor types, representing a true revolution in cancer therapy.139 Since the approval of nivolumab and pembrolizumab in 2014, the number of PD-1/PD-L1 inhibitors has grown rapidly. Currently, there are six PD-1/PD-L1 inhibitors that have been approved by the Food and Drug Administration (FDA): three against PD-1 (pembrolizumab, nivolumab and cemiplimab) and three targeting PD-L1 (atezolizumab, durvalumab and avelumab). In parallel with this rapidly expanding arsenal, the spectrum of advanced malignancies for which each of these agents is indicated has broadened, including melanoma (pembrolizumab and nivolumab), non-small cell lung cancer (pembrolizumab, nivolumab, and atezolizumab), urothelial cancer (pembrolizumab, nivolumab, atezolizumab, durvalumab, avelumab), renal cell carcinoma (nivolumab), head and neck cancer (pembrolizumab and nivolumab), Hodgkin lymphoma (pembrolizumab and nivolumab), microsatellite instability or mismatch repair deficient (dMMR) cancers including dMMR colorectal cancer (pembrolizumab, nivolumab), gastric cancer (pembrolizumab), hepatocellular carcinoma (nivolumab), Merkel cell carcinoma (avelumab)140, and cutaneous squamous cell carcinoma (cemiplimab).141 Although PD-1/PD-L1 inhibitors represent a revolution in the field of oncology, a significant proportion of patients do not benefit from this class of agents; therefore, predicting tumor responses to PD-1/PD-L1 blockade represents a major issue. So far, the most widely recognized parameter of tumor response is PD-L1 expression in tumor cells and/or immune cells, for which four assays have been approved to guide treatment decisions for different agents (Dako 22C3, Dako 28-8, Ventana SP142 and Ventana SP263).142 However, despite the clinical utility of PD-L1 expression as a biomarker of the response to PD-1/PD-L1 inhibitors, its predictive value still appears to be unsatisfactory, to the point that other parameters are being explored (including tumor mutational burden [TMB] and the presence and the numbers of TIL and genetic polymorphisms, among others). In this context, all possible limitations inherent to PD-L1 expression should be taken into account, including intra- and intertumor heterogeneity as well as technical issues represented by the use of different mAbs and different diagnostic materials (cytology versus diagnostic biopsies versus surgical specimens), in order to improve the predictive potential of PD-L1 immunohistochemistry.98,99,143145 In any case, a significant proportion of patients would still be not eligible or not responsive to PD-1/PD-L1 axis inhibitors. For this reason, as illustrated above, ongoing research is aimed at finding additional checkpoints other than PD-1/PD-L1 and CTLA-4 to be exploited for targeting by therapeutic antibodies either alone or in combination.

Importantly, while the TCR-mediated antitumor activity of T lymphocytes correlates with TMB, this is not the case for NK cells. Therefore, therapeutic blockade of inhibitory checkpoint or their ligands may still be effective in tumors with NK cell infiltration. For this reason, there is a growing interest in the less-explored NK cell compartment for therapeutic purposes.146,147 As indicated, promising inhibitory checkpoints are LAG-3, TIM-3, TIGIT, HLA-E, and HLA-G, among others.

In this context, two HLA-cl I b molecules expressed on tumors may be particularly relevant.

HLA-E is overexpressed in many tumors.148 As illustrated below, a recent work demonstrated that monalizumab, a humanized anti-NKG2A antibody, enhanced NK cell activity against various tumor cells and rescued CD8+ T-cell function alone or in combination with PD-1/PD-L1 axis blockade.15 HLA-G is physiologically expressed by trophoblast cells, which are mainly involved in maternal-fetal tolerance.149 HLA-G can interact with immunoglobulin-like transcript 2 (ILT2) expressed on NK, T and B lymphocytes, monocytes/macrophages, and dendritic cells and with ILT4, which is expressed only by myeloid cells, as well as KIR2DL4 expressed on NK cells. Under normal conditions, HLA-G is virtually absent in adult tissues; by contrast, most tumors express HLA-G at different levels, either on their cell membranes or released as soluble isoforms. Different studies have shown that HLA-G acts by impairing the cytolytic function and proliferation of peripheral and uterine NK cells and of cytotoxic T cells, the maturation and function of dendritic cells while inducing Tregs and myeloid suppressive cells, thus inhibiting different actors involved in antitumor responses.150 Therefore, HLA-G plays a key role in the induction of immune tolerance, representing an important escape mechanism of tumor cells. However, the therapeutic role of HLA-G-based immunotherapy is still virtually unexplored. We speculate that the use of T or NK cells expressing chimeric antigen receptors (CAR-T or CAR-NK) targeting HLA-G could represent a promising approach for the adoptive immunotherapy of different tumors. Studies in this context are in progressing in our labs.

Exploiting NK cells for translational applications

The majority of current tumor immunotherapies are based on the use of mAbs that target tumor antigens or unlock inhibitory checkpoints expressed by T lymphocytes or the generation of T cells expressing CARs specific for tumor antigens. However, recent contributions reassessed or confirmed the importance of NK cells in tumor therapy. Indeed, as illustrated under the headings below, NK cells can fulfill unique antitumor activities such as ADCC, thanks to the expression of CD16 or the killing of HLA-cl I-deficient tumor cells that are undetectable by cytolytic T lymphocytes. In addition, masking the HLA-cl I-specific KIR or NKG2A inhibitory receptors may induce NK cell-mediated cytolytic activity against HLA-cl I+ target cells. Finally, the generation of CAR-NK cells with extensive proliferative capacity is now available and may be implemented with CAR-T cell therapy, exploiting the homing properties and functional characteristics typical of NK cells.

Targeting NKG2A alone or in combination with other therapeutic mAbs

As underlined above, blocking monoclonal antibodies specific for inhibitory checkpoints have revealed unprecedented potential in tumor therapy, representing a true revolution in the prognosis of a subset of otherwise highly aggressive cancers, including NSCL cancer, metastatic melanoma, kidney tumors and others.139 In spite of this major progress, the majority of patients do not respond to treatment with mAbs targeting the CTLA-4 or PD-1-mediated inhibitory pathways. This partial failure may be due to one or more mechanisms of tumor escape. Thus, as mentioned above, in many instances, tumor progression is accompanied by a partial or total loss of classical HLA-cl I antigens, which are necessary for tumor antigen presentation to cytolytic T lymphocytes. In addition, the tumor microenvironment may greatly contribute to tumor escape. Indeed, an important mechanism is the release of cytokines (e.g., TGF-β) and soluble inhibitory factors (e.g., kynurenine, PGE2), not only from tumor cells but also from tumor-infiltrating cells (e.g., myeloid-derived suppressor cells, M2 macrophages, fibroblasts/MSC). Additionally, the hypoxic environment typical of many tumors may play a substantial negative role.151 Of note, all of these mechanisms impair both T- and NK cell function by targeting receptor-associated polypeptides that are crucial for the transmission of activating signals. Although different therapeutic approaches are being explored to antagonize such soluble inhibitors, a particularly relevant approach is to identify other inhibitory checkpoints expressed by T or NK cells that, when blocked, may unleash the antitumor effector activity of these cells and possibly favor their proliferation.

Most promising progress towards this goal has recently been reported by André et al.,15 who showed that NKG2A may represent an important checkpoint in tumor therapy. Indeed, NKG2A targeting, alone or in combination with other checkpoint inhibitors or with mAbs specific for tumor antigens, led to impressive results both in preclinical and preliminary clinical studies in highly aggressive human tumors (Fig. 1).

Fig. 1.

Fig. 1

Effects of blocking NKG2A alone or in combination with other therapeutic mAbs disrupting the PD-1/PD-L1 axis or directed to tumor-specific antigens. (1) Cytolytic NK cells expressing the NKG2A inhibitory receptor acquire the ability to kill HLA-E+ tumor cells upon mAb-mediated masking of NKG2A. (2) NKG2A+ NK cells that also express PD1 are rescued from their anergic state and kill HLA-E+ PD-L1+ tumor cells upon disruption of the two inhibitory pathways with mAbs specific for NKG2A and PD-L1 (or PD-1), respectively. (3) The therapeutic activity of mAbs directed to tumor-specific antigens (in this case EGFR) is mainly mediated via CD16 (FcγRIII) expressed by highly mature cytolytic NK cells. However, in the case of HLA-E+ tumors, the coexpression of NKG2A may represent a major brake on NK-mediated function. Accordingly, masking of NKG2A may restore NK cell function and allow killing of tumor cells targeted by specific mAbs

As discussed above, specific mAb-mediated blocking of NKG2A and/or KIR can restore NK cytotoxicity, which is inhibited by the interaction with HLA-cl I. Importantly, both inhibitory receptors, while constitutively expressed by mature NK cells, have also been detected in cytolytic T lymphocytes.152,153 The expression of CD94/NKG2A can be induced by prolonged antigenic T-cell stimulation154 or exposure to TGF-β155, a cytokine released in the tumor microenvironment. Such ex novo NKG2A expression leads to the impairment of T-cell function.154,155 Thus, blocking of NKG2A was expected to unleash not only NK cells but also T lymphocytes with potential antitumor activity. This was indeed proven in elegant preclinical studies in suitable murine models. Subsequently, by studying human cancers, André et al.15 showed that HLA-E (the NKG2A ligand) is expressed in many highly aggressive tumors (including those of the pancreas, colon, lung, head and neck, stomach, and liver). Moreover, most tumor cells examined were HLA-E+. Analysis of lymphoid infiltrates showed that NKG2A was expressed in NK cells and in CD8+ T lymphocytes, which could also coexpress PD1. Experiments in vitro confirmed that IFN-γ production and the cytolytic activity of NK and T cells against HLA-E+ tumors were recovered, at least in part, by blocking anti-NKG2A mAbs, and, in the case of coexpression with PD-1, better functional restoration was documented in combination with therapeutic mAbs against PD1/PD-L1 (Fig. 1). This combined treatment could also restore T-cell proliferation, suggesting the possible generation of tumor-specific T-cell memory.

Another promising immunotherapy is based on the combined use of anti-NKG2A and tumor antigen-specific mAbs (e.g., an anti-EGFR mAb). The antitumor effects of these mAbs are, at least in part, dependent on the action of mature NK cells recognizing Ig-opsonized tumor cells via their CD16 to mediate ADCC (Fig. 1). In these in vitro experiments, the combined use of anti-NKG2A and anti-EGFR enhanced NK cell-mediated cytotoxicity against antibody-opsonized tumor cells. In this context, in preliminary clinical studies, the use of monalizumab (anti-NKG2A) combined with the anti-EGFR cetuximab in an ongoing phase II trial provided encouraging results in patients with squamous cell carcinoma of the head and neck (SCCHN). Notably, this study also emphasized the importance of harnessing NK cell antitumor activity, while immunotherapeutic strategies so far have been focused on potentiating T-cell responses. When unlocked from NKG2A blocking, NK cells may not only potentiate their natural cytotoxicity but also kill tumor cells via ADCC in the presence of tumor-specific therapeutic mAbs.

Role of NK cells in haploidentical HSCT for the cure of high-risk leukemia

HSCT is the life-saving therapy for acute leukemias with either adverse cytogenetic or molecular characteristics or poor response to chemotherapy or relapse. However, only 2 out of 3 patients requiring HSCT can find a suitable HLA-compatible donor. A novel transplantation approach has been attempted to rescue such patients for whom no other valuable therapeutic option are available. Thus, haploidentical HSCT, developed in the late 90s, is based on the infusion of “megadoses” of highly purified CD34+ haemopoietic precursors isolated from a parent or a sibling. Given the high degree of HLA incompatibility, deep T-cell depletion is mandatory to avoid severe, life-threatening graft versus host disease (GvHD). In this transplantation setting, NK cells of the donor may express KIRs that are mismatched with the HLA-cl I alleles of the patient. The first wave of lymphoid cells in patient PB occurs at ~2 weeks after transplant and is composed almost exclusively of NK cells. However, these NK cells are CD56bright and express NKG2A but not KIR. More mature KIR+NK cells are detectable only after additional 4-6 weeks. In the absence of T cells, NK cells have been shown to play a prominent role in the anti-leukemia effect.156 While NK cells per se may kill leukemia cells that express low levels of HLA-cl I molecules, the anti-leukemia effect has been primarily related to NK cell alloreactivity consequent to a KIR-HLA mismatch (in the donor vs recipient direction).156 Studies by our group revealed a direct correlation between the size of the alloreactive population and the degree of anti-leukemia cytotoxicity in vitro as well as the survival probability of the patient.157,158 Thus, clinical data in over 80 pediatric patients receiving haplo-HSCT indicated the treatment was particularly good for high-risk ALL with an ~70% 5-year survival probability in the presence of NK alloreactivity and ~40% in the absence of alloreactivity. The overall survival was ~60%. The outcome was less favorable for AML (~40% and ~20% with and without NK alloreactivity, respectively, and 30% overall survival). Although all these patients would have died in the absence of HSCT, the overall survival was not satisfactory, particularly for AML patients. Thus, a novel transplantation setting has been applied in an attempt to fill the gap between the time of HSCT and the generation of mature KIR+ alloreactive NK cells. Indeed, most of the deaths occurred in the first weeks after HSCT due to leukemia relapse or infections. This approach is based on the selective depletion of T cells expressing TCRαβ, which are responsible for GvHD, and B cells (to avoid B cell malignancies in immunocompromised patients). Thus, the infused mononuclear cells contained, in addition to CD34+ progenitors, mature KIR+ (possibly alloreactive) NK cells and TCRγδ T cells,159 plus different myeloid cells. Notably, not only NK cells but also TCRγδ T cells can kill leukemia blasts.160 The prompt availability of effector cells capable of attacking leukemia and exerting control of viral reactivation greatly improved the clinical outcome. Indeed, the overall survival probability was ~70% for AML and over 70% for ALL. Altogether, these data clearly indicate that NK cells exert strong anti-leukemia activity. The ability of CD34+ cells from different sources to give rise to ILC3s, including NCRnegILC3s with LTi-like activity, which contribute to tissue repair and regeneration of lymphoid tissues damaged by chemo/radiotherapy during conditioning, should also be carefully evaluated.161

Concluding remarks

A large body of experimental evidence, particularly from preclinical studies of both in vitro and in vivo animal models, indicates that NK cells play relevant roles in antitumor defenses. This is due to their direct cytolytic activity against tumor cells and their ability to produce cytokines that shape effective (Th1) downstream adaptive responses. However, different mechanisms occurring primarily in the tumor microenvironment may greatly impair NK cell function by compromising their antitumor activity, and they may even play an unwanted role in tumor promotion. This may occur when the NK cells present at the tumor site are few and/or their function is compromised. The result may be NK-mediated tumor editing (i.e., selection of the most resistant tumor cells)162 or the induction of epithelial/mesenchymal transition (that favors tumor spread and metastasis).163 In this context, any therapeutic approach that may favor/reconstitute NK cell function and expansion may be useful in tumor therapy. Thus, mAb-mediated masking of HLA-cl I inhibitory receptors may allow the direct attack of NK cells towards HLA-cl I+ tumor cells. Importantly, our group demonstrated that such inhibitory receptors, particularly NKG2A, also can be expressed on CD8+ CTLs following antigen- or cytokine-induced proliferation or exposure to TGF-β.152,154,155 These data offered clues to interpret recent studies by Andrè et al.15 showing the efficacy of blocking NKG2A in tumor therapy. On the other hand, the recent finding that not only T but also NK cells may express PD-1 revealed a new perspective, especially for the therapy of HLA-cl I-negative tumors (invisible to T lymphocytes).

It should be underscored that the major therapeutic success of haploidentical HSCT in the cure of high-risk leukemia is mostly related to NK cell activity. Great expectations for the therapy of both hematologic malignancies and solid tumors are based on the use of NK cells expressing CARs specific for tumor antigens. These CAR-NK cells may complement/substitute CAR-T lymphocytes in view of their potent cytolytic activity and their particular homing capacity.

Acknowledgements

This study is dedicated to Alessandro Moretta, who sadly passed away on February 17, 2018. His seminal discoveries of inhibitory and activating NK receptors made it possible to understand how NK cells function. Many of the issues described in this review are based on his pioneering studies. We greatly miss his scientific insight and even more his humanity and smile. Supported by grants awarded by Associazione Italiana per la Ricerca sul Cancro (AIRC)-Special Program Metastatic disease: the key unmet need in oncology 5 per mille 2018 Id. 21147 (S.S. and L.M.), AIRC IG2017 Id. 20312 (S.S.), AIRC IG2017 Id.19920 (L.M.), RC-2018 OPBG (P.V. and L.M.); 5 × 1000 Italian Ministry of Health 2015 (M.C.M); and Ministero della Salute RF-2013, GR-2013-02356568 (P.V.).

Competing interests

The authors declare no competing interests.

References

  • 1.Kiessling R, et al. Killer cells: a functional comparison between natural, immune T-cell and antibody-dependent in vitro systems. J. Exp. Med. 1976;143:772–780. doi: 10.1084/jem.143.4.772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Herberman RB, et al. Fc receptors on mouse effector cells mediating natural cytotoxicity against tumor cells. J. Immunol. 1977;119:322–326. [PubMed] [Google Scholar]
  • 3.Karre K, Ljunggren HG, Piontek G, Kiessling R. Selective rejection of H-2-deficient lymphoma variants suggests alternative immune defence strategy. Nature. 1986;319:675–678. doi: 10.1038/319675a0. [DOI] [PubMed] [Google Scholar]
  • 4.Ljunggren HG, Karre K. In search of the ‘missing self’: MHC molecules and NK cell recognition. Immunol. Today. 1990;11:237–244. doi: 10.1016/0167-5699(90)90097-s. [DOI] [PubMed] [Google Scholar]
  • 5.Moretta A, et al. Receptors for HLA class-I molecules in human natural killer cells. Annu. Rev. Immunol. 1996;14:619–648. doi: 10.1146/annurev.immunol.14.1.619. [DOI] [PubMed] [Google Scholar]
  • 6.Moretta A, Pantaleo G, Moretta L, Cerottini JC, Mingari MC. Direct demonstration of the clonogenic potential of every human peripheral blood T cell. Clonal analysis of HLA-DR expression and cytolytic activity. J. Exp. Med. 1983;157:743–754. doi: 10.1084/jem.157.2.743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Moretta A, Pantaleo G, Moretta L, Mingari MC, Cerottini JC. Quantitative assessment of the pool size and subset distribution of cytolytic T lymphocytes within human resting or alloactivated peripheral blood T cell populations. J. Exp. Med. 1983;158:571–585. doi: 10.1084/jem.158.2.571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Moretta A, et al. A novel surface antigen expressed by a subset of human CD3- CD16+natural killer cells. Role in cell activation and regulation of cytolytic function. J. Exp. Med. 1990;171:695–714. doi: 10.1084/jem.171.3.695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Moretta A, et al. Identification of four subsets of human CD3-CD16+natural killer (NK) cells by the expression of clonally distributed functional surface molecules: correlation between subset assignment of NK clones and ability to mediate specific alloantigen recognition. J. Exp. Med. 1990;172:1589–1598. doi: 10.1084/jem.172.6.1589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Moretta A, et al. P58 molecules as putative receptors for major histocompatibility complex (MHC) class I molecules in human natural killer (NK) cells. Anti-p58 antibodies reconstitute lysis of MHC class I-protected cells in NK clones displaying different specificities. J. Exp. Med. 1993;178:597–604. doi: 10.1084/jem.178.2.597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Ciccone E, et al. Evidence of a natural killer (NK) cell repertoire for (allo) antigen recognition: definition of five distinct NK-determined allospecificities in humans. J. Exp. Med. 1992;175:709–718. doi: 10.1084/jem.175.3.709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Colonna M, Samaridis J. Cloning of immunoglobulin-superfamily members associated with HLA-C and HLA-B recognition by human natural killer cells. Science. 1995;268:405–408. doi: 10.1126/science.7716543. [DOI] [PubMed] [Google Scholar]
  • 13.Wagtmann N, et al. Molecular clones of the p58 NK cell receptor reveal immunoglobulin-related molecules with diversity in both the extra- and intracellular domains. Immunity. 1995;2:439–449. doi: 10.1016/1074-7613(95)90025-x. [DOI] [PubMed] [Google Scholar]
  • 14.Braud VM, et al. HLA-E binds to natural killer cell receptors CD94/NKG2A, B and C. Nature. 1998;391:795–799. doi: 10.1038/35869. [DOI] [PubMed] [Google Scholar]
  • 15.Andre P, 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 e1713. doi: 10.1016/j.cell.2018.10.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Mingari, M. C., Pietra, G. & Moretta, L. Immune checkpoint inhibitors: Anti-NKG2A antibodies on board. Trends Immunol. 40, 83–85 (2019). [DOI] [PubMed]
  • 17.Anfossi N, et al. Human NK cell education by inhibitory receptors for MHC class I. Immunity. 2006;25:331–342. doi: 10.1016/j.immuni.2006.06.013. [DOI] [PubMed] [Google Scholar]
  • 18.Kim S, et al. Licensing of natural killer cells by host major histocompatibility complex class I molecules. Nature. 2005;436:709–713. doi: 10.1038/nature03847. [DOI] [PubMed] [Google Scholar]
  • 19.Joncker NT, Fernandez NC, Treiner E, Vivier E, Raulet DH. NK cell responsiveness is tuned commensurate with the number of inhibitory receptors for self-MHC class I: the rheostat model. J. Immunol. 2009;182:4572–4580. doi: 10.4049/jimmunol.0803900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Sivori S, et al. p46, a novel natural killer cell-specific surface molecule that mediates cell activation. J. Exp. Med. 1997;186:1129–1136. doi: 10.1084/jem.186.7.1129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Pessino A, et al. Molecular cloning of NKp46: a novel member of the immunoglobulin superfamily involved in triggering of natural cytotoxicity. J. Exp. Med. 1998;188:953–960. doi: 10.1084/jem.188.5.953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Sivori S, et al. NKp46 is the major triggering receptor involved in the natural cytotoxicity of fresh or cultured human NK cells. Correlation between surface density of NKp46 and natural cytotoxicity against autologous, allogeneic or xenogeneic target cells. Eur. J. Immunol. 1999;29:1656–1666. doi: 10.1002/(SICI)1521-4141(199905)29:05<1656::AID-IMMU1656>3.0.CO;2-1. [DOI] [PubMed] [Google Scholar]
  • 23.Vitale M, et al. 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. doi: 10.1084/jem.187.12.2065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Pende D, 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. doi: 10.1084/jem.190.10.1505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Moretta A, et al. Activating receptors and coreceptors involved in human natural killer cell-mediated cytolysis. Annu. Rev. Immunol. 2001;19:197–223. doi: 10.1146/annurev.immunol.19.1.197. [DOI] [PubMed] [Google Scholar]
  • 26.Pazina T, Shemesh A, Brusilovsky M, Porgador A, Campbell KS. Regulation of the functions of natural cytotoxicity receptors by interactions with diverse ligands and alterations in splice variant expression. Front. Immunol. 2017;8:369. doi: 10.3389/fimmu.2017.00369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Raffaghello L, et al. Downregulation and/or release of NKG2D ligands as immune evasion strategy of human neuroblastoma. Neoplasia. 2004;6:558–568. doi: 10.1593/neo.04316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Moretta L, et al. Different checkpoints in human NK-cell activation. Trends Immunol. 2004;25:670–676. doi: 10.1016/j.it.2004.09.008. [DOI] [PubMed] [Google Scholar]
  • 29.Caligiuri MA. Human natural killer cells. Blood. 2008;112:461–469. doi: 10.1182/blood-2007-09-077438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Moretta L. Dissecting CD56dim human NK cells. Blood. 2010;116:3689–3691. doi: 10.1182/blood-2010-09-303057. [DOI] [PubMed] [Google Scholar]
  • 31.Hammer Q, et al. Peptide-specific recognition of human cytomegalovirus strains controls adaptive natural killer cells. Nat. Immunol. 2018;19:453–463. doi: 10.1038/s41590-018-0082-6. [DOI] [PubMed] [Google Scholar]
  • 32.Della Chiesa M, Sivori S, Carlomagno S, Moretta L, Moretta A. Activating KIRs and NKG2C in viral infections: Toward NK cell memory? Front. Immunol. 2015;6:573. doi: 10.3389/fimmu.2015.00573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Artis D, Spits H. The biology of innate lymphoid cells. Nature. 2015;517:293–301. doi: 10.1038/nature14189. [DOI] [PubMed] [Google Scholar]
  • 34.Vivier E, van de Pavert SA, Cooper MD, Belz GT. The evolution of innate lymphoid cells. Nat. Immunol. 2016;17:790–794. doi: 10.1038/ni.3459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Luetke-Eversloh M, Killig M, Romagnani C. Signatures of human NK cell development and terminal differentiation. Front. Immunol. 2013;4:499. doi: 10.3389/fimmu.2013.00499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Montaldo E, et al. Human innate lymphoid cells. Immunol. Lett. 2016;179:2–8. doi: 10.1016/j.imlet.2016.01.007. [DOI] [PubMed] [Google Scholar]
  • 37.Guia S, Fenis A, Vivier E, Narni-Mancinelli E. Activating and inhibitory receptors expressed on innate lymphoid cells. Semin. Immunopathol. 2018;40:331–341. doi: 10.1007/s00281-018-0685-x. [DOI] [PubMed] [Google Scholar]
  • 38.Soriani A, Stabile H, Gismondi A, Santoni A, Bernardini G. Chemokine regulation of innate lymphoid cell tissue distribution and function. Cytokine Growth Factor Rev. 2018;42:47–55. doi: 10.1016/j.cytogfr.2018.02.003. [DOI] [PubMed] [Google Scholar]
  • 39.Mingari MC, et al. Interleukin-15-induced maturation of human natural killer cells from early thymic precursors: selective expression of CD94/NKG2-A as the only HLA class I-specific inhibitory receptor. Eur. J. Immunol. 1997;27:1374–1380. doi: 10.1002/eji.1830270612. [DOI] [PubMed] [Google Scholar]
  • 40.Yu J, Freud AG, Caligiuri MA. Location and cellular stages of natural killer cell development. Trends Immunol. 2013;34:573–582. doi: 10.1016/j.it.2013.07.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Montaldo E, et al. Human RORgammat(+)CD34(+) cells are lineage-specified progenitors of group 3 RORgammat(+) innate lymphoid cells. Immunity. 2014;41:988–1000. doi: 10.1016/j.immuni.2014.11.010. [DOI] [PubMed] [Google Scholar]
  • 42.Vacca P, et al. CD34+hematopoietic precursors are present in human decidua and differentiate into natural killer cells upon interaction with stromal cells. Proc. Natl Acad. Sci. USA. 2011;108:2402–2407. doi: 10.1073/pnas.1016257108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Poggi A, et al. Extrathymic differentiation of T lymphocytes and natural killer cells from human embryonic liver precursors. Proc. Natl Acad. Sci. USA. 1993;90:4465–4469. doi: 10.1073/pnas.90.10.4465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Vacca P, Moretta L, Moretta A, Mingari MC. Origin, phenotype and function of human natural killer cells in pregnancy. Trends Immunol. 2011;32:517–523. doi: 10.1016/j.it.2011.06.013. [DOI] [PubMed] [Google Scholar]
  • 45.Parham P, Guethlein LA. Genetics of natural killer cells in human health, disease, and survival. Annu. Rev. Immunol. 2018;36:519–548. doi: 10.1146/annurev-immunol-042617-053149. [DOI] [PubMed] [Google Scholar]
  • 46.Moretta A, et al. Existence of both inhibitory (p58) and activatory (p50) receptors for HLA-C molecules in human natural killer cells. J. Exp. Med. 1995;182:875–884. doi: 10.1084/jem.182.3.875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Biassoni R, et al. The human leukocyte antigen (HLA)-C-specific “activatory” or “inhibitory” natural killer cell receptors display highly homologous extracellular domains but differ in their transmembrane and intracytoplasmic portions. J. Exp. Med. 1996;183:645–650. doi: 10.1084/jem.183.2.645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Chewning JH, Gudme CN, Hsu KC, Selvakumar A, Dupont B. KIR2DS1-positive NK cells mediate alloresponse against the C2 HLA-KIR ligand group in vitro. J. Immunol. 2007;179:854–868. doi: 10.4049/jimmunol.179.2.854. [DOI] [PubMed] [Google Scholar]
  • 49.Stewart CA, et al. Recognition of peptide-MHC class I complexes by activating killer immunoglobulin-like receptors. Proc. Natl Acad. Sci. USA. 2005;102:13224–13229. doi: 10.1073/pnas.0503594102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Graef T, et al. KIR2DS4 is a product of gene conversion with KIR3DL2 that introduced specificity for HLA-A*11 while diminishing avidity for HLA-C. J. Exp. Med. 2009;206:2557–2572. doi: 10.1084/jem.20091010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Burian A, et al. HLA-F and MHC-I Open Conformers Bind Natural Killer Cell Ig-Like Receptor KIR3DS1. PLoS ONE. 2016;11:e0163297. doi: 10.1371/journal.pone.0163297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Garcia-Beltran WF, et al. Open conformers of HLA-F are high-affinity ligands of the activating NK-cell receptor KIR3DS1. Nat. Immunol. 2016;17:1067–1074. doi: 10.1038/ni.3513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Carlomagno S, et al. KIR3DS1-mediated recognition of HLA-*B51: modulation of KIR3DS1 responsiveness by self HLA-B allotypes and effect on NK cell licensing. Front. Immunol. 2017;8:581. doi: 10.3389/fimmu.2017.00581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Naiyer Mohammed M., Cassidy Sorcha A., Magri Andrea, Cowton Vanessa, Chen Kevin, Mansour Salah, Kranidioti Hariklia, Mbirbindi Berenice, Rettman Pauline, Harris Scott, Fanning Liam J., Mulder Arend, Claas Franz H. J., Davidson Andrew D., Patel Arvind H., Purbhoo Marco A., Khakoo Salim I. KIR2DS2 recognizes conserved peptides derived from viral helicases in the context of HLA-C. Science Immunology. 2017;2(15):eaal5296. doi: 10.1126/sciimmunol.aal5296. [DOI] [PubMed] [Google Scholar]
  • 55.Parham P. MHC class I molecules and KIRs in human history, health and survival. Nat. Rev. Immunol. 2005;5:201–214. doi: 10.1038/nri1570. [DOI] [PubMed] [Google Scholar]
  • 56.Vitale M, et al. The leukocyte Ig-like receptor (LIR)-1 for the cytomegalovirus UL18 protein displays a broad specificity for different HLA class I alleles: analysis of LIR-1+NK cell clones. Int. Immunol. 1999;11:29–35. doi: 10.1093/intimm/11.1.29. [DOI] [PubMed] [Google Scholar]
  • 57.Colonna M, et al. A common inhibitory receptor for major histocompatibility complex class I molecules on human lymphoid and myelomonocytic cells. J. Exp. Med. 1997;186:1809–1818. doi: 10.1084/jem.186.11.1809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Cosman D, et al. A novel immunoglobulin superfamily receptor for cellular and viral MHC class I molecules. Immunity. 1997;7:273–282. doi: 10.1016/s1074-7613(00)80529-4. [DOI] [PubMed] [Google Scholar]
  • 59.Ponte M, et al. Inhibitory receptors sensing HLA-G1 molecules in pregnancy: decidua-associated natural killer cells express LIR-1 and CD94/NKG2A and acquire p49, an HLA-G1-specific receptor. Proc. Natl Acad. Sci. USA. 1999;96:5674–5679. doi: 10.1073/pnas.96.10.5674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Muccio L, et al. Analysis of memory-like natural killer cells in human cytomegalovirus-infected children undergoing alphabeta+T and B cell-depleted hematopoietic stem cell transplantation for hematological malignancies. Haematologica. 2016;101:371–381. doi: 10.3324/haematol.2015.134155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Vales-Gomez M, Reyburn HT, Erskine RA, Lopez-Botet M, Strominger JL. Kinetics and peptide dependency of the binding of the inhibitory NK receptor CD94/NKG2-A and the activating receptor CD94/NKG2-C to HLA-E. EMBO J. 1999;18:4250–4260. doi: 10.1093/emboj/18.15.4250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Heatley SL, et al. Polymorphism in human cytomegalovirus UL40 impacts on recognition of human leukocyte antigen-E (HLA-E) by natural killer cells. J. Biol. Chem. 2013;288:8679–8690. doi: 10.1074/jbc.M112.409672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Della Chiesa M, et al. Features of Memory-Like and PD-1(+) Human NK Cell Subsets. Front. Immunol. 2016;7:351. doi: 10.3389/fimmu.2016.00351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Guma M, et al. Expansion of CD94/NKG2C+NK cells in response to human cytomegalovirus-infected fibroblasts. Blood. 2006;107:3624–3631. doi: 10.1182/blood-2005-09-3682. [DOI] [PubMed] [Google Scholar]
  • 65.Lopez-Verges S, et al. Expansion of a unique CD57+NKG2Chi natural killer cell subset during acute human cytomegalovirus infection. Proc. Natl Acad. Sci. USA. 2011;108:14725–14732. doi: 10.1073/pnas.1110900108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Tomasello E, et al. Gene structure, expression pattern, and biological activity of mouse killer cell activating receptor-associated protein (KARAP)/DAP-12. J. Biol. Chem. 1998;273:34115–34119. doi: 10.1074/jbc.273.51.34115. [DOI] [PubMed] [Google Scholar]
  • 67.Campbell KS, Yusa S, Kikuchi-Maki A, Catina TL. NKp44 triggers NK cell activation through DAP12 association that is not influenced by a putative cytoplasmic inhibitory sequence. J. Immunol. 2004;172:899–906. doi: 10.4049/jimmunol.172.2.899. [DOI] [PubMed] [Google Scholar]
  • 68.Muccio L, et al. Late development of Fc epsilon R gamma(neg) adaptive natural killer cells upon human cytomegalovirus reactivation in umbilical cord blood transplantation recipients. Front. Immunol. 2018;9:1050. doi: 10.3389/fimmu.2018.01050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Arnon TI, Markel G, Mandelboim O. Tumor and viral recognition by natural killer cells receptors. Semin. Cancer Biol. 2006;16:348–358. doi: 10.1016/j.semcancer.2006.07.005. [DOI] [PubMed] [Google Scholar]
  • 70.Arnon TI, et al. Inhibition of the NKp30 activating receptor by pp65 of human cytomegalovirus. Nat. Immunol. 2005;6:515–523. doi: 10.1038/ni1190. [DOI] [PubMed] [Google Scholar]
  • 71.Pogge von Strandmann E, et al. Human leukocyte antigen-B-associated transcript 3 is released from tumor cells and engages the NKp30 receptor on natural killer cells. Immunity. 2007;27:965–974. doi: 10.1016/j.immuni.2007.10.010. [DOI] [PubMed] [Google Scholar]
  • 72.Reiners KS, et al. Soluble ligands for NK cell receptors promote evasion of chronic lymphocytic leukemia cells from NK cell anti-tumor activity. Blood. 2013;121:3658–3665. doi: 10.1182/blood-2013-01-476606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Baychelier F, et al. Identification of a cellular ligand for the natural cytotoxicity receptor NKp44. Blood. 2013;122:2935–2942. doi: 10.1182/blood-2013-03-489054. [DOI] [PubMed] [Google Scholar]
  • 74.Rosental B, et al. Proliferating cell nuclear antigen is a novel inhibitory ligand for the natural cytotoxicity receptor NKp44. J. Immunol. 2011;187:5693–5702. doi: 10.4049/jimmunol.1102267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Brandt CS, et al. The B7 family member B7-H6 is a tumor cell ligand for the activating natural killer cell receptor NKp30 in humans. J. Exp. Med. 2009;206:1495–1503. doi: 10.1084/jem.20090681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Textor S, et al. The proto-oncogene Myc drives expression of the NK cell-activating NKp30 ligand B7-H6 in tumor cells. Oncoimmunology. 2016;5:e1116674. doi: 10.1080/2162402X.2015.1116674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Matta J, et al. Induction of B7-H6, a ligand for the natural killer cell-activating receptor NKp30, in inflammatory conditions. Blood. 2013;122:394–404. doi: 10.1182/blood-2013-01-481705. [DOI] [PubMed] [Google Scholar]
  • 78.Narni-Mancinelli Emilie, Gauthier Laurent, Baratin Myriam, Guia Sophie, Fenis Aurore, Deghmane Ala-Eddine, Rossi Benjamin, Fourquet Patrick, Escalière Bertrand, Kerdiles Yann M., Ugolini Sophie, Taha Muhamed-Kheir, Vivier Eric. Complement factor P is a ligand for the natural killer cell–activating receptor NKp46. Science Immunology. 2017;2(10):eaam9628. doi: 10.1126/sciimmunol.aam9628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Gaggero S, et al. Nidogen-1 is a novel extracellular ligand for the NKp44 activating receptor. Oncoimmunology. 2018;7:e1470730. doi: 10.1080/2162402X.2018.1470730. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Schlecker E, 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. doi: 10.1158/0008-5472.CAN-13-3017. [DOI] [PubMed] [Google Scholar]
  • 81.Pesce S, et al. B7-H6-mediated downregulation of NKp30 in NK cells contributes to ovarian carcinoma immune escape. Oncoimmunology. 2015;4:e1001224. doi: 10.1080/2162402X.2014.1001224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.De Maria A, et al. The impaired NK cell cytolytic function in viremic HIV-1 infection is associated with a reduced surface expression of natural cytotoxicity receptors (NKp46, NKp30 and NKp44) Eur. J. Immunol. 2003;33:2410–2418. doi: 10.1002/eji.200324141. [DOI] [PubMed] [Google Scholar]
  • 83.Bauer S, et al. Activation of NK cells and T cells by NKG2D, a receptor for stress-inducible MICA. Science. 1999;285:727–729. [PubMed] [Google Scholar]
  • 84.Raulet DH, Gasser S, Gowen BG, Deng W, Jung H. Regulation of ligands for the NKG2D activating receptor. Annu. Rev. Immunol. 2013;31:413–441. doi: 10.1146/annurev-immunol-032712-095951. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Lanier LL. NKG2D receptor and its ligands in host defense. Cancer Immunol. Res. 2015;3:575–582. doi: 10.1158/2326-6066.CIR-15-0098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Sivori S, et al. 2B4 functions as a co-receptor in human NK cell activation. Eur. J. Immunol. 2000;30:787–793. doi: 10.1002/1521-4141(200003)30:3<787::AID-IMMU787>3.0.CO;2-I. [DOI] [PubMed] [Google Scholar]
  • 87.Bottino C, et al. NTB-A [correction of GNTB-A], a novel SH2D1A-associated surface molecule contributing to the inability of natural killer cells to kill Epstein-Barr virus-infected B cells in X-linked lymphoproliferative disease. J. Exp. Med. 2001;194:235–246. doi: 10.1084/jem.194.3.235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Shibuya A, et al. DNAM-1, a novel adhesion molecule involved in the cytolytic function of T lymphocytes. Immunity. 1996;4:573–581. doi: 10.1016/s1074-7613(00)70060-4. [DOI] [PubMed] [Google Scholar]
  • 89.Marcenaro E, et al. CD59 is physically and functionally associated with natural cytotoxicity receptors and activates human NK cell-mediated cytotoxicity. Eur. J. Immunol. 2003;33:3367–3376. doi: 10.1002/eji.200324425. [DOI] [PubMed] [Google Scholar]
  • 90.Vitale M, et al. Identification of NKp80, a novel triggering molecule expressed by human NK cells. Eur. J. Immunol. 2001;31:233–242. doi: 10.1002/1521-4141(200101)31:1<233::AID-IMMU233>3.0.CO;2-4. [DOI] [PubMed] [Google Scholar]
  • 91.Sivori S, et al. CpG and double-stranded RNA trigger human NK cells by Toll-like receptors: induction of cytokine release and cytotoxicity against tumors and dendritic cells. Proc. Natl Acad. Sci. USA. 2004;101:10116–10121. doi: 10.1073/pnas.0403744101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Sivori S, Carlomagno S, Moretta L, Moretta A. Comparison of different CpG oligodeoxynucleotide classes for their capability to stimulate human NK cells. Eur. J. Immunol. 2006;36:961–967. doi: 10.1002/eji.200535781. [DOI] [PubMed] [Google Scholar]
  • 93.Sivori S, et al. Heterogeneity of TLR3 mRNA transcripts and responsiveness to poly (I:C) in human NK cells derived from different donors. Int. Immunol. 2007;19:1341–1348. doi: 10.1093/intimm/dxm105. [DOI] [PubMed] [Google Scholar]
  • 94.Sivori S, et al. A novel KIR-associated function: evidence that CpG DNA uptake and shuttling to early endosomes is mediated by KIR3DL2. Blood. 2010;116:1637–1647. doi: 10.1182/blood-2009-12-256586. [DOI] [PubMed] [Google Scholar]
  • 95.Ochoa MC, et al. Antibody-dependent cell cytotoxicity: immunotherapy strategies enhancing effector NK cells. Immunol. Cell Biol. 2017;95:347–355. doi: 10.1038/icb.2017.6. [DOI] [PubMed] [Google Scholar]
  • 96.Vivier E, et al. Innate or adaptive immunity? The example of natural killer cells. Science. 2011;331:44–49. doi: 10.1126/science.1198687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Chiossone L, Vienne M, Kerdiles YM, Vivier E. Natural killer cell immunotherapies against cancer: checkpoint inhibitors and more. Semin. Immunol. 2017;31:55–63. doi: 10.1016/j.smim.2017.08.003. [DOI] [PubMed] [Google Scholar]
  • 98.Munari E, et al. PD-L1 expression comparison between primary and relapsed non-small cell lung carcinoma using whole sections and clone SP263. Oncotarget. 2018;9:30465–30471. doi: 10.18632/oncotarget.25770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Munari Enrico, Zamboni Giuseppe, Lunardi Gianluigi, Marchionni Luigi, Marconi Marcella, Sommaggio Marco, Brunelli Matteo, Martignoni Guido, Netto George J., Hoque Mohammad O., Moretta Francesca, Mingari Maria Cristina, Pegoraro Maria Cristina, Inno Alessandro, Paiano Simona, Terzi Alberto, Cavazza Alberto, Rossi Giulio, Mariotti Francesca Romana, Vacca Paola, Moretta Lorenzo, Bogina Giuseppe. PD-L1 Expression Heterogeneity in Non–Small Cell Lung Cancer: Defining Criteria for Harmonization between Biopsy Specimens and Whole Sections. Journal of Thoracic Oncology. 2018;13(8):1113–1120. doi: 10.1016/j.jtho.2018.04.017. [DOI] [PubMed] [Google Scholar]
  • 100.Ishida Y, Agata Y, Shibahara K, Honjo T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J. 1992;11:3887–3895. doi: 10.1002/j.1460-2075.1992.tb05481.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Nishimura H, Nose M, Hiai H, Minato N, Honjo T. Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor. Immunity. 1999;11:141–151. doi: 10.1016/s1074-7613(00)80089-8. [DOI] [PubMed] [Google Scholar]
  • 102.Okazaki T, Chikuma S, Iwai Y, Fagarasan S, Honjo T. A rheostat for immune responses: the unique properties of PD-1 and their advantages for clinical application. Nat. Immunol. 2013;14:1212–1218. doi: 10.1038/ni.2762. [DOI] [PubMed] [Google Scholar]
  • 103.Huang AC, et al. T-cell invigoration to tumour burden ratio associated with anti-PD-1 response. Nature. 2017;545:60–65. doi: 10.1038/nature22079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Pesce S, 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;139:335–346. doi: 10.1016/j.jaci.2016.04.025. [DOI] [PubMed] [Google Scholar]
  • 105.Beldi-Ferchiou A, et al. PD-1 mediates functional exhaustion of activated NK cells in patients with Kaposi sarcoma. Oncotarget. 2016;7:72961–72977. doi: 10.18632/oncotarget.12150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Iraolagoitia XL, et al. NK cells restrain spontaneous antitumor CD8+T cell priming through PD-1/PD-L1 interactions with dendritic cells. J. Immunol. 2016;197:953–961. doi: 10.4049/jimmunol.1502291. [DOI] [PubMed] [Google Scholar]
  • 107.Vari F, 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;131:1809–1819. doi: 10.1182/blood-2017-07-796342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Quatrini L, 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;19:954–962. doi: 10.1038/s41590-018-0185-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Hsu J, et al. Contribution of NK cells to immunotherapy mediated by PD-1/PD-L1 blockade. J. Clin. Invest. 2018;128:4654–4668. doi: 10.1172/JCI99317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Bottino C, 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. doi: 10.1084/jem.20030788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Dougall WC, Kurtulus S, Smyth MJ, Anderson AC. TIGIT and CD96: new checkpoint receptor targets for cancer immunotherapy. Immunol. Rev. 2017;276:112–120. doi: 10.1111/imr.12518. [DOI] [PubMed] [Google Scholar]
  • 112.Zhou XM, et al. Intrinsic expression of immune checkpoint molecule TIGIT could help tumor growth in vivo by suppressing the function of NK and CD8(+) T Cells. Front. Immunol. 2018;9:2821. doi: 10.3389/fimmu.2018.02821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Zhang Q, et al. Blockade of the checkpoint receptor TIGIT prevents NK cell exhaustion and elicits potent anti-tumor immunity. Nat. Immunol. 2018;19:723–732. doi: 10.1038/s41590-018-0132-0. [DOI] [PubMed] [Google Scholar]
  • 114.Hung AL, et al. TIGIT and PD-1 dual checkpoint blockade enhances antitumor immunity and survival in GBM. Oncoimmunology. 2018;7:e1466769. doi: 10.1080/2162402X.2018.1466769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Huntington ND, et al. NK cell maturation and peripheral homeostasis is associated with KLRG1 up-regulation. J. Immunol. 2007;178:4764–4770. doi: 10.4049/jimmunol.178.8.4764. [DOI] [PubMed] [Google Scholar]
  • 116.Huang Y, et al. IL-25-responsive, lineage-negative KLRG1(hi) cells are multipotential ‘inflammatory’ type 2 innate lymphoid cells. Nat. Immunol. 2015;16:161–169. doi: 10.1038/ni.3078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Ito M, et al. Killer cell lectin-like receptor G1 binds three members of the classical cadherin family to inhibit NK cell cytotoxicity. J. Exp. Med. 2006;203:289–295. doi: 10.1084/jem.20051986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Salimi M, et al. A role for IL-25 and IL-33-driven type-2 innate lymphoid cells in atopic dermatitis. J. Exp. Med. 2013;210:2939–2950. doi: 10.1084/jem.20130351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Simoni Y, et al. Human innate lymphoid cell subsets possess tissue-type based heterogeneity in phenotype and frequency. Immunity. 2017;46:148–161. doi: 10.1016/j.immuni.2016.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Carrega P, et al. NCR(+)ILC3 concentrate in human lung cancer and associate with intratumoral lymphoid structures. Nat. Commun. 2015;6:8280. doi: 10.1038/ncomms9280. [DOI] [PubMed] [Google Scholar]
  • 121.Baixeras E, et al. Characterization of the lymphocyte activation gene 3-encoded protein. A new ligand for human leukocyte antigen class II antigens. J. Exp. Med. 1992;176:327–337. doi: 10.1084/jem.176.2.327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Lichtenegger FS, et al. Targeting LAG-3 and PD-1 to enhance T cell activation by antigen-presenting cells. Front. Immunol. 2018;9:385. doi: 10.3389/fimmu.2018.00385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Woo SR, et al. Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape. Cancer Res. 2012;72:917–927. doi: 10.1158/0008-5472.CAN-11-1620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Miyazaki T, Dierich A, Benoist C, Mathis D. LAG-3 is not responsible for selecting T helper cells in CD4-deficient mice. Int. Immunol. 1996;8:725–729. doi: 10.1093/intimm/8.5.725. [DOI] [PubMed] [Google Scholar]
  • 125.Wada J, Kanwar YS. Identification and characterization of galectin-9, a novel beta-galactoside-binding mammalian lectin. J. Biol. Chem. 1997;272:6078–6086. doi: 10.1074/jbc.272.9.6078. [DOI] [PubMed] [Google Scholar]
  • 126.DeKruyff RH, et al. T cell/transmembrane, Ig, and mucin-3 allelic variants differentially recognize phosphatidylserine and mediate phagocytosis of apoptotic cells. J. Immunol. 2010;184:1918–1930. doi: 10.4049/jimmunol.0903059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Chiba S, 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. doi: 10.1038/ni.2376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Huang YH, et al. CEACAM1 regulates TIM-3-mediated tolerance and exhaustion. Nature. 2015;517:386–390. doi: 10.1038/nature13848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Gao X, et al. TIM-3 expression characterizes regulatory T cells in tumor tissues and is associated with lung cancer progression. PLoS ONE. 2012;7:e30676. doi: 10.1371/journal.pone.0030676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Lu X, et al. Tumor antigen-specific CD8(+) T cells are negatively regulated by PD-1 and Tim-3 in human gastric cancer. Cell Immunol. 2017;313:43–51. doi: 10.1016/j.cellimm.2017.01.001. [DOI] [PubMed] [Google Scholar]
  • 131.Shayan G, et al. Adaptive resistance to anti-PD1 therapy by Tim-3 upregulation is mediated by the PI3K-Akt pathway in head and neck cancer. Oncoimmunology. 2017;6:e1261779. doi: 10.1080/2162402X.2016.1261779. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Fourcade J, et al. Upregulation of Tim-3 and PD-1 expression is associated with tumor antigen-specific CD8+T cell dysfunction in melanoma patients. J. Exp. Med. 2010;207:2175–2186. doi: 10.1084/jem.20100637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.da Silva IP, et al. Reversal of NK-cell exhaustion in advanced melanoma by Tim-3 blockade. Cancer Immunol. Res. 2014;2:410–422. doi: 10.1158/2326-6066.CIR-13-0171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Xu L, et al. Increased Tim-3 expression in peripheral NK cells predicts a poorer prognosis and Tim-3 blockade improves NK cell-mediated cytotoxicity in human lung adenocarcinoma. Int. Immunopharmacol. 2015;29:635–641. doi: 10.1016/j.intimp.2015.09.017. [DOI] [PubMed] [Google Scholar]
  • 135.Das M, Zhu C, Kuchroo VK. Tim-3 and its role in regulating anti-tumor immunity. Immunol. Rev. 2017;276:97–111. doi: 10.1111/imr.12520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Thomassen E, Renshaw BR, Sims JE. Identification and characterization of SIGIRR, a molecule representing a novel subtype of the IL-1R superfamily. Cytokine. 1999;11:389–399. doi: 10.1006/cyto.1998.0452. [DOI] [PubMed] [Google Scholar]
  • 137.Mantovani A, Locati M, Polentarutti N, Vecchi A, Garlanda C. Extracellular and intracellular decoys in the tuning of inflammatory cytokines and Toll-like receptors: the new entry TIR8/SIGIRR. J. Leukoc. Biol. 2004;75:738–742. doi: 10.1189/jlb.1003473. [DOI] [PubMed] [Google Scholar]
  • 138.Molgora M, et al. IL-1R8 is a checkpoint in NK cells regulating anti-tumour and anti-viral activity. Nature. 2017;551:110–114. doi: 10.1038/nature24293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer. 2012;12:252–264. doi: 10.1038/nrc3239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Gong J, Chehrazi-Raffle A, Reddi S, Salgia R. Development of PD-1 and PD-L1 inhibitors as a form of cancer immunotherapy: a comprehensive review of registration trials and future considerations. J. Immunother. Cancer. 2018;6:8. doi: 10.1186/s40425-018-0316-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Migden MR, et al. PD-1 blockade with cemiplimab in advanced cutaneous squamous-cell carcinoma. N. Engl. J. Med. 2018;379:341–351. doi: 10.1056/NEJMoa1805131. [DOI] [PubMed] [Google Scholar]
  • 142.Buttner R, et al. Programmed death-ligand 1 immunohistochemistry testing: A review of analytical assays and clinical implementation in non-small-cell lung cancer. J. Clin. Oncol.: Off. J. Am. Soc. Clin. Oncol. 2017;35:3867–3876. doi: 10.1200/JCO.2017.74.7642. [DOI] [PubMed] [Google Scholar]
  • 143.Munari, E. et al. PD-L1 Assays 22C3 and SP263 are not interchangeable in non-small cell lung cancer when considering clinically relevant cutoffs: An interclone evaluation by differently trained pathologists. Am. J. Surg. Pathol.42, 1384–1389 (2018). [DOI] [PubMed]
  • 144.Munari E, et al. PD-L1 expression heterogeneity in non-small cell lung cancer: evaluation of small biopsies reliability. Oncotarget. 2017;8:90123–90131. doi: 10.18632/oncotarget.21485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Munari Enrico, Zamboni Giuseppe, Sighele Giorgia, Marconi Marcella, Sommaggio Marco, Lunardi Gianluigi, Rossi Giulio, Cavazza Alberto, Moretta Francesca, Gilioli Eliana, Caliò Anna, Netto George J., Hoque Mohammad O., Martignoni Guido, Brunelli Matteo, Vacca Paola, Moretta Lorenzo, Bogina Giuseppe. Expression of programmed cell death ligand 1 in non–small cell lung cancer: Comparison between cytologic smears, core biopsies, and whole sections using the SP263 assay. Cancer Cytopathology. 2018;127(1):52–61. doi: 10.1002/cncy.22083. [DOI] [PubMed] [Google Scholar]
  • 146.Pietra G, et al. Human natural killer cells: news in the therapy of solid tumors and high-risk leukemias. Cancer Immunol. Immunother. 2016;65:465–476. doi: 10.1007/s00262-015-1744-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Kim N, Kim HS. Targeting checkpoint receptors and molecules for therapeutic modulation of natural killer cells. Front. Immunol. 2018;9:2041. doi: 10.3389/fimmu.2018.02041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Sivori S, et al. Inhibitory CD94 molecules identified by the Z199 monoclonal antibody recognize different HLA-class I molecules. Transplant. Proc. 1996;28:3199–3203. [PubMed] [Google Scholar]
  • 149.Pende D, et al. HLA-G recognition by human natural killer cells. Involvement of CD94 both as inhibitory and as activating receptor complex. Eur. J. Immunol. 1997;27:1875–1880. doi: 10.1002/eji.1830270809. [DOI] [PubMed] [Google Scholar]
  • 150.Morandi F, Rizzo R, Fainardi E, Rouas-Freiss N, Pistoia V. Recent advances in our understanding of HLA-G biology: Lessons from a wide spectrum of human diseases. J. Immunol. Res. 2016;2016:4326495. doi: 10.1155/2016/4326495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Vitale M, Cantoni C, Pietra G, Mingari MC, Moretta L. Effect of tumor cells and tumor microenvironment on NK-cell function. Eur. J. Immunol. 2014;44:1582–1592. doi: 10.1002/eji.201344272. [DOI] [PubMed] [Google Scholar]
  • 152.Mingari MC, et al. Cytolytic T lymphocytes displaying natural killer (NK)-like activity: expression of NK-related functional receptors for HLA class I molecules (p58 and CD94) and inhibitory effect on the TCR-mediated target cell lysis or lymphokine production. Int. Immunol. 1995;7:697–703. doi: 10.1093/intimm/7.4.697. [DOI] [PubMed] [Google Scholar]
  • 153.Mingari MC, et al. Human CD8 + T lymphocyte subsets that express HLA class I-specific inhibitory receptors represent oligoclonally or monoclonally expanded cell populations. Proc. Natl Acad. Sci. USA. 1996;93:12433–12438. doi: 10.1073/pnas.93.22.12433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Mingari MC, et al. HLA class I-specific inhibitory receptors in human T lymphocytes: interleukin 15-induced expression of CD94/NKG2A in superantigen- or alloantigen-activated CD8+T cells. Proc. Natl Acad. Sci. USA. 1998;95:1172–1177. doi: 10.1073/pnas.95.3.1172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Bertone S, et al. Transforming growth factor-beta-induced expression of CD94/NKG2A inhibitory receptors in human T lymphocytes. Eur. J. Immunol. 1999;29:23–29. doi: 10.1002/(SICI)1521-4141(199901)29:01<23::AID-IMMU23>3.0.CO;2-Y. [DOI] [PubMed] [Google Scholar]
  • 156.Ruggeri L, et al. Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants. Science. 2002;295:2097–2100. doi: 10.1126/science.1068440. [DOI] [PubMed] [Google Scholar]
  • 157.Pende D, et al. Anti-leukemia activity of alloreactive NK cells in KIR ligand-mismatched haploidentical HSCT for pediatric patients: evaluation of the functional role of activating KIR and redefinition of inhibitory KIR specificity. Blood. 2009;113:3119–3129. doi: 10.1182/blood-2008-06-164103. [DOI] [PubMed] [Google Scholar]
  • 158.Pende D, et al. Analysis of the receptor-ligand interactions in the natural killer-mediated lysis of freshly isolated myeloid or lymphoblastic leukemias: evidence for the involvement of the Poliovirus receptor (CD155) and Nectin-2 (CD112) Blood. 2005;105:2066–2073. doi: 10.1182/blood-2004-09-3548. [DOI] [PubMed] [Google Scholar]
  • 159.Pistoia V, et al. Human gammadelta T-Cells: From surface receptors to the therapy of high-risk leukemias. Front. Immunol. 2018;9:984. doi: 10.3389/fimmu.2018.00984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Locatelli F, et al. NK cells mediate a crucial graft-versus-leukemia effect in haploidentical-HSCT to Cure High-risk Acute Leukemia. Trends Immunol. 2018;39:577–590. doi: 10.1016/j.it.2018.04.009. [DOI] [PubMed] [Google Scholar]
  • 161.Moretta F, et al. The generation of human innate lymphoid cells is influenced by the source of hematopoietic stem cells and by the use of G-CSF. Eur. J. Immunol. 2016;46:1271–1278. doi: 10.1002/eji.201546079. [DOI] [PubMed] [Google Scholar]
  • 162.Balsamo M, 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. doi: 10.1002/eji.201142179. [DOI] [PubMed] [Google Scholar]
  • 163.Huergo-Zapico L, et al. NK-cell editing mediates epithelial-to-mesenchymal transition via phenotypic and proteomic changes in melanoma cell lines. Cancer Res. 2018;78:3913–3925. doi: 10.1158/0008-5472.CAN-17-1891. [DOI] [PubMed] [Google Scholar]
  • 164.Rajagopalan, S. & Long, E. O. KIR2DL4 (CD158d): An activation receptor for HLA-G. Front Immunol3, 258 (2012). 10.3389/fimmu.2012.00258. [DOI] [PMC free article] [PubMed]

Articles from Cellular and Molecular Immunology are provided here courtesy of Nature Publishing Group

RESOURCES