Europe PMC
Nothing Special   »   [go: up one dir, main page]

Europe PMC requires Javascript to function effectively.

Either your web browser doesn't support Javascript or it is currently turned off. In the latter case, please turn on Javascript support in your web browser and reload this page.

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


Natural killer (NK) cells are innate lymphoid cells endowed with cytolytic activity and a capacity to secrete cytokines and chemokines. Several lines of evidence suggest that NK cells play an important role in anti-tumor immunity. Some therapies against hematological malignacies make use of the immune properties of NK cells, such as their ability to kill residual leukemic blasts efficiently after conditioning during haploidentical hematopoietic stem cell transplantation. However, knowledge on NK cell infiltration and the status of NK cell responsiveness in solid tumors is limited so far. The pro-angiogenic role of the recently described NK cell-like type 1 innate lymphoid cells (ILC1s) and their phenotypic resemblance to NK cells are confounding factors that add a level of complexity, at least in mice. Here, we review the current knowledge on the presence and function of NK cells in solid tumors as well as the immunotherapeutic approaches designed to harness NK cell functions in these conditions, including those that aim to reinforce conventional anti-tumor therapies to increase the chances of successful treatment.

Free full text 


Logo of cmiLink to Publisher's site
Cell Mol Immunol. 2019 May; 16(5): 415–422.
Published online 2019 Mar 25. https://doi.org/10.1038/s41423-019-0224-2
PMCID: PMC6474204
PMID: 30911118

Targeting natural killer cells in solid tumors

Abstract

Natural killer (NK) cells are innate lymphoid cells endowed with cytolytic activity and a capacity to secrete cytokines and chemokines. Several lines of evidence suggest that NK cells play an important role in anti-tumor immunity. Some therapies against hematological malignacies make use of the immune properties of NK cells, such as their ability to kill residual leukemic blasts efficiently after conditioning during haploidentical hematopoietic stem cell transplantation. However, knowledge on NK cell infiltration and the status of NK cell responsiveness in solid tumors is limited so far. The pro-angiogenic role of the recently described NK cell-like type 1 innate lymphoid cells (ILC1s) and their phenotypic resemblance to NK cells are confounding factors that add a level of complexity, at least in mice. Here, we review the current knowledge on the presence and function of NK cells in solid tumors as well as the immunotherapeutic approaches designed to harness NK cell functions in these conditions, including those that aim to reinforce conventional anti-tumor therapies to increase the chances of successful treatment.

Subject terms: Innate lymphoid cells, Tumour immunology

NK cells

Natural killer (NK) cells belong to the innate lymphoid cell (ILC) family.1 The various ILC subtypes are characterized principally by their cytokine secretion profiles, which resemble those of CD4+ T helper cells; ILC1s produce the type 1 cytokine interferon-γ (IFN-γ), ILC2s produce the type 2 cytokines interleukin-5 (IL-5) and IL-13, and ILC3s produce the type 3 cytokines IL-17 and IL-22. NK cells are the predominant population of ILCs with cytolytic functions.

NK cells are present primarily in the peripheral blood, spleen and bone marrow, but they can infiltrate tissues and are also found in the liver, lungs, gut, lymph nodes and uterus.2 Recruitment to inflamed tissues depends on the expression of several chemokine receptors, such as CX3CR1, CXCR2 and CXCR1. In mice, NK cells are defined as CD3NKp46+CD127.1,3 In human peripheral blood, NK cells are defined as CD14CD19CD3 and can be divided into two populations, CD56bright and CD56dim NK cells, on the basis of their level of expression of the CD56 antigen4,5 (see Table 1). CD56dim NK cells express CD16 (FcγRIIIA) and combine cytotoxicity and cytokine production as effector functions, while CD56bright NK cells are more specialized in the secretion of cytokines. NK cell effector functions are tightly regulated by the balance between activating and inhibitory signals delivered by cell surface receptors.5 Accordingly, NK cells express activating and inhibitory receptors that bind to major histocompatibility complex (MHC) class I molecules, including killer cell immunoglobulin-like receptors (KIRs) in humans and Ly49 in mice.46 These receptors are highly polymorphic. In contrast, the inhibitory receptor CD94/NKG2A and the activating receptor CD94-NKG2C are invariant. CD94-NKG2A dimers recognize human leukocyte antigen (HLA)-E in humans and Qa-1 in mice. The inhibitory receptors KIR, Ly49 and CD94-NKG2A detect self-MHC-I ligands on healthy cells and help prevent NK cell activation and cell killing. Several activating receptors, including CD16, NKG2D, SLAM-family members and the natural cytotoxicity receptors (NCRs), NKp30, NKp44 and NKp46, can be targeted to induce NK cell-mediated anti-tumor immunity.5,7,8 NKp46 is highly conserved among mammalian species, whereas NKp30 and NKp44 are expressed only in humans. Human NK cells express NKp30 constitutively, whereas NKp44 expression is induced upon activation. The full activation of NK cells requires the coengagement of several different cell surface receptors.9,10

Table 1

Phenotype of human ILC1s

NK CD56brightNK CD56dimCD127+ILC1Ref.
SURFACE MARKERS
CD127+/−+ 108, 109
CD56+++ 108, 110112
CD161+++/−+ 108, 111
CD117+/− 108, 111, 112
CD337 (NKp30)++++ 1, 111
CD336 (NKp44)On activated cellsOn activated cells 1, 110, 111
CD335 (NKp46)+++ 1, 110112
NKp80++ 1, 103
CD200R+ 102
IL12Rβ+++ 1, 108
CCR6LowLow+ 108, 113
CD16+/−++ 108, 110, 114
KIR+/− 111, 112
2DL4+n.d. 111
CD94/NKG2A+++/−+/− 108, 110112
NKG2C+++ 110
NKG2D++++ 110, 115
Perforin+++ 111
CD25 (IL2Ra)+ 108, 111
IL1R1++++ 108, 111, 112
CD62L+++/− 111, 112, 116
HLA-DR++/− 111, 117
CD122+++n.d. 1
CD57+/− 111, 112, 118
CD160+++ 111, 112
CX3CR1++n.d. 111, 112
CD197 (CCR7)++n.d. 111, 112
CD181 (CXCR1)++n.d. 111, 112
CD183 (CXCR3)+++/−+ 1, 111, 112
TRANSCRIPTION FACTORS
Tbet++ 1
Eomes+ 1

Intraepithelial and liver ILC1s were excluded

ILC1s type 1 innate lymphoid cells, NK natural killer, n.d. not determined

Anti-tumor immunity of NK cells

NK cells form an immunological synapse with target cells, leading to the direct exocytosis of cytotoxic granules containing both perforin (a membrane-disrupting protein) and granzymes (a family of proteolytic enzymes), resulting in the specific lysis of target cells.11 NK cells can also express membrane-bound tumor necrosis factor (TNF) and death-inducing ligands, such as FAS ligand and TNF-related apoptosis-inducing ligand (TRAIL), upon activation. Cytotoxicity is one of the keys to anti-tumor immunity, as shown by mice lacking perforin, which lose control of tumor growth.12,13 In addition to their cytotoxic activities, NK cells also secrete cytokines, such as IFN-γ, TNF-α, growth factors (e.g., granulocyte–macrophage colony-stimulating factor) and chemokines (CCL3, CCL4 and CCL5), which shape the innate and adaptive immune responses.14 Chemokine secretion underlies NK cell colocalization with other hematopoietic cells, such as dendritic cells (DCs), in inflamed tissues.15 Furthermore, IFN-γ production helps to shape T cell responses in lymph nodes,16 activates myeloid cells and has effects on angiogenesis.17 The NK cell-mediated killing of target cells also affects T cell anti-tumor responses, possibly by promoting antigen cross-presentation to CD8+ cytotoxic T cells.18

In the 1980s, several studies reported a higher incidence of cancers in individuals with defective NK cell function.19,20 During the same period, mutant mice with impaired NK cell activity and mice treated with an NK cell-depleting antibody were reported to display higher rates of tumor growth and metastasis.21,22 Many studies have since reported poor NK cell function in cancer patients,2325 and case reports have been published on NK cell deficiencies26 that are characterized by an absence of NK cells or NK cell function or are caused by mutations of genes such as GATA227 or MCM4,28 suggesting a link to higher rates of malignancy. However, these immunodeficiencies also affect cells other than NK cells, and the relationship between a high risk of cancer and low levels of NK cell activity in humans remains purely correlative. Recent studies showed that the incidence of cancers was not higher in NK cell-deficient patients than in the general population, but a compensatory mechanism involving redundant leukocyte populations could not be excluded.29,30 In addition, the considered cohorts were too small, and the time of follow-up was insufficient to properly analyze the involvement of NK cells in tumor immunosurveillance. In mice, NK cells have been shown to control the growth and metastasis of transplantable tumors in studies based on the antibody-mediated depletion of NK cells.31 However, in many of these models, mouse NK cells were depleted with an antibody against NK1.1 that can also target ILC1s, subsets of activated T cells and, at least in theory, invariant natural killer T cells. Other studies used polyclonal anti-asialo GM1 serum, but asialo GM1 was detected on a subpopulation of NKT, CD8+ T and γδ T cells32,33 and on some activated CD4+ T cells, macrophages, eosinophils and basophils under certain experimental conditions.3437 NKp46 was recently identified as the cell marker displaying the highest degree of restriction to NK cells, but even this marker is also expressed by discrete subsets of T cells (in particular γδ T cells), ILC1s and a fraction of ILC3s.1 Therefore, there is no formal proof that NK cells are absolutely required for immunosurveillance against tumor formation under natural conditions. However, NK cells may have an antileukemic effect in patients undergoing allogeneic hematopoietic stem cell transplantation if donor NK cells express inhibitory KIRs mismatched with the patient’s cells for MHC class I molecules because they recognize recipient leukemia cells as foreign cells.3841 In addition, the adoptive transfer of genetically modified NK cells, checkpoint inhibitors and bispecific antibodies recently developed to target NK cells and tumor antigens are promising immunotherapeutic strategies that take advantage of the anti-tumor activities of NK cells to eliminate cancer.42

The presence of NK cells in solid tumors

Several studies have shown that the natural cytotoxicity of peripheral blood is significantly lower in patients with various types of solid tumors than in healthy individuals.43 The downregulation of the activating receptors NKp46, NKp30 and NKG2D at the cell surface of peripheral blood NK cells, and their magnitudes, were correlated with tumor progression in patients with cervical cancer.44 However, little information is available concerning NK cell infiltration into tumors.

An early study reported a correlation between the risk of local recurrence after tumor resection and preoperative levels of NK cells in patients with colorectal carcinoma; low levels of these cells were associated with a higher risk of local recurrence than high levels of these cells.45 More recently, colorectal tumors have been shown to be almost devoid of NK cells, despite efficient T cell infiltration.46 Similarly, melanoma skin lesions display poor CD56+ NK cell infiltration (<1% of the infiltrating cells).47 However, CD56dimKIR+CD57+ NK cells, which are highly cytotoxic, were present in tumor-infiltrated lymph nodes,48 and the levels of NKp30 and NKG2D expression were inversely correlated with the number of tumor cells in infiltrated lymph nodes adjacent to metastatic melanomas,49 suggesting a possible role for NK cells in tumor control.

In breast cancer patients, tumor-infiltrating NK cell counts have been associated with the response to neoadjuvant treatments, including anti-HER2 mAbs,50,51 and therapies with anti-HER2 trastuzumab–docetaxel and the antibody drug conjugate trastuzumab emtansine (T-DM1).5254 Recently, baseline NK tumor-infiltrating lymphocytes (TILs) were defined as independent biomarkers with great predictive value for complete response to anti-HER2 antibody-based treatment.55 NK cell infiltration of breast tumors is thus an indication of good prognosis.

NK cells invade clear cell renal tumors, and this invasion was associated with a favorable prognosis.56 NK cells were distributed throughout the tumor mass, rather than being restricted to the surrounding stroma.57,58 In this pathology, infiltrating NK cells were abundant but only weakly cytotoxic. Infiltrating NK cells had a CD56bright phenotype and overexpress NKG2A, but they exhibited lower levels of KIRs and leukocyte immunoglobulin-like receptor subfamily B member 1 (LILRB1) than autologous peripheral NK cells.

Head and neck (H&N) cancers are infiltrated with NK cells (Fig. (Fig.1),1), and this infiltration was associated with longer survival. Poor peripheral blood NK cell function was correlated with the development of metastases in H&N cancer59 and pharyngeal cancer.25,60 NK TILs displayed a decreased frequency in the mature CD56dimCD16hi population, with a concomitant decrease in expression of the maturation marker CD57 (Figs. (Figs.22 and and3).3). The frequencies of NK TILs expressing the inhibitory KIRs, KIR3DL1 and KIR2DL1/2/3, were lower among infiltrating than among circulating NK cells, but we observed an increase in the frequency of NK TILs expressing the inhibitory receptor NKG2A (Fig. 2 and see ref. 61).

An external file that holds a picture, illustration, etc.
Object name is 41423_2019_224_Fig1_HTML.jpg

NK infiltration in head and neck squamous cell cancer and lung small cell cancer samples Number of NKp46+ cells per mm2 measured by IHC in tumor samples and normal adjacent tissue (NAT) and percentage of tissue slice with NK cells are indicated (A and B, left panels). NK cells frequencies defined as CD45+, CD3, CD56+, CD4 and/or CD14 among CD45+ leukocytes were measured in blood, NAT and tumor samples was by flow cytometry. The box and whiskers represent the maximum, 75 percentile, median, 25 percentile, and minimum. Each dot indicates a sample

An external file that holds a picture, illustration, etc.
Object name is 41423_2019_224_Fig2_HTML.jpg

Contour plots showing the positivity thresholds for indicated markers. NK cells were defined by flow cytometry as CD45+, CD3, CD56+, CD4 and/or CD14 and represented by red contour plots. T cells defined as CD45+, CD3+, CD56 are represented in blue contour plots. Representative expression of several markers, on blood or tumor samples from head and neck squamous cell cancer and non-small cell lung cancer patients, exemplifying the staining and the positioning of the thresholds are shown

An external file that holds a picture, illustration, etc.
Object name is 41423_2019_224_Fig3_HTML.jpg

Expression profile of surface markers on NK cells from head and neck squamous cell cancer samples. NK cells were analyzed by flow cytometry and defined as CD45+, CD3, CD56+, CD4 and/or CD14 in blood, normal adjacent tissue (NAT) and tumor samples. Frequencies of NK cells expressing indicated surface marker are shown. Box and whiskers represent maximum, 75 percentile, median, 25 percentile, and minimum. Each dot indicates a sample. For each marker the Wilcoxon test is used when matched data for the three kinds of samples were available, otherwise the Kruskal-Wallis test is used. Significant p-values below 0.05 are shown

Checkpoint blockade immunotherapy targeting the programmed cell death protein 1/programmed death-ligand 1 (PD-1/PD-L1) inhibitory axis produced remarkable results in the treatment of several types of cancer.6267 PD-1 is mostly expressed by T cells, but NK cells with an activated and more responsive phenotype can also express PD-1.68,69 We detected a few NK TILs expressing PD-1 in H&N tumors (Fig. (Fig.33 and see ref. 61). CD96 and TIGIT are both checkpoints currently under consideration for possible use in immunotherapy for cancer. We observed an increase in the frequency of NK TILs expressing CD96 but not TIGIT,70 which are coinhibitory receptors recognizing nectin and nectin-like ligands overexpressed in various cancer cell types.71

NK cell infiltration per se has no impact on clinical outcome in non-small-cell lung cancer (NSCLC) (Fig. (Fig.11 and see refs. 72,73). However, NK cells are the least abundant immune cell lineage in lung adenocarcinoma early lesions compared to normal lung tissue.72 NK cells infiltrated lung tumors (Fig. 1 and see refs. 72,73) and were found in the stroma on NSCLC biopsy, rather than in direct contact with tumor cells.73 Nevertheless, the clinical outcome was more dependent on the NK cell phenotype and function than on the NK cell density in patients with NSCLC. Indeed, tumor-infiltrating NK cells had a profoundly impaired cytotoxic potential despite their expression of several activation markers, including NKp44, CD69 and HLA-DR (Fig. (Fig.44 and see refs. 72,73). However, other activating receptors, such as NKp30 and NKp80, were strongly downregulated on intratumoral NK cells, and this may impair cytotoxicity.74 Furthermore, NSCLC samples had higher levels of CD56hiCD16CD57 NK cells than the surrounding normal lung tissues, and in addition to NKG2A, NK cells from these samples expressed inhibitory KIRs that are normally expressed by CD56lowCD16+ NK cells (Fig. (Fig.44 and see ref. 73). We observed frequencies of NK TILs expressing higher levels of CD96 than those in the surrounding tissues and on circulating NK cells (Fig. 4). We also detected a few NK TILs expressing PD-1 in lung tumors (Fig. 4). Overall, these results indicate that NK cells can invade some solid tumors. When NK cells infiltrate solid tumors, high numbers of NK cells may be associated with better survival. However, in most cases, anti-tumor properties of NK cells are attenuated at the tumor bed. Thus, the targeting of NK cells by immunotherapy is an attractive anti-cancer strategy.

An external file that holds a picture, illustration, etc.
Object name is 41423_2019_224_Fig4_HTML.jpg

Expression profile of surface markers in natural killer (NK) cells from lung small-cell cancer samples. NK cells were analyzed by flow cytometry and defined as CD45+, CD3, CD56+, CD4 and/or CD14 in blood, normal adjacent tissue (NAT) and tumor samples. Frequencies of NK cells expressing the indicated surface markers are shown. Box and whiskers represent the maximum, 75 percentile, median, 25 percentile and minimum. Each dot indicates a sample. For each marker, the Wilcoxon test was used when matched data for the three types of samples were available; otherwise, the Kruskal–Wallis test was used. Significant p values below 0.05 are shown

The tumor microenvironment represses NK cell recruitment and/or anti-tumor NK cell functions

Some tumors are poorly permeable to NK cells. Tumor cells themselves may affect the ability of NK cells to infiltrate the tumor mass via several mechanisms. For example, neuroblastoma cells have been reported to modulate the NK cell chemokine-receptor repertoire by producing transforming growth factor-β (TGF-β).75 Neuroblastoma cells induce the upregulation of CXCR4 and CXCR3 expression on all NK cells and the downregulation of CX3CR1 by the CD56dim subset. Another mechanism by which tumors evade NK cell attack is based on the prevention of cell contact. Ovarian tumor cells interfere with the formation of immunological synapses with NK cells by expressing the mucin 16 (MUC16) glycoprotein, which has antiadhesion properties.76 Tumor cells may also display altered patterns of activating or inhibitory receptor ligand expression, thus preventing NK cell activation. For example, melanoma cells become resistant to NK cell attack after multiple contacts through an upregulation of their surface expression of both classical and nonclassical HLA class I molecules.77 Another well-known mechanism of NK cell-mediated lysis evasion involves the modulation of NKG2D ligands at the tumor cell surface or the secretion of the soluble NKG2D ligand MICA or soluble NKp30 ligands B7H6 and HLA-B-associated transcript 3/BCL2-associated athanogene 6 (BAT3/BAG6).78,79 Finally, NK cell activities may also be downregulated by the engagement of the NKp44 receptor with proliferating cell nuclear antigen, which is overexpressed in various tumor types.80,81

Cells from the tumor microenvironment may also have suppressive activities against NK cells, such as myeloid-derived suppressor cells, tumor-associated macrophages, which can be isolated from patients with hepatocellular carcinoma,82 or cancer-associated fibroblasts.77,83,84 The physical status of the tumor microenvironment, including characteristics such as hypoxia and the production of various soluble factors by the tumor cells or other cells, may also impair NK cell function. For example, TGF-β, indoleamine 2,3-dioxygenase (IDO), IL-4 and prostaglandin E2 (PGE2) can interfere with NK cell activation.77,8587

NK cell-based immunotherapies for solid tumors

Efforts should be made to improve NK cell recruitment to the tumor bed. Tumor NK cell infiltration is an important parameter controlled by chemokines and adhesion molecules that may differ from those regulating T cell migration.46,88 An antibody-based NK cell-homing protein, the NK cell-recruiting protein-conjugated antibody (NRP-body), has recently been developed.89 The NRP-body increased NK cell infiltration along a CXCL16 gradient in a murine primary and metastatic pancreatic ductal adenocarcinoma tumor model. NRP-body administration increased the infiltration of transferred NK cells into tumor tissues and reduced tumor burden relative to that in controls, thereby increasing the overall survival. The combination of the NRP-body with NK cell therapy may, therefore, be of potential therapeutic value.

Other strategies consist of promoting NK cell activation. This is, for example, achieved using cytokine treatments. NKTR-214 is a PEGylated IL-2 that preferentially activates IL-2Rβ.90 In preclinical models, NKTR-214 increased the expansion of the NK cell population and strongly increased the CD8+ T cell to regulatory T cell ratio. NKTR-214 is currently in phase I/II clinical trials for various solid tumors (NCT02983045, NCT02869295, NCT03138889, NCT03435640 and NCT03282344).

As mentioned above, activated NK cells have been shown to express PD-1 in certain circumstances.91,92 NK cells from patients with multiple myeloma express PD-1, and the anti-PD-1 antibody (CT-011) could potentially be used to restore NK cell-mediated anti-tumor activity.93 A combination of anti-PD-1 and anti-CTLA4 mAbs has recently been shown to be effective for the treatment of melanoma.65,94 Monalizumab is an IgG4-blocking mAb directed against NKG2A, which, in combination with the anti-PD-L1 antibody, promotes NK cell effector functions and antibody-dependent cell-mediated cytotoxicity (ADCC) and effector T cell responses.61 A combination of monalizumab and cetuximab (anti-epidermal growth factor receptor (EGFR)) gave positive results in a phase II clinical trial in patients with previously treated squamous cell carcinoma of the head and neck.61 Monalizumab is being investigated in several studies, particularly in combination with durvalumab, an anti-PD-L1 antibody, in solid tumors (phase I, NCT02671435).

Another approach to direct NK cell-mediated cytotoxicity against tumors was achieved through chimeric antigen receptor (CAR) expression, which is an interesting strategy for treating refractory cancers. Various tumor antigens, including EGFR, epithelial cell adhesion molecule, human epidermal, ganglioside GD2, growth factor receptor 2 (HER2; also known as ERBB2), the inactive tyrosine-protein kinase transmembrane receptor ROR1 and Wilms’ tumor protein (WT1), have been targeted by CAR NK therapy with exciting clinical results.95 Another approach based on NKG2D-expressing CAR NK cells promotes anti-tumor NK cell activity in an osteosarcoma mouse model.96 These cells have been tested in one phase I study in patients with metastatic solid tumors.97

Finally, bispecific killer cell engagers have been designed to promote the NK cell-mediated lysis of tumor targets. Two bispecific CD16-based antibodies (AFM22 and AFM24) bind to the EGFR variant III (which is expressed by several types of solid tumors, including glioblastoma) and wild-type EGFR, respectively, and are in preclinical development. Other bispecific antibodies have been generated and are in preclinical tests (Gauthier et al., unpublished data). These NK cell engagers (NKCEs) target the activating NK cell receptor NKp46 together with a tumor antigen and an Fc fragment to promote ADCC via the activating receptor CD16, which is expressed on NK cells. These NKCEs were more potent in vitro than clinical therapeutic antibodies targeting the same tumor antigen. In vivo, these antibodies efficiently controlled tumor growth in mouse models of solid and invasive tumors. Interestingly, these antibodies promoted the infiltration/proliferation of NK cells within tumors.

Fuzzy frontiers between NK cells and ILC1s

NK cells and ILC1s have different developmental paths, but they share several features; however, the properties differentiating NK cells from ILC1s are still poorly defined, particularly in humans. NK cells are cytotoxic cells that circulate in the bloodstream, whereas ILC1s are tissue-resident cells.98,99 ILC1s are generally weakly cytotoxic or noncytotoxic and act as a first line of defense against infections with viruses and certain parasite and bacteria, such as Toxoplasma gondii100 or Clostridium difficile.101 Both of these cell types mainly produce IFN-γ. The phenotypic characterization of ILC1s is often problematic. In both humans and mice, ILC1s preferentially express CD49a and TRAIL, but the specificity of these markers is tissue dependent, and their expression is often lost upon cell activation. ILC1s share some phenotypic markers with NK cells and ILC3s, such as NK1.1 in mice, and NCRs, such as NKp44 in humans and NKp46 in both humans and mice, in at least some organs (Table 1). In humans, CD127 may not be an absolute ILC1 marker because a majority of CD56bright CD16 NK cells in peripheral blood express CD127, as ILC1s. In contrast, highly cytotoxic CD56dim CD16+ NK cells do not express this marker. CD200R expression has been shown to distinguish ILC1s from NK cells in mice.102 Another marker of human NK cells that is not expressed on ILC1s is NKp80.103 Human ILC1s are, thus, negatively selected as LinCD127+CRTH2cKitNKp80 cells in flow cytometry, and specific positive staining is not currently possible in immunohistochemistry.

Recent studies have shown that NK cells can be converted into ILC1s (converted ILC1s),104 ILC3s can acquire an ILC1-like phenotype (ex-ILC3)105,106 and markers previously used to differentiate ILC populations are often specific to particular immune contexts or tissues. These observations are of particular interest because converted ILC1s may facilitate tumor growth in the TGFβ-rich environment of mouse models of solid tumors, as TGFβ induces the conversion of CD49b+CD49aEOMES+ NK cells into CD49bCD49a+EOMES ILC1s.104 Tumor ILC1s express higher levels of inhibitory receptors, including NKG2A, killer cell lectin-like receptor subfamily G member 1 (KLRG1), cytotoxic T lymphocyte antigen 4 (CTLA4) and lymphocyte activation gene 3 protein (LAG3), and they have low levels of DNAM1 and increased levels of TIGIT, potentially enabling tumors to escape immune surveillance by the innate immune system.104,107 Given the potential pro-angiogenic role of ILC1s, further work in the phenotypic characterization of these cells is clearly required.

Conclusions

Unlike T cells or dendritic cells, NK cells are often underappreciated at the tumor bed, leading to speculation that these cells might only be involved in the control of hematologic malignancies and tumor metastasis. However, the cytolytic activities of NK cells promote CD8+ T cell cross-priming, and NK cell-derived cytokines and chemokines attract and activate DCs, macrophages, and neutrophils. NK cells can, therefore, make a significant contribution to anti-tumor immune responses. Remarkably, high levels of NK cell infiltration have been associated with a better prognosis, at least for some tumors. Thus, it is of interest to promote NK cell infiltration of tumors and to develop immunotherapeutic approaches designed to harness NK cell functions in the tumor bed, including those that aim to reinforce conventional anti-tumor therapies to increase the chances of successful treatment.

Acknowledgements

The laboratory of E.V. is supported by funding from the European Research Council (ERC) under the European Union Horizon 2020 research and innovation program (Targeting innate lymphoid cells (TILC), grant agreement number 694502), the Agence Nationale de la Recherche (PIONeeR Project (ANR-17-RHUS-0007)), Equipe Labellisée “La Ligue”, Ligue Nationale contre le Cancer, MSDAvenir, Innate Pharma and institutional grants to the CIML (Institut National Français de Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS) and Aix-Marseille University) and to Marseille Immunopôle.

Competing interests

G.H., P.A. and E.V. are employees of Innate Pharma. The other authors declare no competing interests.

References

1. Vivier E, et al. Innate lymphoid cells: 10 years on. Cell. 2018;174:1054–1066. [Abstract] [Google Scholar]
2. Shi FD, Ljunggren HG, La Cava A, Van Kaer L. Organ-specific features of natural killer cells. Nat. Rev. Immunol. 2011;11:658–671. [Europe PMC free article] [Abstract] [Google Scholar]
3. Walzer T, et al. Identification, activation, and selective in vivo ablation of mouse NK cells via NKp46. Proc. Natl. Acad. Sci. USA. 2007;104:3384–3389. [Europe PMC free article] [Abstract] [Google Scholar]
4. Biassoni, R., Bottino, C., Cantoni, C. & Moretta, A. Human natural killer receptors and their ligands. Curr. Protoc. Immunol. Chapter 14, Unit 14.10 (2002). [Abstract]
5. Moretta L, et al. Surface NK receptors and their ligands on tumor cells. Semin. Immunol. 2006;18:151–158. [Abstract] [Google Scholar]
6. Guia S, Fenis A, Vivier E, Narni-Mancinelli E. Activating and inhibitory receptors expressed on innate lymphoid cells. Semin. Immunopathol. 2018;40:331–341. [Abstract] [Google Scholar]
7. Moretta A, et al. Receptors for HLA class-I molecules in human natural killer cells. Annu. Rev. Immunol. 1996;14:619–648. [Abstract] [Google Scholar]
8. Wu N, Veillette A. SLAM family receptors in normal immunity and immune pathologies. Curr. Opin. Immunol. 2016;38:45–51. [Abstract] [Google Scholar]
9. Bryceson YT, Ljunggren HG, Long EO. Minimal requirement for induction of natural cytotoxicity and intersection of activation signals by inhibitory receptors. Blood. 2009;114:2657–2666. [Europe PMC free article] [Abstract] [Google Scholar]
10. Bryceson YT, March ME, Ljunggren HG, Long EO. Activation, coactivation, and costimulation of resting human natural killer cells. Immunol. Rev. 2006;214:73–91. [Europe PMC free article] [Abstract] [Google Scholar]
11. Vivier E, Tomasello E, Baratin M, Walzer T, Ugolini S. Functions of natural killer cells. Nat. Immunol. 2008;9:503–510. [Abstract] [Google Scholar]
12. Smyth MJ, et al. Perforin-mediated cytotoxicity is critical for surveillance of spontaneous lymphoma. J. Exp. Med. 2000;192:755–760. [Europe PMC free article] [Abstract] [Google Scholar]
13. Street SE, et al. Host perforin reduces tumor number but does not increase survival in oncogene-driven mammary adenocarcinoma. Cancer Res. 2007;67:5454–5460. [Abstract] [Google Scholar]
14. Vivier E, et al. Innate or adaptive immunity? The example of natural killer cells. Science. 2011;331:44–49. [Europe PMC free article] [Abstract] [Google Scholar]
15. Moretta A, et al. Early liaisons between cells of the innate immune system in inflamed peripheral tissues. Trends Immunol. 2005;26:668–675. [Abstract] [Google Scholar]
16. Martin-Fontecha A, et al. Induced recruitment of NK cells to lymph nodes provides IFN-gamma for T(H)1 priming. Nat. Immunol. 2004;5:1260–1265. [Abstract] [Google Scholar]
17. Hayakawa Y, et al. IFN-gamma-mediated inhibition of tumor angiogenesis by natural killer T-cell ligand, alpha-galactosylceramide. Blood. 2002;100:1728–1733. [Abstract] [Google Scholar]
18. Krebs P, et al. NK-cell-mediated killing of target cells triggers robust antigen-specific T-cell-mediated and humoral responses. Blood. 2009;113:6593–6602. [Europe PMC free article] [Abstract] [Google Scholar]
19. Roder JC, et al. A new immunodeficiency disorder in humans involving NK cells. Nature. 1980;284:553–555. [Abstract] [Google Scholar]
20. Sullivan JL, Byron KS, Brewster FE, Purtilo DT. Deficient natural killer cell activity in x-linked lymphoproliferative syndrome. Science. 1980;210:543–545. [Abstract] [Google Scholar]
21. Gorelik E, Wiltrout RH, Okumura K, Habu S, Herberman RB. Role of NK cells in the control of metastatic spread and growth of tumor cells in mice. Int J. Cancer. 1982;30:107–112. [Abstract] [Google Scholar]
22. Talmadge JE, Meyers KM, Prieur DJ, Starkey JR. Role of natural killer cells in tumor growth and metastasis: C57BL/6 normal and beige mice. J. Natl. Cancer Inst. 1980;65:929–935. [Abstract] [Google Scholar]
23. Nakajima T, Mizushima N, Nakamura J, Kanai K. Surface markers of NK cells in peripheral blood of patients with cirrhosis and hepatocellular carcinoma. Immunol. Lett. 1986;13:7–10. [Abstract] [Google Scholar]
24. Pross HF, Lotzova E. Role of natural killer cells in cancer. Nat. Immun. 1993;12:279–292. [Abstract] [Google Scholar]
25. Schantz SP, Shillitoe EJ, Brown B, Campbell B. Natural killer cell activity and head and neck cancer: a clinical assessment. J. Natl. Cancer Inst. 1986;77:869–875. [Abstract] [Google Scholar]
26. Orange JS. Natural killer cell deficiency. J. Allergy Clin. Immunol. 2013;132:515–525. [Europe PMC free article] [Abstract] [Google Scholar]
27. Spinner MA, et al. GATA2 deficiency: a protean disorder of hematopoiesis, lymphatics, and immunity. Blood. 2014;123:809–821. [Europe PMC free article] [Abstract] [Google Scholar]
28. Gineau L, et al. Partial MCM4 deficiency in patients with growth retardation, adrenal insufficiency, and natural killer cell deficiency. J. Clin. Invest. 2012;122:821–832. [Europe PMC free article] [Abstract] [Google Scholar]
29. Ebbo M, et al. Low circulating natural killer cell counts are associated with severe disease in patients with common variable immunodeficiency. EBioMedicine. 2016;6:222–230. [Europe PMC free article] [Abstract] [Google Scholar]
30. Vely F, et al. Evidence of innate lymphoid cell redundancy in humans. Nat. Immunol. 2016;17:1291–1299. [Europe PMC free article] [Abstract] [Google Scholar]
31. Smyth MJ, Hayakawa Y, Takeda K, Yagita H. New aspects of natural-killer-cell surveillance and therapy of cancer. Nat. Rev. Cancer. 2002;2:850–861. [Abstract] [Google Scholar]
32. Lee U, Santa K, Habu S, Nishimura T. Murine asialo GM1+CD8+T cells as novel interleukin-12-responsive killer T cell precursors. Jpn J. Cancer Res. 1996;87:429–432. [Europe PMC free article] [Abstract] [Google Scholar]
33. Trambley J, et al. Asialo GM1(+) CD8(+) T cells play a critical role in costimulation blockade-resistant allograft rejection. J. Clin. Invest. 1999;104:1715–1722. [Europe PMC free article] [Abstract] [Google Scholar]
34. Kataoka S, Konishi Y, Nishio Y, Fujikawa-Adachi K, Tominaga A. Antitumor activity of eosinophils activated by IL-5 and eotaxin against hepatocellular carcinoma. DNA Cell Biol. 2004;23:549–560. [Abstract] [Google Scholar]
35. Nishikado H, Mukai K, Kawano Y, Minegishi Y, Karasuyama H. NK cell-depleting anti-asialo GM1 antibody exhibits a lethal off-target effect on basophils in vivo. J. Immunol. 2011;186:5766–5771. [Abstract] [Google Scholar]
36. Slifka MK, Pagarigan RR, Whitton JL. NK markers are expressed on a high percentage of virus-specific CD8+and CD4+T cells. J. Immunol. 2000;164:2009–2015. [Abstract] [Google Scholar]
37. Wiltrout RH, et al. Reactivity of anti-asialo GM1 serum with tumoricidal and non-tumoricidal mouse macrophages. J. Leukoc. Biol. 1985;37:597–614. [Abstract] [Google Scholar]
38. Cooley S, et al. Donor selection for natural killer cell receptor genes leads to superior survival after unrelated transplantation for acute myelogenous leukemia. Blood. 2010;116:2411–2419. [Europe PMC free article] [Abstract] [Google Scholar]
39. Hsu KC, et al. Improved outcome in HLA-identical sibling hematopoietic stem-cell transplantation for acute myelogenous leukemia predicted by KIR and HLA genotypes. Blood. 2005;105:4878–4884. [Europe PMC free article] [Abstract] [Google Scholar]
40. Ruggeri L, et al. Role of natural killer cell alloreactivity in HLA-mismatched hematopoietic stem cell transplantation. Blood. 1999;94:333–339. [Abstract] [Google Scholar]
41. Ruggeri L, et al. Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants. Science. 2002;295:2097–2100. [Abstract] [Google Scholar]
42. Chiossone L, Dumas PY, Vienne M, Vivier E. Natural killer cells and other innate lymphoid cells in cancer. Nat. Rev. Immunol. 2018;18:671–688. [Abstract] [Google Scholar]
43. Brittenden J, Heys SD, Ross J, Eremin O. Natural killer cells and cancer. Cancer. 1996;77:1226–1243. [Abstract] [Google Scholar]
44. Garcia-Iglesias T, et al. Low NKp30, NKp46 and NKG2D expression and reduced cytotoxic activity on NK cells in cervical cancer and precursor lesions. BMC Cancer. 2009;9:186. [Europe PMC free article] [Abstract] [Google Scholar]
45. Tartter PI, Steinberg B, Barron DM, Martinelli G. The prognostic significance of natural killer cytotoxicity in patients with colorectal cancer. Arch. Surg. 1987;122:1264–1268. [Abstract] [Google Scholar]
46. Halama N, et al. Natural killer cells are scarce in colorectal carcinoma tissue despite high levels of chemokines and cytokines. Clin. Cancer Res. 2011;17:678–689. [Abstract] [Google Scholar]
47. Erdag G, et al. Immunotype and immunohistologic characteristics of tumor-infiltrating immune cells are associated with clinical outcome in metastatic melanoma. Cancer Res. 2012;72:1070–1080. [Europe PMC free article] [Abstract] [Google Scholar]
48. Ali TH, et al. Enrichment of CD56(dim)KIR+CD57+highly cytotoxic NK cells in tumour-infiltrated lymph nodes of melanoma patients. Nat. Commun. 2014;5:5639. [Europe PMC free article] [Abstract] [Google Scholar]
49. Messaoudene M, et al. Mature cytotoxic CD56(bright)/CD16(+) natural killer cells can infiltrate lymph nodes adjacent to metastatic melanoma. Cancer Res. 2014;74:81–92. [Abstract] [Google Scholar]
50. Denkert C, et al. Tumour-infiltrating lymphocytes and prognosis in different subtypes of breast cancer: a pooled analysis of 3771 patients treated with neoadjuvant therapy. Lancet Oncol. 2018;19:40–50. [Abstract] [Google Scholar]
51. Salgado R, et al. Tumor-infiltrating lymphocytes and associations with pathological complete response and event-free survival in HER2-positive early-stage breast cancer treated with lapatinib and trastuzumab: a secondary analysis of the NeoALTTO Trial. JAMA Oncol. 2015;1:448–454. [Europe PMC free article] [Abstract] [Google Scholar]
52. Arnould L, et al. Trastuzumab-based treatment of HER2-positive breast cancer: an antibody-dependent cellular cytotoxicity mechanism? Br. J. Cancer. 2006;94:259–267. [Europe PMC free article] [Abstract] [Google Scholar]
53. Gennari R, et al. Pilot study of the mechanism of action of preoperative trastuzumab in patients with primary operable breast tumors overexpressing HER2. Clin. Cancer Res. 2004;10:5650–5655. [Abstract] [Google Scholar]
54. Muller P, et al. Trastuzumab emtansine (T-DM1) renders HER2+breast cancer highly susceptible to CTLA-4/PD-1 blockade. Sci. Transl. Med. 2015;7:315ra188. [Abstract] [Google Scholar]
55. Muntasell A, et al. NK cell infiltrates and HLA class I expression in primary HER2+breast cancer predict and uncouple pathological response and disease-free survival. Clin. Cancer Res. 2019;25:1535–1545. [Abstract] [Google Scholar]
56. Eckl J, et al. Transcript signature predicts tissue NK cell content and defines renal cell carcinoma subgroups independent of TNM staging. J. Mol. Med. 2012;90:55–66. [Abstract] [Google Scholar]
57. Schleypen JS, et al. Cytotoxic markers and frequency predict functional capacity of natural killer cells infiltrating renal cell carcinoma. Clin. Cancer Res. 2006;12:718–725. [Abstract] [Google Scholar]
58. Schleypen JS, et al. Renal cell carcinoma-infiltrating natural killer cells express differential repertoires of activating and inhibitory receptors and are inhibited by specific HLA class I allotypes. Int. J. Cancer. 2003;106:905–912. [Abstract] [Google Scholar]
59. Schantz SP, Ordonez NG. Quantitation of natural killer cell function and risk of metastatic poorly differentiated head and neck cancer. Nat. Immun. Cell Growth Regul. 1991;10:278–288. [Abstract] [Google Scholar]
60. Schantz SP, Savage HE, Racz T, Taylor DL, Sacks PG. Natural killer cells and metastases from pharyngeal carcinoma. Am. J. Surg. 1989;158:361–366. [Abstract] [Google Scholar]
61. 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.e13. [Europe PMC free article] [Abstract] [Google Scholar]
62. Baumeister SH, Freeman GJ, Dranoff G, Sharpe AH. Coinhibitory pathways in immunotherapy for cancer. Annu Rev. Immunol. 2016;34:539–573. [Abstract] [Google Scholar]
63. 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. [Abstract] [Google Scholar]
64. Okazaki T, Honjo T. PD-1 and PD-1 ligands: from discovery to clinical application. Int. Immunol. 2007;19:813–824. [Abstract] [Google Scholar]
65. Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science. 2015;348:69–74. [Abstract] [Google Scholar]
66. Sharma P, Allison JP. Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell. 2015;161:205–214. [Europe PMC free article] [Abstract] [Google Scholar]
67. Sharma P, Allison JP. The future of immune checkpoint therapy. Science. 2015;348:56–61. [Abstract] [Google Scholar]
68. Barry KC, et al. A natural killer-dendritic cell axis defines checkpoint therapy-responsive tumor microenvironments. Nat. Med. 2018;24:1178–1191. [Europe PMC free article] [Abstract] [Google Scholar]
69. Hsu J, et al. Contribution of NK cells to immunotherapy mediated by PD-1/PD-L1 blockade. J. Clin. Invest. 2018;128:4654–4668. [Europe PMC free article] [Abstract] [Google Scholar]
70. Dougall WC, Kurtulus S, Smyth MJ, Anderson AC. TIGIT and CD96: new checkpoint receptor targets for cancer immunotherapy. Immunol. Rev. 2017;276:112–120. [Abstract] [Google Scholar]
71. Anderson AC, Joller N, Kuchroo VK. Lag-3, Tim-3, and TIGIT: co-inhibitory receptors with specialized functions in immune regulation. Immunity. 2016;44:989–1004. [Europe PMC free article] [Abstract] [Google Scholar]
72. Lavin Y, et al. Innate immune landscape in early lung adenocarcinoma by paired single-cell analyses. Cell. 2017;169:750–765.e17. [Europe PMC free article] [Abstract] [Google Scholar]
73. Carrega P, et al. Natural killer cells infiltrating human nonsmall-cell lung cancer are enriched in CD56 bright CD16(-) cells and display an impaired capability to kill tumor cells. Cancer. 2008;112:863–875. [Abstract] [Google Scholar]
74. Platonova S, et al. Profound coordinated alterations of intratumoral NK cell phenotype and function in lung carcinoma. Cancer Res. 2011;71:5412–5422. [Abstract] [Google Scholar]
75. Castriconi R, et al. Neuroblastoma-derived TGF-beta1 modulates the chemokine receptor repertoire of human resting NK cells. J. Immunol. 2013;190:5321–5328. [Abstract] [Google Scholar]
76. Gubbels JA, et al. MUC16 provides immune protection by inhibiting synapse formation between NK and ovarian tumor cells. Mol. Cancer. 2010;9:11. [Europe PMC free article] [Abstract] [Google Scholar]
77. 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. [Abstract] [Google Scholar]
78. 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. [Abstract] [Google Scholar]
79. 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. [Europe PMC free article] [Abstract] [Google Scholar]
80. Horton NC, Mathew SO, Mathew PA. Novel interaction between proliferating cell nuclear antigen and HLA I on the surface of tumor cells inhibits NK cell function through NKp44. PLoS One. 2013;8:e59552. [Europe PMC free article] [Abstract] [Google Scholar]
81. 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. [Europe PMC free article] [Abstract] [Google Scholar]
82. Hoechst B, et al. Myeloid derived suppressor cells inhibit natural killer cells in patients with hepatocellular carcinoma via the NKp30 receptor. Hepatology. 2009;50:799–807. [Europe PMC free article] [Abstract] [Google Scholar]
83. Li T, et al. Hepatocellular carcinoma-associated fibroblasts trigger NK cell dysfunction via PGE2 and IDO. Cancer Lett. 2012;318:154–161. [Abstract] [Google Scholar]
84. Li T, et al. Colorectal carcinoma-derived fibroblasts modulate natural killer cell phenotype and antitumor cytotoxicity. Med. Oncol. 2013;30:663. [Abstract] [Google Scholar]
85. Castriconi R, et al. Transforming growth factor beta 1 inhibits expression of NKp30 and NKG2D receptors: consequences for the NK-mediated killing of dendritic cells. Proc. Natl. Acad. Sci. USA. 2003;100:4120–4125. [Europe PMC free article] [Abstract] [Google Scholar]
86. Marcenaro E, et al. IL-12 or IL-4 prime human NK cells to mediate functionally divergent interactions with dendritic cells or tumors. J. Immunol. 2005;174:3992–3998. [Abstract] [Google Scholar]
87. Pietra G, et al. Melanoma cells inhibit natural killer cell function by modulating the expression of activating receptors and cytolytic activity. Cancer Res. 2012;72:1407–1415. [Abstract] [Google Scholar]
88. Faveeuw C, Di Mauro ME, Price AA, Ager A. Roles of alpha(4) integrins/VCAM-1 and LFA-1/ICAM-1 in the binding and transendothelial migration of T lymphocytes and T lymphoblasts across high endothelial venules. Int. Immunol. 2000;12:241–251. [Abstract] [Google Scholar]
89. Lee J, et al. An antibody designed to improve adoptive NK-cell therapy inhibits pancreatic cancer progression in a murine model. Cancer Immunol. Res. 2019;7:219–229. [Abstract] [Google Scholar]
90. Charych DH, et al. NKTR-214, an engineered cytokine with biased IL2 receptor binding, increased tumor exposure, and marked efficacy in mouse tumor models. Clin. Cancer Res. 2016;22:680–690. [Abstract] [Google Scholar]
91. Alvarez IB, et al. Role played by the programmed death-1-programmed death ligand pathway during innate immunity against Mycobacterium tuberculosis. J. Infect. Dis. 2010;202:524–532. [Abstract] [Google Scholar]
92. Norris S, et al. PD-1 expression on natural killer cells and CD8(+) T cells during chronic HIV-1 infection. Viral Immunol. 2012;25:329–332. [Abstract] [Google Scholar]
93. Benson DM, et al. The PD-1/PD-L1 axis modulates the natural killer cell versus multiple myeloma effect: a therapeutic target for CT-011, a novel monoclonal anti-PD-1 antibody. Blood. 2010;116:2286–2294. [Europe PMC free article] [Abstract] [Google Scholar]
94. Amaria RN, et al. Neoadjuvant immune checkpoint blockade in high-risk resectable melanoma. Nat. Med. 2018;24:1649–1654. [Europe PMC free article] [Abstract] [Google Scholar]
95. Liu D, et al. Chimeric antigen receptor (CAR)-modified natural killer cell-based immunotherapy and immunological synapse formation in cancer and HIV. Protein Cell. 2017;8:861–877. [Europe PMC free article] [Abstract] [Google Scholar]
96. Chang YH, et al. A chimeric receptor with NKG2D specificity enhances natural killer cell activation and killing of tumor cells. Cancer Res. 2013;73:1777–1786. [Abstract] [Google Scholar]
97. Lonez C, et al. Study protocol for THINK: a multinational open-label phase I study to assess the safety and clinical activity of multiple administrations of NKR-2 in patients with different metastatic tumour types. BMJ Open. 2017;7:e017075. [Europe PMC free article] [Abstract] [Google Scholar]
98. Gasteiger G, Fan X, Dikiy S, Lee SY, Rudensky AY. Tissue residency of innate lymphoid cells in lymphoid and nonlymphoid organs. Science. 2015;350:981–985. [Europe PMC free article] [Abstract] [Google Scholar]
99. Peng H, et al. Liver-resident NK cells confer adaptive immunity in skin-contact inflammation. J. Clin. Invest. 2013;123:1444–1456. [Europe PMC free article] [Abstract] [Google Scholar]
100. Klose CSN, et al. Differentiation of type 1 ILCs from a common progenitor to all helper-like innate lymphoid cell lineages. Cell. 2014;157:340–356. [Abstract] [Google Scholar]
101. Abt MC, et al. Innate immune defenses mediated by two ILC subsets are critical for protection against acute Clostridium difficile infection. Cell Host Microbe. 2015;18:27–37. [Europe PMC free article] [Abstract] [Google Scholar]
102. Weizman OE, et al. ILC1 confer early host protection at initial sites of viral infection. Cell. 2017;171:795–808 e712. [Europe PMC free article] [Abstract] [Google Scholar]
103. Freud AG, et al. NKp80 defines a critical step during human natural killer cell development. Cell Rep. 2016;16:379–391. [Europe PMC free article] [Abstract] [Google Scholar]
104. Gao Y, et al. Tumor immunoevasion by the conversion of effector NK cells into type 1 innate lymphoid cells. Nat. Immunol. 2017;18:1004–1015. [Abstract] [Google Scholar]
105. Nussbaum K, et al. Tissue microenvironment dictates the fate and tumor-suppressive function of type 3 ILCs. J. Exp. Med. 2017;214:2331–2347. [Europe PMC free article] [Abstract] [Google Scholar]
106. Vonarbourg C, et al. Regulated expression of nuclear receptor RORgammat confers distinct functional fates to NK cell receptor-expressing RORgammat(+) innate lymphocytes. Immunity. 2010;33:736–751. [Europe PMC free article] [Abstract] [Google Scholar]
107. Cortez VS, et al. SMAD4 impedes the conversion of NK cells into ILC1-like cells by curtailing non-canonical TGF-beta signaling. Nat. Immunol. 2017;18:995–1003. [Europe PMC free article] [Abstract] [Google Scholar]
108. Hazenberg MD, Spits H. Human innate lymphoid cells. Blood. 2014;124:700–709. [Abstract] [Google Scholar]
109. Judge CJ, et al. CD56(bright) NK IL-7Ralpha expression negatively associates with HCV level, and IL-7-induced NK function is impaired during HCV and HIV infections. J. Leukoc. Biol. 2017;102:171–184. [Europe PMC free article] [Abstract] [Google Scholar]
110. Fuchs A. ILC1s in tissue inflammation and infection. Front. Immunol. 2016;7:104. [Europe PMC free article] [Abstract] [Google Scholar]
111. Michel T, et al. Human CD56bright NK cells: an update. J. Immunol. 2016;196:2923–2931. [Abstract] [Google Scholar]
112. Poli A, et al. CD56bright natural killer (NK) cells: an important NK cell subset. Immunology. 2009;126:458–465. [Abstract] [Google Scholar]
113. Lima M, et al. Chemokine receptor expression on normal blood CD56(+) NK-cells elucidates cell partners that comigrate during the innate and adaptive immune responses and identifies a transitional NK-cell population. J. Immunol. Res. 2015;2015:839684. [Europe PMC free article] [Abstract] [Google Scholar]
114. Mittal D, Vijayan D, Smyth MJ. Overcoming acquired PD-1/PD-L1 resistance with CD38 blockade. Cancer Discov. 2018;8:1066–1068. [Abstract] [Google Scholar]
115. Lopez-Verges S, et al. CD57 defines a functionally distinct population of mature NK cells in the human CD56dimCD16+NK-cell subset. Blood. 2010;116:3865–3874. [Europe PMC free article] [Abstract] [Google Scholar]
116. Kloverpris HN, et al. Innate lymphoid cells are depleted irreversibly during acute HIV-1 infection in the absence of viral suppression. Immunity. 2016;44:391–405. [Europe PMC free article] [Abstract] [Google Scholar]
117. Cruz-Zarate D, et al. Innate lymphoid cells have decreased HLA-DR expression but retain their responsiveness to TLR ligands during sepsis. J. Immunol. 2018;201:3401–3410. [Abstract] [Google Scholar]
118. Moretta L. Dissecting CD56dim human NK cells. Blood. 2010;116:3689–3691. [Abstract] [Google Scholar]

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

Citations & impact 


Impact metrics

Jump to Citations

Citations of article over time

Alternative metrics

Altmetric item for https://www.altmetric.com/details/57676286
Altmetric
Discover the attention surrounding your research
https://www.altmetric.com/details/57676286

Article citations


Go to all (128) article citations

Data 


Data behind the article

This data has been text mined from the article, or deposited into data resources.

Similar Articles 


To arrive at the top five similar articles we use a word-weighted algorithm to compare words from the Title and Abstract of each citation.