Next Article in Journal
Effects of Antiangiogenetic Drugs on Microcirculation and Macrocirculation in Patients with Advanced-Stage Renal Cancer
Next Article in Special Issue
Exploiting NK Cell Surveillance Pathways for Cancer Therapy
Previous Article in Journal
Flavonoids in Cancer and Apoptosis
Previous Article in Special Issue
The Potential for Cancer Immunotherapy in Targeting Surgery-Induced Natural Killer Cell Dysfunction
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

NK Cell-Based Immunotherapy in Cancer Metastasis

by
Seila Lorenzo-Herrero
1,2,3,†,
Alejandro López-Soto
1,2,3,†,
Christian Sordo-Bahamonde
1,2,3,
Ana P Gonzalez-Rodriguez
2,3,4,
Massimo Vitale
5 and
Segundo Gonzalez
1,2,3,*
1
Department of Functional Biology, Immunology, University of Oviedo, 33006 Oviedo, Spain
2
Instituto Universitario de Oncología del Principado de Asturias, IUOPA, 33006 Oviedo, Spain
3
Instituto de Investigación Biosanitaria del Principado de Asturias (IISPA), 33011 Oviedo, Spain
4
Department of Hematology, Hospital Universitario Central de Asturias (HUCA), 33011 Oviedo, Spain
5
UOC Immunologia, Ospedale Policlinico San Martino Genova, 16132 Genoa, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally.
Cancers 2019, 11(1), 29; https://doi.org/10.3390/cancers11010029
Submission received: 15 November 2018 / Revised: 11 December 2018 / Accepted: 20 December 2018 / Published: 28 December 2018
(This article belongs to the Special Issue Natural Killer Cells and Cancer Therapy)

Abstract

:
Metastasis represents the leading cause of cancer-related death mainly owing to the limited efficacy of current anticancer therapies on advanced malignancies. Although immunotherapy is rendering promising results in the treatment of cancer, many adverse events and factors hampering therapeutic efficacy, especially in solid tumors and metastases, still need to be solved. Moreover, immunotherapeutic strategies have mainly focused on modulating the activity of T cells, while Natural Killer (NK) cells have only recently been taken into consideration. NK cells represent an attractive target for cancer immunotherapy owing to their innate capacity to eliminate malignant tumors in a non-Major Histocompatibility Complex (MHC) and non-tumor antigen-restricted manner. In this review, we analyze the mechanisms and efficacy of NK cells in the control of metastasis and we detail the immunosubversive strategies developed by metastatic cells to evade NK cell-mediated immunosurveillance. We also share current and cutting-edge clinical approaches aimed at unleashing the full anti-metastatic potential of NK cells, including the adoptive transfer of NK cells, boosting of NK cell activity, redirecting NK cell activity against metastatic cells and the release of evasion mechanisms dampening NK cell immunosurveillance.

1. Introduction

Natural Killer (NK) cells are cytotoxic immune cells with an innate capacity for eliminating transformed cells in a non-major histocompatibility complex (MHC) and non-tumor antigen-restricted manner [1]. The activation of NK cells depends on a balance of signals provided by inhibitory and activating receptors that detect changes in the patterns of expression of their ligands on the surface of tumor cells. Inhibitory NK cell receptors recognize self-proteins and transmit inhibitory signals that maintain tolerance to normal cells [1,2]. Killer cell immunoglobulin-like receptors (KIRs) and the heterodimer CD94-Natural Killer Group 2A (NKG2A) are inhibitory receptors that recognize self-MHC class I molecules, whereas other inhibitory receptors, such as T cell immunoreceptor with Ig and ITIM domains (TIGIT) receptor, bind to other self-molecules [2,3,4]. Transformed cells frequently downregulate MHC class I molecules, thereby avoiding recognition by CD8+ cytotoxic T cells, but concomitantly inducing the activation of NK cells by missing self-recognition. Activating receptors, including, but not limited to, killer cell lectin-like receptor K1 (KLRK1—best known as NKG2D), DNAX accessory molecule-1 (CD226—best known as DNAM-1) and the natural cytotoxicity receptors NKp46, NKp44, and NKp30, recognize stress-inducible ligands on tumor cells that are scarcely expressed in their normal counterparts [2,3,4,5]. Natural killer group 2D (NKG2D) is a particularly relevant activating receptor, which recognizes a group of stress-inducible molecules termed MHC class I polypeptide-related sequence A and B (MICA and MICB) and UL16 binding protein molecules (ULBP1-6), which are restrictedly expressed on stressed and transformed cells [6,7]. Thus, by this complex pattern of receptors, NK cells may kill a broad range of cancer cells. Indeed, the engagement of activating receptors by tumor-expressed ligands, along with a lack of co-engagement of an appropriate number of inhibitory receptors, results in the exocytosis of cytotoxic granules containing perforin and granzymes that induce apoptotic cell death of the target cells. Additionally, NK cells can eliminate target cells through Fas ligand and tumor necrosis factor (TNF)-related apoptosis-inducing signals [2,3]. Finally, NK cells may also kill tumor cells bound by specific IgG antibodies through FcγRIII receptors (also named as CD16s), a process known as antibody-dependent cellular cytotoxicity (ADCC). The latter is a relevant process underlying the therapeutic activity of certain monoclonal antibodies [8]. NK cells also regulate the innate and adaptive immune response through the secretion of cytokines with potent antitumor activity, such as interferon-gamma (IFN-γ).

2. The Metastatic Cascade

The metastatic cascade involves tumor cell detachment from their neighbors, local invasion of surrounding tissues—as either a singular cells or groups of tumor cells—and the tumors entrance into the nearby pre-existing or neo-formed vasculature. In this scenario, typical features of such aggressive tumor cells are the partial loss of epithelial markers and the acquisition of a mesenchymal-like phenotype typically associated with migratory and invasive properties, which happens during the so-called epithelial-to-mesenchymal transition (EMT) (see below) [9]. Upon intravasation into the vascular bed, single or circulating tumor cell (CTC) clusters travel throughout the circulatory system until they lodge at secondary sites and extravasate, via transendothelial migration, through capillary walls. Once at the target site, disseminated tumor cells (DTCs) can survive for a short period or for decades as dormant and indolent entities before micrometastasis proliferation is triggered, giving rise to overt macrometastasis and organ colonization [10].
Metastasis formation is an extremely selective process wherein a progressive decimation of the tumor population occurs. The metastatic cascade may be envisioned as a sequence of selective microenvironments demanding specific malignant attributes. Among these tumor-extrinsic factors, the host’s anti-tumor immune response is a major hindrance that cancer cells must avoid to successfully seed distant metastases. In fact, owing to cancer immunosurveillance, a myriad of immune components can be detected at every step of the metastatic process as a forced companion of tumor cells, an immune contexture that constitutes a strong prognostic factor of the outcome of the disease [11]. This pairing results in a complicated bidirectional relationship whereby the immune system shapes the immunogenicity of the developing tumor by operating both tumor-promoting and tumor-obstructive mechanisms throughout the metastatic process.

3. NK Cells and the Immunosurveillance of Metastasis

Accumulating data evidence a central role of NK cells in the control of metastasis in preclinical models [12]. Nevertheless, NK cells show a poor capacity for infiltrating and eliminating large solid tumors in humans [13,14,15,16,17]. Indeed, NK cells are scarcely present in the tumor bed of colorectal cancers, despite the high levels of local chemokines and the normal numbers of NK cells observed in adjacent mucosa [18]. Moreover, tumor-infiltrating NK cells are frequently enriched by poorly cytotoxic CD56bright or poorly functional CD56dim NK cells with a decreased capacity for eliminating tumor cells [19,20,21,22]. Furthermore, NK cells are preferentially located in the stroma, rather than in direct contact with tumor cells in colorectal carcinomas, melanomas and Gastro-Intestinal Stromal Tumors (GISTs) [17,23]. It has recently been reported that tumor hyperploidy induced NK cell antitumor activity occurs mainly through the activation of NKG2D and DNAM-1 receptors [24] and that the level of aneuploidy negatively correlated with the NK cell infiltration of solid tumors, providing rational support for these observations [25]. The abovementioned observations notwithstanding, NK cells are sometimes successful in infiltrating primary tumors. The level of NK cell infiltration was associated with better prognoses of metastatic colorectal carcinomas and metastatic renal cell carcinomas [26,27]. There is a correlation between the density of infiltrating NK cells, metastasis, and the patient’s prognosis in terms of esophageal cancer, gastric carcinomas, and prostate cancer [28,29,30]. Furthermore, patients with prostate carcinomas with high NKp46 and NKp30 expression on intra-tumoral NK cells showed a high tendency toward no progression 1 year after surgery [22,31]. The levels of NKp46+ cell infiltration inversely correlated with the presence of metastases upon the diagnosis of gastrointestinal sarcoma tumors [32].
Along the same thread, the immune evasion mechanisms developed by tumor cells, including the downregulation of NK cell activating receptors or the shedding of NK cell activating ligands (NKARLs) as soluble proteins, are associated with poor prognosis and metastasis of several types of cancer (see below). Similarly, dendritic cell (DC)-induced IFN-γ production by NK cells, as well as the expression of the NKp30 splice variant (immunosuppressive NKp30C variant vs. immunostimulatory NKp30A and B variants), was found to constitute an independent predictor of the long term survival of patients with advanced gastrointestinal stromal cancer treated with Imatinib Mesylate [33] and those with neuroblastoma treated with chemotherapy [34]. NK cells have also been involved in the control of metastasis after cancer surgery [35]. Major surgery induces significant immunosuppression, mainly associated with the profound suppression of NK cells affecting both cytotoxic activity and cytokine production, thereby increasing the risk of metastasis in preclinical models and patients with cancer [35].
NK cells are present in lymph nodes and blood and may participate in the immunosurveillance of disseminated cells in the metastatic cascade. In melanoma patients, NK cells with cytotoxic capacity are recruited or generated in tumor-draining lymph nodes suggesting a potential role in metastasis immunosurveillance, although their prognostic significance has not yet been established [21,36,37]. Furthermore, the majority of tumor cells that enter into circulation are eliminated by NK cells within the first 24 h [38]. Hence, the number of circulating NK cells correlated with the number of CTCs and metastasis in triple-negative breast cancer and pancreatic cancer patients [39,40]. Conversely, and consistent with the immunoediting process, the activity of circulating NK cells generally decreases as the cancer progresses, thereby increasing the risk of metastatic disease in prostate cancer pharyngeal, carcinoma and gastric cancer patients [31,41,42]. These data, along with supporting the prominent role of NK cells in the control of metastasis, display an opportunity for immunotherapeutic intervention in cancer patients.

4. EMT and NK Cells

EMT is one of the key steps of the metastatic cascade whereby carcinoma cells lose epithelial characteristics and acquire a mesenchymal phenotype, which endows tumor cells with invasiveness, motility and resistance to apoptosis [9]. In addition, activation of the EMT program impinges on the immunomodulatory properties and the immunogenicity of cancer cells [43,44]. For instance, we reported that the induction of EMT in a colorectal cancer cell line, by the forced expression of the master regulator of the EMT process Snail family transcriptional repressor 1 (SNAI1; best known as Snail), is associated with the increased immunogenicity of tumor cells in vitro [45]. Furthermore, immunohistochemistry analyses showed that in well-differentiated colorectal tumors preserving the epithelial architecture, MICA/B was confined to the luminal part of the epithelial layer, thereby impeding the interaction with NKG2D-expressing immune cells located in the basal part of the epithelial tissue [45]. Conversely, disruption of the epithelial phenotype during EMT was associated with the loss of the polarized expression of MICA/B, likely rendering tumor cells sensitive to immune elimination. In fact, a significant decrease of MICA/B expression and a marked increase of NKG2D+ tumor-infiltrating lymphocytes were observed in less-differentiated tumors with mesenchymal characteristics. These suggest that the EMT process may constitute a cancer immune checkpoint and that colorectal tumor cells must develop mechanisms to escape from NKG2D-mediated immune responses in order to progress through the metastatic process [45,46]. Along similar lines, prostate cancer cells were shown to strongly downregulate MHC class I expression during TGF-β/EGF-induced EMT, rendering tumor cells resistant to CD8 T cell-mediated 0cytotoxicity, but increasing their susceptibility to NK cells in a process independent of Snail transcription factor [47]. In lung cancer, TGF-β-induced EMT confers increased tumor cell susceptibility to NK cell killing activity through the induction of E-cadherin and cell adhesion molecule 1 (CADM1) expression [48]. Accordingly, transplantation of epithelial tumors expressing neu oncogene into syngeneic mice induced an immune-mediated rejection of cancer cells [49]. Consistent with cancer immunoediting, these mice subsequently relapsed with tumors enriched in neu-negative variant cancer cells with a mesenchymal phenotype. These data together suggest that the EMT transdifferentiation may be an immune checkpoint crucial to the control of metastasis by NK cells.
NK cells may control the development of cancer, principally during the initial steps of malignant transformation, but, in a specific tumorigenic context and mainly in the last stages of tumor transformation, they may also favor tumor progression [23]. In line with this, Huergo-Zapico and colleagues recently showed the unexpected role of NK cells in the promotion of pro-metastatic features of melanoma cells through the triggering of the EMT process, thereby promoting a tumor phenotype switching from proliferative to invasive [50]. NK cells were found to increase tumor resistance to NK cell-mediated killing by inducing the expression of NK cell-inhibitory MHC class I molecules on the surface of melanoma cells. These changes were mostly dependent on NKp30 or NKG2D engagement and release of IFN-γ and TNF-α by NK cells. Worth noting was the expression of the inhibitory immune checkpoint programmed death ligand 1 (CD274—best known as PD-L1), induced by IFN-γ produced by activated immune cells, including NK cells, which constitutes a prominent mechanism of tumor “adaptive resistance” to immunosurveillance [51]. Interestingly, PD-L1 expression has been reported to be downregulated by the EMT-repressor microRNA-200 (miR-200) in Non-Small-Cell Lung Carcinoma (NSCLC) [52,53] and breast carcinoma cells [54], hence unveiling a link between inhibitory immune checkpoint expression and the acquisition of a mesenchymal phenotype in cancer. Accordingly, a number of studies demonstrate a correlation between PD-L1 expression and EMT score in several types of malignancies, such as lung cancer and breast carcinomas, suggesting that the group of patients in whom malignant progression is driven by EMT activators may respond to treatment with PD1/PD-L1 antagonists [53]. Overall, the EMT process may have crucial influence over the immunosurveillance of cancer mediated by NK cells, hence opening a potential new window for therapeutic intervention.

5. Metastasis and Evasion of NK Cell Surveillance

Immune evasion is a hallmark of cancer and metastatic cells develop the most refined de facto immunosubversive mechanisms [55]. Thus, in patients with advanced cancers, tumor cells exhibit decreased expression of NKARLs. Consequently, metastatic cancer cells are more likely to escape from NK cell antitumor surveillance, thereby increasing the probability of malignant dissemination. A manifold program of suppressive mechanisms has been reported to reduce NKARL expression in cancer, including, but not limited to, the proteolytic shedding of soluble NKARLs as well as epigenetic changes involving histone deacetylation [56] or microRNA overexpression [57,58,59]. Shedding of soluble MICA depends on its interaction with the chaperon molecule protein disulfide isomerase family A member six (PDIA6—best known as ERp5) on the surface of tumor cells [60]. ERp5 forms a transitory disulphide bond with MICA, which induces a conformational change in its α3 domain. This allows the proteolytic cleavage of MICA by proteases, including ADAM10, ADAM17 and MMP14, which are overexpressed in cancer cells [61,62]. ERp5 that had been identified as a metastasis-promoting factor in a mouse model of breast cancer was highly detected in human samples of invasive breast cancer [63]. Further, membrane ERp5 was functionally associated with soluble MICA shedding in chronic lymphocytic leukemia patients [64] and enhanced levels of soluble MICA correlated with membrane ERp5 expression in myeloma and lymphoma cells [65,66].
It has been widely reported that low cell surface expression of MICA/B or elevated sera levels of soluble MICA and MICB correlate with metastasis in different types of cancer [67,68,69,70,71,72,73,74]. Elevated sera levels of soluble ULBP2 are an indicator of progression in melanoma patients [75]. Low expression of ULBP4 also favors metastasis in nasopharyngeal carcinomas [76]. By contrast, the tumor tissue expression levels of B7-H6, a ligand of the activating NCR receptor NKp30, correlated with the metastasis and cancer progression of ovarian cancer [77]. Meanwhile, the serum concentration of soluble B7-H6 correlated with the down-regulation of NKp30, bone marrow metastasis, and chemo-resistance in high-risk neuroblastoma patients [34]. NK cells also expressed inhibitory receptors, such as PD1, which participated in the suppression of NK cell activity in advanced and metastatic patients [78,79,80].
Immunosuppressive factors also played a crucial role in the evasion of NK cell-antitumor activity. TGF-β is a key immunosuppressive cytokine produced by several cell subtypes, including tumor cells, Tregs, stromal cells and myeloid-derived suppressor cells (MDSCs). The pleiotropic activity of TGF-β is of pivotal relevance for the EMT process and cancer progression and, at the same time, is a major suppressor of NKG2D expression, NK cell cytotoxicity, and IFN-γ production of and metastasis in advanced cancer patients [22,81,82,83]. Furthermore, TGF-β-dependent signaling in the tumor microenvironment promotes the conversion of NK cells into type 1 innate lymphoid cell (ILC1) populations, which are unable to control tumor growth and metastasis [84]. In addition, TGF-β and other immunosuppressive molecules produced by tumor cells may recruit and activate immune cells including Tregs [85], MDSCs [86], CD11b+Ly6G+ neutrophils [87], and indoleamine 2,3-dioxygenase 1 (IDO1)-expressing DCs [88] that further suppress NK cell activity.
Metastatic cells develop specific mechanisms that allow them to survive outside of the tumor nest and to cross the blood to extravasate into distant tissues. Blood is a challenging territory for metastatic cells and there is increasing evidence that the survival of CTCs is greatly enhanced by platelets. Thus, thrombocytopenia inhibits tumor growth and metastasis in mice, whereas depletion of NK cells reverts this anti-metastatic phenotype [89]. Platelets favor the escape of CTCs from NK cells via at least five different mechanisms. They (1) physically shield CTCs from immune recognition [90], (2) secrete immunosuppressive factors including TGF-β [91], (3) transfer MHC class I molecules to cancer cells to establish a state of pseudo-self-tolerance [92], (4) induce the shedding of NKG2D ligands [93], and (5) express NK cell inhibitory ligands such as glucocorticoid-induced TNF-related ligand (GITRL) [94]. Along with the above described immunosuppressive environment, tumor infiltrating lymph nodes and circulating NK cells from patients with metastasis display reduced levels of NK cell activating receptors and, consequently, decreased cytotoxicity and IFN-γ production compared with healthy donors or patients with less-advanced diseases [19,20,95].
To conclude, immune evasion is a major obstacle in the development of effective anticancer therapies. The recent advances in understanding the immune evasion mechanisms developed by disseminated cells are opening new opportunities for novel and more effective cancer immunotherapies.

6. Immunotherapy in the Treatment of Metastasis

The treatment of cancer metastasis is one of the greatest challenges in medicine. Indeed, metastasis is responsible for more than 90% of cancer-associated mortality. Conventional therapeutic strategies, including surgical resection, chemotherapy and radiotherapy are relatively efficient in the elimination of primary tumors, but show limited efficacy in the elimination of metastases. Although immunotherapies are a promising strategy against cancer metastasis, current immunotherapies are mainly focused on T cells, and many toxicity and efficacy complications remain to be solved. NK cells have potent anti-tumor and regulatory activity and are key regulators of T cell-mediated immunity [96], which supports the idea that NK cell immunotherapy may be an alternative or complementary to T cell immunotherapy (Table 1, Figure 1).

6.1. Adoptive Transfer of NK Cells

NK cells are considered ideal targets for adoptive transfer owing to their capability to eliminate tumor cells in a non-MHC and non-tumor antigen-restricted manner. However, the number and the cytotoxic capacity of circulating NK cells is limited and, therefore, it is necessary to expand and activate NK cells before their infusion into patients. In hematopoietic stem cell transplantation, blood NK cells recover very early after transplantation and the number of NK cells correlates with the clinical outcome [97,98]. Initial clinical trials in patients with metastatic cancer showed that the transfer of expanded autologous NK cells in monotherapy was well tolerated, although no clinical response was observed [99,100,101]. Nonetheless, the efficacy of allogeneic NK cells in controlling human cancers is supported by more compelling evidence. The transfer of allogenic NK cells in hematological malignancies, and, particularly, the adoptive transfer of KIR-ligand mismatched donors in patients with Acute Myeloid Leukemia (AML) has shown impressive clinical results [102]. The lack of engagement of inhibitory KIR receptors, by their MHC class I ligands in these patients, showed how the full potential of NK cells may translate to outstanding clinical responses. In solid tumors, initial clinical studies of allogenic NK cell adoptive transfer have shown that this therapy is safe and well-tolerated and may result in a clinical response in metastatic cancer patients [103,104,105,106,107,108]. Nevertheless, the therapeutic adoptive transfer of NK cells in the patient has not yet been definitely developed. There is very limited experience in terms of the adequate conditioning regimen, the source of NK cells, the NK cell activation, and the expansion protocols, or the need to combine therapies to unleash the full anti-tumor potential of NK cells. Thus, many procedures to obtain clinical grade NK cells from peripheral blood, bone marrow, or cord blood have been developed and their quality may vary. In addition, many different approaches to expand and activate NK cells exist. Hence, the priming of NK cells with CTV-1 leukemia cell line lysate CNDO-109 results in enhanced cytotoxicity against NK cell-resistant cell lines and NK cell activation in high-risk patients with AML in a phase I clinical trial [109]. NK cells can also be pre-activated with IL-12, IL-15 and IL-18. This approach has been used to generate the so-called Cytokine-induced memory-like (CIML) NK cells with potent in vitro and in vivo anti-leukemia responses along with improved clinical responses in patients with AML in the context of a first-in-human phase I clinical trial [110]. As indicated in Table 1, a number of different NK-based therapies have been studied or are under consideration. These approaches (and their effectiveness) greatly vary depending on the tumor type, tumor cell phenotype, patient, and feasibility of the treatments.
CAR T cells are emerging as a new revolution in the treatment of cancer [111]. Similarly, a novel way to boost the therapeutic activity of NK cell transfer is to manipulate and redirect NK cell antitumor activity by chimeric antigen receptor (CAR) engineering. NK cells should be considered an alternative to T cells since they are non-MHC-restricted “serial killers” of tumor cells and, therefore, can be used as universal donors for cancer immunotherapy. Despite the fact that the evaluation of CAR NK cells in solid tumors and metastases are mostly in preclinical studies and initial clinical trials [112,113], CAR NK cells may theoretically show several advantages over CAR T cells [114]. First, CAR NK cells are considered safer than their T cell counterparts. Owing to the fact that NK cells are short-living cells, CAR NK cell therapy is not expected to be associated with long-term problems characteristic of CAR T cell therapy, such as the risk of autoimmunity or malignant transformation. Furthermore, the so-called “cytokine storm” initiated by CAR T cells may be severe, even fatal, and is largely mediated by the production of pro-inflammatory cytokines by T cells. NK cells produce cytokines, including IFN-γ and GM-CSF, with a lower toxicity profile and are considered safer than those produced by T cells. A recent phase I clinical trial evaluating the safety and efficacy of using CAR NK-92 cells targeting CD33 in patients with relapsed and refractory AML showed no major adverse effects, indicating that CAR NK cells may be a safe alternative for CAR T cells [115]. CAR NK cells could be also more efficient in terms of eliminating tumor cells than CAR T cells as they are endowed with spontaneous cytotoxic activity provided by an array of activating receptors that may target cancer cells in an MHC and antigen-unrestricted manner, which may potentiate the activity of the CAR. Additionally, NK cells are endowed with intrinsic ADCC activity that enables the use of combination therapies involving CAR NK cells and monoclonal antibodies. Nevertheless, there are some drawbacks to the use of CAR NK cells [114]. NK cells are difficult to obtain, expand and manipulate as they show quite low transfection efficiency even when viral vectors are used [116].
NK cell lines, such NK-92 cells, may be an alternative source of NK cells for cancer immunotherapy. They have several advantages over autologous or allogenic NK cells. NK-92 cells are an unlimited, homogeneous and well-defined population of highly active NK cells that can be obtained at a low cost [117]. However, NK-92 cells have several disadvantages, such as their lack of typical NK cell activating receptors, including CD16, NKp44, and NKp46, and, as with any other tumor cell line, NK-92 cells carry multiple cytogenetic abnormalities and are latently infected by Epstein-Barr virus [118]. Hence, NK cell lines have to be irradiated before infusion for safety. Unmodified NK-92 cells have been employed to treat patients with advanced cancers, with encouraging responses observed in patients with advanced lung cancer [119]. Furthermore, NK-92 cells may constitute a suitable source of NK cells for CAR engineering since they show a transfection efficiency of about 50% even with non-viral methods. Preclinical data demonstrate that CAR NK-92 cells targeting the receptor tyrosine kinase ErbB2/HER2 or the epidermal growth factor receptor (EGFR) protect mice against glioblastoma [120,121,122]. In a preclinical study, CAR NK-92 cells targeting EGFR combined with oncolytic herpes simple virus 1 have been shown to be a novel strategy for breast cancer brain metastases [123]. CAR NK-92 cells targeting ERbB2/HER2 also reduced lung metastasis in a renal carcinoma murine model, showing their potential in relation to the treatment of the disseminated disease [113].

6.2. Cytokine-Based NK Cell Therapy

IL-2 was one of the first cytokines employed to treat human cancers. It showed great therapeutic potential in controlling metastatic diseases owing to its pleiotropic capability of boosting T and NK cell antitumor activity [124]. Although high doses of IL-2 improved the survival of a fraction of treated patients with metastatic renal cancer and melanomas [125], it caused life-threatening toxicities including vascular leak syndrome (VLS) [124]. Furthermore, both high- and low-dose IL-2 therapy induced the expansion in patients of immunosuppressive CD4+CD25+Foxp3+ Treg cells expressing the high affinity IL-2 receptor α-chain (CD25) [126,127,128]. Thus, IL-2 monotherapy is not the optimal or standard treatment for these patients and efforts have been made to further improve the efficacy of IL-2 therapy in combination with other anticancer immunotherapy regimens [124]. Early combination therapies were initiated incorporating immune cells such as lymphokine activated killer (LAK) cells and T cells. Co-administration of LAK cells with IL-2 yielded a clinical response rate of 20 to 35%, however, mostly with a transient response in solid tumors [129]. Subsequently, IL-2 was also used in conjunction with adoptive T cell therapy, significantly improving efficacy [130,131,132]. Similarly, IL-2 is also frequently associated with the adoptive transfer of NK cells for the promotion of the expansion of NK cells in vivo.
Cytokines that activate NK cell development, survival and activity without activating Tregs (such as IL-12, IL-15, IL-18, and IL-21) are now being studied [133]. A compelling body of studies supports the idea that IL-15 is a cytokine with a high potential to be harnessed in cancer therapy [134]. First, while IL-15 shared the strong stimulatory effect on cytotoxic immune cells with IL-2, treatment with IL-15 did not result in immunosuppression and did not cause vascular leak syndrome in mice or nonhuman primates [135,136]. Second, preclinical studies demonstrated that IL-15 induced prolonged expansion and activation of NK cells and CD8 memory T cells, therefore resulting in tumor regression, decreased metastasis, and increased survival in several experimental models [135,136,137,138,139,140]. These observations supported the use of IL-15 as an alternative treatment in patients with metastatic malignancies and led to the implementation of IL-15-based therapies in several clinical trials (source https://clinicaltrials.gov/). These involved the administration of IL-15 alone or in combination with NK transfer or chemotherapy in patients with solid tumors and hematological malignancies [134]. A hyper-proliferation and an increased number of circulating NK cells was reported in the first in-human phase I clinical trial of IL-15 in patients with metastatic malignant melanoma and metastatic renal cell cancer [141]. Preliminary antitumor evaluation showed no objective responses, but the clearance of lung metastases in two patients was observed [141]. In a phase I/II clinical trial, IL-15-stimulated NK cells induced a clinical response in four out of six pediatric patients with solid refractory tumors [142]. Despite these promising clinical results, therapies involving IL-15 must deal with short in vivo half-life of the cytokine. Several approaches have been developed to overcome this limitation, largely by generating super agonist IL-15/IL-15Rα conjugates that exhibit greater activity than that of naïve IL-15 [134].
Other cytokines that activate NK cells without stimulating Tregs are being evaluated. IL-12 and IL-18 showed, in monotherapy, rather limited anti-tumor activity in humans, while a combination of IL-12/IL-18/IL-15 induced sustained in vitro and in vivo effector functions in human and mice NK cells respectively [143]. IL-21 represents another interesting cytokine for cancer immunotherapy as it induces the antitumor activities of CD8 T cells, NK cells, and NKT cells [144,145]. It showed antitumor activity in several preclinical cancer models [146,147,148,149] and in a phase II study in patients with metastatic melanomas [150]. However, at high concentrations, IL-21 can also lead to dose-limiting side effects including 3/4 grade granulocytopenia and liver toxicity. It may also perform pro-inflammatory activities in many tissues and promote colitis-associated colon cancer [151]. Thus, a more profound understanding of the biology of IL-21 is needed to ensure its maximal therapeutic benefit.
Multiple experimental and preclinical studies reporting the effectiveness of several drugs in the boosting of NK cell antitumor activity have been published, including that of inhibitors of TGF-β, antagonists of the high adenosine receptor A2, immunomodulatory drugs such as lenalidomide, DNA complexes (CpG), demethylating agents, histone deacetylases inhibitors, etc. [12]. Despite such efforts, the clinical potential of these NK cell-activating strategies in the treatment of metastasis remains to be established.

6.3. Modulation of NK Cell Receptors or Ligands

Activating and inhibitory receptors or their ligands can be targeted by monoclonal antibodies (mAbs) to increase therapeutic NK cell activity. The most widely used strategy in cancer immunotherapy is to employ tumor-specific antibodies that promote ADCC through the ligation of CD16 receptors on NK cells. Rituximab (mAb to the B cell marker CD20), trastuzumab (mAb to ErbB2/HER2) and cetuximab (mAb to EGFR) have demonstrated marked efficacy in the treatment of various solid and hematological tumors [8]. No therapeutic effect was achieved in mice deficient in Fc receptors [152] and polymorphisms in the genes encoding these receptors modulated their clinical response [153,154], suggesting that Fc receptors and ADCC play a crucial role in the therapeutic activity of these mAbs.
Trastuzumab, the first antibody developed against a growth factor, was successfully introduced in the treatment of HER2+ breast cancers, which represent 15–20% of this tumor type. Trastuzumab combined with chemotherapy has dramatically improved the survival of HER2+ patients [155] and even trastuzumab in monotherapy improves the clinical response and survival of patients with metastatic breast cancer [156]. However, high proportions of patients relapse and experience brain metastasis after treatment with trastuzumab. Currently, several combinations of strategies for improving the efficacy of mAbs are being tested. For example, several clinical trials are now assessing the safety and efficacy of the combination of autologous NK cells with trastuzumab in patients with HER2+ breast or gastric cancers (source https://clinicaltrials.gov/).
A step toward improving ADCC activity for cancer immunotherapy is the development of the so-called bispecific or trispecific antibodies. Bispecific antibodies can bind simultaneously two different antigens, which leads to a wide range of applications including redirecting T cells or NK cells to tumor cells. The so-called bispecific trifunctional antibodies have dual specificity while preserving ADCC potential. The most successful example of this class of new drugs is catuxomab, which consists of one “Fab” recognizing the epithelial cell adhesion molecule (EpCAM), the other "Fab" recognizing a CD3 antibody, and the Fc region binding to NK cells or macrophages. Catuxomab is now approved in the European Union for the treatment of patients with malignant ascites from EPCAM-positive tumors [157].
Targeting NKG2D ligands may also stimulate NK cell activity. Chronic exposure to MICA/B downregulates NKG2D expression at the surface of NK and CD8 T cells, suppressing their activity. The MICA/B targeting antibody IPH4301 mediates potent NK cell-stimulatory effects in vitro and in murine models and prevents NKG2D downregulation [12]. Recently, antibodies targeting the α3 domain of MICA, preventing soluble MICA/B shedding, have been shown to inhibit tumor growth in multiple murine models and to reduce human melanoma metastases in a humanized mouse model, mainly by NK cell activation through NKG2D and CD16 receptors [158]. These antibodies are good candidates for clinical testing.

6.4. Targeting Inhibitory Receptors

Blocking the inhibitory pathways dampening NK cell function in cancer is an encouraging strategy for NK cell immunotherapy [159] (Figure 1). Checkpoint inhibitors are currently the most promising drugs for cancer immunotherapy. Treatment with anti-CTLA4 and PD1/PD-L1 antibodies showed impressive results in patients with metastatic cancer, causing durable tumor regression in a significant group of patients [160]. A combination of both checkpoint inhibitors had cooperative clinical effects [161]. The clinical benefit of these antibodies is thought to mainly rely on the reactivation of exhausted T cells. However, NK cells also express checkpoint molecules and, consequently, they may also contribute to the observed therapeutic benefit. In fact, the PD1/PDL1 blockade in several mouse models elicited a strong NK cell-mediated response that was indispensable for the full therapeutic effect of immunotherapy [162]. Further, NK cells positively regulated the abundance of the stimulatory DCs through the production of cytokines such as FLT3LG, CCL5 and XCL1 in mouse and human tumors. Both infiltrating NK cells and stimulatory DCs correlated with increased overall survival and responsiveness to anti-PD1 immunotherapy in melanoma patients [80,163]. PD1 was also shown to inhibit NK cell activity in multiple myeloma and digestive cancers, particularly in patients with advanced diseases and PD1 blockade restored NK cell cytotoxicity and IFN-γ production [78,79]. These recent data suggest that NK cells may contribute to checkpoint inhibitor clinical efficacy, particularly in hematological malignancies and patients with metastases. This matter clearly warrants further investigation.
Overexpression of MHC class I molecules in cancer cells inhibits NK cell activation through the interaction with KIR receptors and blocking this interaction may have therapeutic benefits for cancer patients. IPH2101 is an antibody that binds to the high affinity inhibitory receptors KIR2DL-1, KIR2DL-2, and KIR2DL-3, blocking their interaction with HLA-C group 1 and 2 allotypes, which boosts NK cell anti-tumor activity in preclinical studies [164]. Nevertheless, IPH2101 failed to demonstrate any clinical benefit in patients with smoldering multiple myeloma in a clinical trial, probably due to an unexpected NK cell inhibitory effect [165]. Monalizumab, an antibody that blocks NKG2A-HLA-E interaction, has been shown to induce NK cell antitumor activity against leukemia cells from patients with chronic lymphocytic leukemia [166] and is currently being tested for efficacy and efficiency in several hematological malignancies and advanced solid tumors (source: https://clinicaltrials.gov).
The inhibitory receptors TIGIT and CD96 (TACTILE) have recently emerged as new checkpoints for cancer immunotherapy. They inhibit NK cell activity and counterbalance CD226 activation of NK cells. TIGIT was associated with NK cell exhaustion in tumor bearing mice and patients with colon cancer and TIGIT blockade promoted NK cell anti-tumor immunity in several mouse models [167]. CD96 blockade also inhibited experimental metastases in three different murine models, the therapeutic effect of which depended on NK cells, DNAM-1, and IFN-γ, but was independent of Fc receptors [168]. Furthermore, blocking CD96 in Tigit−/− mice significantly reduced experimental and spontaneous metastases. Finally, the TIM-3 expression on NK cells was upregulated on metastases and correlated with a poor prognosis in patients with lung adenocarcinoma, melanomas, and gastric cancer; and TIM-3 blockade restored NK cell anti-tumor activity ex vivo [169,170,171].
To conclude, a more profound understanding of the evasion mechanisms developed by tumor cells is needed in order to develop new strategies for cancer immunotherapy. Due to the wide variety of immune evasion mechanisms specifically developed by each type of cancer, it is likely that the abovementioned, novel NK cell activating therapies may only benefit a specific group of patients and a more personalized therapeutic approach might be required in the future.

7. Conclusions

Cancer metastasis is the leading cause of cancer-related death. Current cancer therapies, including surgery, chemotherapy and radiotherapy, have shown a limited capacity for improving the survival of metastatic patients. Conversely, T cell-mediated immunotherapy has obtained impressive clinical results in some metastatic patients, particularly melanoma patients. Further studies aimed at overcoming the immunosuppressive tumor microenvironment and managing the adverse effects induced by possible self-reactive T cells are needed to fully exploit the therapeutic potential of these cells. As an alternative, or even as a complement, the use of NK cell immunotherapy shows great potential in the treatment of metastatic patients. NK cells harbor innate and potent anti-metastatic activity, which is independent of MHC restriction and tumor antigen expression, and they may be envisioned as a universal source of antitumor cells for cancer immunotherapy. Now, the challenge is to translate the increasing amount of basic research into new therapeutic strategies. Initial NK cell immunotherapies for metastatic patients have shown encouraging clinical results and some NK cell immunotherapies could be associated with a better toxic profile than that of T cells. Nevertheless, further experimental and clinical studies have to be performed in order to completely understand the antitumor and anti-metastatic properties of NK cells and the most effective way to fully unleash their therapeutic potential.

Author Contributions

All the authors participated in the preparation, writing and revision of the manuscript.

Funding

This work was supported by the Spanish grant of Instituto de Salud Carlos III (PI16/01485) and FEDER European Union. S.L.H. holds a Severo Ochoa Grant (BP14-150). The funders had no role in the study design, data collection, or analysis, the decision to publish, or the preparation of the manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Chiossone, L.; Dumas, P.Y.; Vienne, M.; Vivier, E. Natural killer cells and other innate lymphoid cells in cancer. Nat. Rev. Immunol. 2018, 18, 671–688. [Google Scholar] [CrossRef] [PubMed]
  2. Sun, J.C.; Lanier, L.L. NK cell development, homeostasis and function: Parallels with CD8(+) T cells. Nat. Rev. Immunol. 2011, 11, 645–657. [Google Scholar] [CrossRef] [PubMed]
  3. Cerwenka, A.; Lanier, L.L. Natural killer cell memory in infection, inflammation and cancer. Nat. Rev. Immunol. 2016, 16, 112–123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Moretta, L.; Locatelli, F.; Pende, D.; Sivori, S.; Falco, M.; Bottino, C.; Mingari, M.C.; Moretta, A. Human NK receptors: From the molecules to the therapy of high risk leukemias. FEBS Lett. 2011, 585, 1563–1567. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Moretta, A.; Bottino, C.; Vitale, M.; Pende, D.; Cantoni, C.; Mingari, M.C.; Biassoni, R.; Moretta, L. Activating receptors and coreceptors involved in human natural killer cell-mediated cytolysis. Annu. Rev. Immunol. 2001, 19, 197–223. [Google Scholar] [CrossRef] [PubMed]
  6. Lopez-Soto, A.; Huergo-Zapico, L.; Acebes-Huerta, A.; Villa-Alvarez, M.; Gonzalez, S. NKG2D signaling in cancer immunosurveillance. Int. J. Cancer 2015, 136, 1741–1750. [Google Scholar] [CrossRef] [PubMed]
  7. Huergo-Zapico, L.; Acebes-Huerta, A.; Lopez-Soto, A.; Villa-Alvarez, M.; Gonzalez-Rodriguez, A.P.; Gonzalez, S. Molecular Bases for the Regulation of NKG2D Ligands in Cancer. Front. Immunol. 2014, 5, 106. [Google Scholar] [CrossRef]
  8. Weiner, L.M.; Surana, R.; Wang, S. Monoclonal antibodies: Versatile platforms for cancer immunotherapy. Nat. Rev. Immunol. 2010, 10, 317–327. [Google Scholar] [CrossRef]
  9. Brabletz, T.; Kalluri, R.; Nieto, M.A.; Weinberg, R.A. EMT in cancer. Nat. Rev. Cancer 2018, 18, 128–134. [Google Scholar] [CrossRef] [Green Version]
  10. Massague, J.; Obenauf, A.C. Metastatic colonization by circulating tumour cells. Nature 2016, 529, 298–306. [Google Scholar] [CrossRef] [Green Version]
  11. Mlecnik, B.; Bindea, G.; Kirilovsky, A.; Angell, H.K.; Obenauf, A.C.; Tosolini, M.; Church, S.E.; Maby, P.; Vasaturo, A.; Angelova, M.; et al. The tumor microenvironment and Immunoscore are critical determinants of dissemination to distant metastasis. Sci. Transl. Med. 2016, 8, 327. [Google Scholar] [CrossRef] [PubMed]
  12. Lopez-Soto, A.; Gonzalez, S.; Smyth, M.J.; Galluzzi, L. Control of Metastasis by NK Cells. Cancer Cells 2017, 32, 135–154. [Google Scholar] [CrossRef] [PubMed]
  13. Balsamo, M.; Vermi, W.; Parodi, M.; Pietra, G.; Manzini, C.; Queirolo, P.; Lonardi, S.; Augugliaro, R.; Moretta, A.; Facchetti, F.; et al. Melanoma cells become resistant to NK-cell-mediated killing when exposed to NK-cell numbers compatible with NK-cell infiltration in the tumor. Eur. J. Immunol. 2012, 42, 1833–1842. [Google Scholar] [CrossRef] [PubMed]
  14. Sconocchia, G.; Spagnoli, G.C.; Del Principe, D.; Ferrone, S.; Anselmi, M.; Wongsena, W.; Cervelli, V.; Schultz-Thater, E.; Wyler, S.; Carafa, V.; et al. Defective infiltration of natural killer cells in MICA/B-positive renal cell carcinoma involves beta(2)-integrin-mediated interaction. Neoplasia 2009, 11, 662–671. [Google Scholar] [CrossRef] [PubMed]
  15. Sconocchia, G.; Arriga, R.; Tornillo, L.; Terracciano, L.; Ferrone, S.; Spagnoli, G.C. Melanoma cells inhibit NK cell functions. Cancer Res. 2012, 72, 5428–5429. [Google Scholar] [CrossRef] [PubMed]
  16. Erdag, G.; Schaefer, J.T.; Smolkin, M.E.; Deacon, D.H.; Shea, S.M.; Dengel, L.T.; Patterson, J.W.; Slingluff, C.L., Jr. Immunotype and immunohistologic characteristics of tumor-infiltrating immune cells are associated with clinical outcome in metastatic melanoma. Cancer Res. 2012, 72, 1070–1080. [Google Scholar] [CrossRef] [PubMed]
  17. Stojanovic, A.; Cerwenka, A. Natural killer cells and solid tumors. J. Innate Immun. 2011, 3, 355–364. [Google Scholar] [CrossRef]
  18. Halama, N.; Braun, M.; Kahlert, C.; Spille, A.; Quack, C.; Rahbari, N.; Koch, M.; Weitz, J.; Kloor, M.; Zoernig, I.; et al. Natural killer cells are scarce in colorectal carcinoma tissue despite high levels of chemokines and cytokines. Clin. Cancer Res. 2011, 17, 678–689. [Google Scholar] [CrossRef] [PubMed]
  19. Peng, Y.P.; Zhu, Y.; Zhang, J.J.; Xu, Z.K.; Qian, Z.Y.; Dai, C.C.; Jiang, K.R.; Wu, J.L.; Gao, W.T.; Li, Q.; et al. Comprehensive analysis of the percentage of surface receptors and cytotoxic granules positive natural killer cells in patients with pancreatic cancer, gastric cancer, and colorectal cancer. J. Transl. Med. 2013, 11, 262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Saito, H.; Osaki, T.; Ikeguchi, M. Decreased NKG2D expression on NK cells correlates with impaired NK cell function in patients with gastric cancer. Gastric Cancer 2012, 15, 27–33. [Google Scholar] [CrossRef] [PubMed]
  21. Vuletic, A.; Jurisic, V.; Jovanic, I.; Milovanovic, Z.; Nikolic, S.; Konjevic, G. Distribution of several activating and inhibitory receptors on CD3(−)CD56(+) NK cells in regional lymph nodes of melanoma patients. J. Surg. Res. 2013, 183, 860–868. [Google Scholar] [CrossRef] [PubMed]
  22. Pasero, C.; Gravis, G.; Guerin, M.; Granjeaud, S.; Thomassin-Piana, J.; Rocchi, P.; Paciencia-Gros, M.; Poizat, F.; Bentobji, M.; Azario-Cheillan, F.; et al. Inherent and Tumor-Driven Immune Tolerance in the Prostate Microenvironment Impairs Natural Killer Cell Antitumor Activity. Cancer Res. 2016, 76, 2153–2165. [Google Scholar] [CrossRef] [Green Version]
  23. Cantoni, C.; Huergo-Zapico, L.; Parodi, M.; Pedrazzi, M.; Mingari, M.C.; Moretta, A.; Sparatore, B.; Gonzalez, S.; Olive, D.; Bottino, C.; et al. NK Cells, Tumor Cell Transition, and Tumor Progression in Solid Malignancies: New Hints for NK-Based Immunotherapy? J. Immunol. Res. 2016, 2016, 4684268. [Google Scholar] [CrossRef] [PubMed]
  24. Acebes-Huerta, A.; Lorenzo-Herrero, S.; Folgueras, A.R.; Huergo-Zapico, L.; Lopez-Larrea, C.; Lopez-Soto, A.; Gonzalez, S. Drug-induced hyperploidy stimulates an antitumor NK cell response mediated by NKG2D and DNAM-1 receptors. Oncoimmunology 2016, 5, e1074378. [Google Scholar] [CrossRef] [PubMed]
  25. Davoli, T.; Uno, H.; Wooten, E.C.; Elledge, S.J. Tumor aneuploidy correlates with markers of immune evasion and with reduced response to immunotherapy. Science 2017, 355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Donskov, F.; von der Maase, H. Impact of immune parameters on long-term survival in metastatic renal cell carcinoma. J. Clin. Oncol. 2006, 24, 1997–2005. [Google Scholar] [CrossRef] [PubMed]
  27. Coca, S.; Perez-Piqueras, J.; Martinez, D.; Colmenarejo, A.; Saez, M.A.; Vallejo, C.; Martos, J.A.; Moreno, M. The prognostic significance of intratumoral natural killer cells in patients with colorectal carcinoma. Cancer 1997, 79, 2320–2328. [Google Scholar] [CrossRef] [Green Version]
  28. Xu, B.; Chen, L.; Li, J.; Zheng, X.; Shi, L.; Wu, C.; Jiang, J. Prognostic value of tumor infiltrating NK cells and macrophages in stage II+III esophageal cancer patients. Oncotarget 2016, 7, 74904–74916. [Google Scholar] [CrossRef]
  29. Gannon, P.O.; Poisson, A.O.; Delvoye, N.; Lapointe, R.; Mes-Masson, A.M.; Saad, F. Characterization of the intra-prostatic immune cell infiltration in androgen-deprived prostate cancer patients. J. Immunol. Methods 2009, 348, 9–17. [Google Scholar] [CrossRef]
  30. Ishigami, S.; Natsugoe, S.; Tokuda, K.; Nakajo, A.; Che, X.; Iwashige, H.; Aridome, K.; Hokita, S.; Aikou, T. Prognostic value of intratumoral natural killer cells in gastric carcinoma. Cancer 2000, 88, 577–583. [Google Scholar] [CrossRef] [Green Version]
  31. Pasero, C.; Gravis, G.; Granjeaud, S.; Guerin, M.; Thomassin-Piana, J.; Rocchi, P.; Salem, N.; Walz, J.; Moretta, A.; Olive, D. Highly effective NK cells are associated with good prognosis in patients with metastatic prostate cancer. Oncotarget 2015, 6, 14360–14373. [Google Scholar] [CrossRef] [PubMed]
  32. Delahaye, N.F.; Rusakiewicz, S.; Martins, I.; Menard, C.; Roux, S.; Lyonnet, L.; Paul, P.; Sarabi, M.; Chaput, N.; Semeraro, M.; et al. Alternatively spliced NKp30 isoforms affect the prognosis of gastrointestinal stromal tumors. Nat. Med. 2011, 17, 700–707. [Google Scholar] [CrossRef] [PubMed]
  33. Menard, C.; Blay, J.Y.; Borg, C.; Michiels, S.; Ghiringhelli, F.; Robert, C.; Nonn, C.; Chaput, N.; Taieb, J.; Delahaye, N.F.; et al. Natural killer cell IFN-gamma levels predict long-term survival with imatinib mesylate therapy in gastrointestinal stromal tumor-bearing patients. Cancer Res. 2009, 69, 3563–3569. [Google Scholar] [CrossRef] [PubMed]
  34. Semeraro, M.; Rusakiewicz, S.; Zitvogel, L.; Kroemer, G. Natural killer cell mediated immunosurveillance of pediatric neuroblastoma. Oncoimmunology 2015, 4, e1042202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Angka, L.; Khan, S.T.; Kilgour, M.K.; Xu, R.; Kennedy, M.A.; Auer, R.C. Dysfunctional Natural Killer Cells in the Aftermath of Cancer Surgery. Int. J. Mol. Sci. 2017, 18, 1787. [Google Scholar] [CrossRef] [PubMed]
  36. Ali, T.H.; Pisanti, S.; Ciaglia, E.; Mortarini, R.; Anichini, A.; Garofalo, C.; Tallerico, R.; Santinami, M.; Gulletta, E.; Ietto, C.; 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. [Google Scholar] [CrossRef] [PubMed]
  37. Messaoudene, M.; Fregni, G.; Fourmentraux-Neves, E.; Chanal, J.; Maubec, E.; Mazouz-Dorval, S.; Couturaud, B.; Girod, A.; Sastre-Garau, X.; Albert, S.; et al. Mature cytotoxic CD56(bright)/CD16(+) natural killer cells can infiltrate lymph nodes adjacent to metastatic melanoma. Cancer Res. 2014, 74, 81–92. [Google Scholar] [CrossRef]
  38. Uchida, A. The cytolytic and regulatory role of natural killer cells in human neoplasia. Biochim. Biophys. Acta Rev. Cancer 1986, 865, 329–340. [Google Scholar] [CrossRef]
  39. Liu, X.; Ran, R.; Shao, B.; Rugo, H.S.; Yang, Y.; Hu, Z.; Wei, Z.; Wan, F.; Kong, W.; Song, G.; et al. Combined peripheral natural killer cell and circulating tumor cell enumeration enhance prognostic efficiency in patients with metastatic triple-negative breast cancer. Chin. J. Cancer Res. 2018, 30, 315–326. [Google Scholar] [CrossRef]
  40. Hoshikawa, M.; Aoki, T.; Matsushita, H.; Karasaki, T.; Hosoi, A.; Odaira, K.; Fujieda, N.; Kobayashi, Y.; Kambara, K.; Ohara, O.; et al. NK cell and IFN signatures are positive prognostic biomarkers for resectable pancreatic cancer. Biochem. Biophys. Res. Commun. 2018, 495, 2058–2065. [Google Scholar] [CrossRef]
  41. Schantz, S.P.; Campbell, B.H.; Guillamondegui, O.M. Pharyngeal carcinoma and natural killer cell activity. Am. J. Surg. 1986, 152, 467–474. [Google Scholar] [CrossRef]
  42. Lee, J.; Park, K.H.; Ryu, J.H.; Bae, H.J.; Choi, A.; Lee, H.; Lim, J.; Han, K.; Park, C.H.; Jung, E.S.; et al. Natural killer cell activity for IFN-gamma production as a supportive diagnostic marker for gastric cancer. Oncotarget 2017, 8, 70431–70440. [Google Scholar] [CrossRef] [PubMed]
  43. Ricciardi, M.; Zanotto, M.; Malpeli, G.; Bassi, G.; Perbellini, O.; Chilosi, M.; Bifari, F.; Krampera, M. Epithelial-to-mesenchymal transition (EMT) induced by inflammatory priming elicits mesenchymal stromal cell-like immune-modulatory properties in cancer cells. Br. J. Cancer 2015, 112, 1067–1075. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Knutson, K.L.; Lu, H.; Stone, B.; Reiman, J.M.; Behrens, M.D.; Prosperi, C.M.; Gad, E.A.; Smorlesi, A.; Disis, M.L. Immunoediting of cancers may lead to epithelial to mesenchymal transition. J. Immunol. 2006, 177, 1526–1533. [Google Scholar] [CrossRef] [PubMed]
  45. Lopez-Soto, A.; Huergo-Zapico, L.; Galvan, J.A.; Rodrigo, L.; de Herreros, A.G.; Astudillo, A.; Gonzalez, S. Epithelial-mesenchymal transition induces an antitumor immune response mediated by NKG2D receptor. J. Immunol. 2013, 190, 4408–4419. [Google Scholar] [CrossRef] [PubMed]
  46. Lopez-Soto, A.; Zapico, L.H.; Acebes-Huerta, A.; Rodrigo, L.; Gonzalez, S. Regulation of NKG2D signaling during the epithelial-to-mesenchymal transition. Oncoimmunology 2013, 2, e25820. [Google Scholar] [CrossRef] [PubMed]
  47. Chen, X.H.; Liu, Z.C.; Zhang, G.; Wei, W.; Wang, X.X.; Wang, H.; Ke, H.P.; Zhang, F.; Wang, H.S.; Cai, S.H.; et al. TGF-beta and EGF induced HLA-I downregulation is associated with epithelial-mesenchymal transition (EMT) through upregulation of snail in prostate cancer cells. Mol. Immunol. 2015, 65, 34–42. [Google Scholar] [CrossRef] [PubMed]
  48. Chockley, P.J.; Chen, J.; Chen, G.; Beer, D.G.; Standiford, T.J.; Keshamouni, V.G. Epithelial-mesenchymal transition leads to NK cell-mediated metastasis-specific immunosurveillance in lung cancer. J. Clin. Investig. 2018, 128, 1384–1396. [Google Scholar] [CrossRef] [PubMed]
  49. Kmieciak, M.; Knutson, K.L.; Dumur, C.I.; Manjili, M.H. HER-2/neu antigen loss and relapse of mammary carcinoma are actively induced by T cell-mediated anti-tumor immune responses. Eur. J. Immunol. 2007, 37, 675–685. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Huergo-Zapico, L.; Parodi, M.; Cantoni, C.; Lavarello, C.; Fernandez-Martinez, J.L.; Petretto, A.; DeAndres-Galiana, E.J.; Balsamo, M.; Lopez-Soto, A.; Pietra, G.; 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. [Google Scholar] [CrossRef] [PubMed]
  51. Bellucci, R.; Martin, A.; Bommarito, D.; Wang, K.; Hansen, S.H.; Freeman, G.J.; Ritz, J. Interferon-gamma-induced activation of JAK1 and JAK2 suppresses tumor cell susceptibility to NK cells through upregulation of PD-L1 expression. Oncoimmunology 2015, 4, e1008824. [Google Scholar] [CrossRef] [PubMed]
  52. Raimondi, C.; Carpino, G.; Nicolazzo, C.; Gradilone, A.; Gianni, W.; Gelibter, A.; Gaudio, E.; Cortesi, E.; Gazzaniga, P. PD-L1 and epithelial-mesenchymal transition in circulating tumor cells from non-small cell lung cancer patients: A molecular shield to evade immune system? Oncoimmunology 2017, 6, e1315488. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Chen, L.; Gibbons, D.L.; Goswami, S.; Cortez, M.A.; Ahn, Y.H.; Byers, L.A.; Zhang, X.; Yi, X.; Dwyer, D.; Lin, W.; et al. Metastasis is regulated via microRNA-200/ZEB1 axis control of tumour cell PD-L1 expression and intratumoral immunosuppression. Nat. Commun. 2014, 5, 5241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Noman, M.Z.; Janji, B.; Abdou, A.; Hasmim, M.; Terry, S.; Tan, T.Z.; Mami-Chouaib, F.; Thiery, J.P.; Chouaib, S. The immune checkpoint ligand PD-L1 is upregulated in EMT-activated human breast cancer cells by a mechanism involving ZEB-1 and miR-200. Oncoimmunology 2017, 6, e1263412. [Google Scholar] [CrossRef] [PubMed]
  55. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: the next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed]
  56. Lopez-Soto, A.; Folgueras, A.R.; Seto, E.; Gonzalez, S. HDAC3 represses the expression of NKG2D ligands ULBPs in epithelial tumour cells: Potential implications for the immunosurveillance of cancer. Oncogene 2009, 28, 2370–2382. [Google Scholar] [CrossRef] [PubMed]
  57. Yadav, D.; Ngolab, J.; Lim, R.S.; Krishnamurthy, S.; Bui, J.D. Cutting edge: down-regulation of MHC class I-related chain A on tumor cells by IFN-gamma-induced microRNA. J. Immunol. 2009, 182, 39–43. [Google Scholar] [CrossRef] [PubMed]
  58. Heinemann, A.; Zhao, F.; Pechlivanis, S.; Eberle, J.; Steinle, A.; Diederichs, S.; Schadendorf, D.; Paschen, A. Tumor suppressive microRNAs miR-34a/c control cancer cell expression of ULBP2, a stress-induced ligand of the natural killer cell receptor NKG2D. Cancer Res. 2012, 72, 460–471. [Google Scholar] [CrossRef]
  59. Stern-Ginossar, N.; Gur, C.; Biton, M.; Horwitz, E.; Elboim, M.; Stanietsky, N.; Mandelboim, M.; Mandelboim, O. Human microRNAs regulate stress-induced immune responses mediated by the receptor NKG2D. Nat. Immunol. 2008, 9, 1065–1073. [Google Scholar] [CrossRef]
  60. Kaiser, B.K.; Yim, D.; Chow, I.T.; Gonzalez, S.; Dai, Z.; Mann, H.H.; Strong, R.K.; Groh, V.; Spies, T. Disulphide-isomerase-enabled shedding of tumour-associated NKG2D ligands. Nature 2007, 447, 482–486. [Google Scholar] [CrossRef]
  61. Chitadze, G.; Lettau, M.; Bhat, J.; Wesch, D.; Steinle, A.; Furst, D.; Mytilineos, J.; Kalthoff, H.; Janssen, O.; Oberg, H.H.; et al. Shedding of endogenous MHC class I-related chain molecules A and B from different human tumor entities: heterogeneous involvement of the “a disintegrin and metalloproteases” 10 and 17. Int. J. Cancer 2013, 133, 1557–1566. [Google Scholar] [CrossRef] [PubMed]
  62. Liu, G.; Atteridge, C.L.; Wang, X.; Lundgren, A.D.; Wu, J.D. The membrane type matrix metalloproteinase MMP14 mediates constitutive shedding of MHC class I chain-related molecule A independent of A disintegrin and metalloproteinases. J. Immunol. 2010, 184, 3346–3350. [Google Scholar] [CrossRef] [PubMed]
  63. Gumireddy, K.; Sun, F.; Klein-Szanto, A.J.; Gibbins, J.M.; Gimotty, P.A.; Saunders, A.J.; Schultz, P.G.; Huang, Q. In vivo selection for metastasis promoting genes in the mouse. Proc. Natl. Acad. Sci. USA 2007, 104, 6696–6701. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Huergo-Zapico, L.; Gonzalez-Rodriguez, A.P.; Contesti, J.; Gonzalez, E.; Lopez-Soto, A.; Fernandez-Guizan, A.; Acebes-Huerta, A.; de Los Toyos, J.R.; Lopez-Larrea, C.; Groh, V.; et al. Expression of ERp5 and GRP78 on the membrane of chronic lymphocytic leukemia cells: association with soluble MICA shedding. Cancer Immunol. 2012, 61, 1201–1210. [Google Scholar] [CrossRef] [PubMed]
  65. Jinushi, M.; Vanneman, M.; Munshi, N.C.; Tai, Y.T.; Prabhala, R.H.; Ritz, J.; Neuberg, D.; Anderson, K.C.; Carrasco, D.R.; Dranoff, G. MHC class I chain-related protein A antibodies and shedding are associated with the progression of multiple myeloma. Proc. Natl. Acad. Sci. USA 2008, 105, 1285–1290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Zocchi, M.R.; Catellani, S.; Canevali, P.; Tavella, S.; Garuti, A.; Villaggio, B.; Zunino, A.; Gobbi, M.; Fraternali-Orcioni, G.; Kunkl, A.; et al. High ERp5/ADAM10 expression in lymph node microenvironment and impaired NKG2D ligands recognition in Hodgkin lymphomas. Blood 2012, 119, 1479–1489. [Google Scholar] [CrossRef] [PubMed]
  67. Holdenrieder, S.; Stieber, P.; Peterfi, A.; Nagel, D.; Steinle, A.; Salih, H.R. Soluble MICA in malignant diseases. Int. J. Cancer 2006, 118, 684–687. [Google Scholar] [CrossRef]
  68. Wang, L.P.; Niu, H.; Xia, Y.F.; Han, Y.L.; Niu, P.; Wang, H.Y.; Zhou, Q.L. Prognostic significance of serum sMICA levels in non-small cell lung cancer. Eur. Rev. Med. Pharm. Sci. 2015, 19, 2226–2230. [Google Scholar]
  69. Zhao, Y.K.; Jia, C.M.; Yuan, G.J.; Liu, W.; Qiu, Y.; Zhu, Q.G. Expression and clinical value of the soluble major histocompatibility complex class I-related chain A molecule in the serum of patients with renal tumors. Genet. Mol. Res. 2015, 14, 7233–7240. [Google Scholar] [CrossRef]
  70. Wang, J.; Li, C.; Yang, D.; Jian, X.C.; Jiang, C.H. Clinico-pathological significance of MHC-I type chain-associated protein A expression in oral squamous cell carcinoma. Asian Pac. J. Cancer Prev. 2012, 13, 715–718. [Google Scholar] [CrossRef]
  71. Fang, L.; Gong, J.; Wang, Y.; Liu, R.; Li, Z.; Wang, Z.; Zhang, Y.; Zhang, C.; Song, C.; Yang, A.; et al. MICA/B expression is inhibited by unfolded protein response and associated with poor prognosis in human hepatocellular carcinoma. J. Exp. Clin. Cancer Res. 2014, 33, 76. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Zhang, J.; Xu, Z.; Zhou, X.; Zhang, H.; Yang, N.; Wu, Y.; Chen, Y.; Yang, G.; Ren, T. Loss of expression of MHC class I-related chain A (MICA) is a frequent event and predicts poor survival in patients with hepatocellular carcinoma. Int. J. Clin. Exp. Pathol. 2014, 7, 3123–3131. [Google Scholar] [PubMed]
  73. Holdenrieder, S.; Stieber, P.; Peterfi, A.; Nagel, D.; Steinle, A.; Salih, H.R. Soluble MICB in malignant diseases: Analysis of diagnostic significance and correlation with soluble MICA. Cancer Immunol. Immunother. 2006, 55, 1584–1589. [Google Scholar] [CrossRef] [PubMed]
  74. Tamaki, S.; Sanefuzi, N.; Kawakami, M.; Aoki, K.; Imai, Y.; Yamanaka, Y.; Yamamoto, K.; Ishitani, A.; Hatake, K.; Kirita, T. Association between soluble MICA levels and disease stage IV oral squamous cell carcinoma in Japanese patients. Hum. Immunol. 2008, 69, 88–93. [Google Scholar] [CrossRef] [PubMed]
  75. Paschen, A.; Sucker, A.; Hill, B.; Moll, I.; Zapatka, M.; Nguyen, X.D.; Sim, G.C.; Gutmann, I.; Hassel, J.; Becker, J.C.; et al. Differential clinical significance of individual NKG2D ligands in melanoma: Soluble ULBP2 as an indicator of poor prognosis superior to S100B. Clin. Cancer Res. 2009, 15, 5208–5215. [Google Scholar] [CrossRef] [PubMed]
  76. Xu, Y.; Zhou, L.; Zong, J.; Ye, Y.; Chen, G.; Chen, Y.; Liao, X.; Guo, Q.; Qiu, S.; Lin, S.; et al. Decreased expression of the NKG2D ligand ULBP4 may be an indicator of poor prognosis in patients with nasopharyngeal carcinoma. Oncotarget 2017, 8, 42007–42019. [Google Scholar] [CrossRef] [PubMed]
  77. Zhou, Y.; Xu, Y.; Chen, L.; Xu, B.; Wu, C.; Jiang, J. B7-H6 expression correlates with cancer progression and patient’s survival in human ovarian cancer. Int. J. Clin. Exp. Pathol. 2015, 8, 9428–9433. [Google Scholar] [PubMed]
  78. Benson, D.M., Jr.; Bakan, C.E.; Mishra, A.; Hofmeister, C.C.; Efebera, Y.; Becknell, B.; Baiocchi, R.A.; Zhang, J.; Yu, J.; Smith, M.K.; et al. The PD-1/PD-L1 axis modulates the natural killer cell versus multiple myeloma effect: a therapeutic target for CT-011, a novel monoclonal anti-PD-1 antibody. Blood 2010, 116, 2286–2294. [Google Scholar] [CrossRef] [Green Version]
  79. Liu, Y.; Cheng, Y.; Xu, Y.; Wang, Z.; Du, X.; Li, C.; Peng, J.; Gao, L.; Liang, X.; Ma, C. Increased expression of programmed cell death protein 1 on NK cells inhibits NK-cell-mediated anti-tumor function and indicates poor prognosis in digestive cancers. Oncogene 2017, 36, 6143–6153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Barry, K.C.; Hsu, J.; Broz, M.L.; Cueto, F.J.; Binnewies, M.; Combes, A.J.; Nelson, A.E.; Loo, K.; Kumar, R.; Rosenblum, M.D.; et al. A natural killer-dendritic cell axis defines checkpoint therapy-responsive tumor microenvironments. Nat. Med. 2018, 24, 1178–1191. [Google Scholar] [CrossRef]
  81. Yoon, S.J.; Heo, D.S.; Kang, S.H.; Lee, K.H.; Kim, W.S.; Kim, G.P.; Lee, J.A.; Lee, K.S.; Bang, Y.J.; Kim, N.K. Natural killer cell activity depression in peripheral blood and ascites from gastric cancer patients with high TGF-beta 1 expression. Anticancer Res. 1998, 18, 1591–1596. [Google Scholar] [PubMed]
  82. Hata, A.; Shi, Y.; Massague, J. TGF-beta signaling and cancer: structural and functional consequences of mutations in Smads. Mol. Med. Today 1998, 4, 257–262. [Google Scholar] [CrossRef]
  83. Espinoza, J.L.; Takami, A.; Yoshioka, K.; Nakata, K.; Sato, T.; Kasahara, Y.; Nakao, S. Human microRNA-1245 down-regulates the NKG2D receptor in natural killer cells and impairs NKG2D-mediated functions. Haematologica 2012, 97, 1295–1303. [Google Scholar] [CrossRef] [PubMed]
  84. Gao, Y.; Souza-Fonseca-Guimaraes, F.; Bald, T.; Ng, S.S.; Young, A.; Ngiow, S.F.; Rautela, J.; Straube, J.; Waddell, N.; Blake, S.J.; et al. Tumor immunoevasion by the conversion of effector NK cells into type 1 innate lymphoid cells. Nat. Immunol. 2017, 18, 1004–1015. [Google Scholar] [CrossRef] [PubMed]
  85. Smyth, M.J.; Teng, M.W.; Swann, J.; Kyparissoudis, K.; Godfrey, D.I.; Hayakawa, Y. CD4+CD25+ T regulatory cells suppress NK cell-mediated immunotherapy of cancer. J. Immunol. 2006, 176, 1582–1587. [Google Scholar] [CrossRef] [PubMed]
  86. Li, H.; Han, Y.; Guo, Q.; Zhang, M.; Cao, X. Cancer-expanded myeloid-derived suppressor cells induce anergy of NK cells through membrane-bound TGF-beta 1. J. Immunol. 2009, 182, 240–249. [Google Scholar] [CrossRef] [PubMed]
  87. Sceneay, J.; Chow, M.T.; Chen, A.; Halse, H.M.; Wong, C.S.; Andrews, D.M.; Sloan, E.K.; Parker, B.S.; Bowtell, D.D.; Smyth, M.J.; et al. Primary tumor hypoxia recruits CD11b+/Ly6Cmed/Ly6G+ immune suppressor cells and compromises NK cell cytotoxicity in the premetastatic niche. Cancer Res. 2012, 72, 3906–3911. [Google Scholar] [CrossRef]
  88. Della Chiesa, M.; Carlomagno, S.; Frumento, G.; Balsamo, M.; Cantoni, C.; Conte, R.; Moretta, L.; Moretta, A.; Vitale, M. The tryptophan catabolite L-kynurenine inhibits the surface expression of NKp46- and NKG2D-activating receptors and regulates NK-cell function. Blood 2006, 108, 4118–4125. [Google Scholar] [CrossRef]
  89. Palumbo, J.S.; Talmage, K.E.; Massari, J.V.; La Jeunesse, C.M.; Flick, M.J.; Kombrinck, K.W.; Hu, Z.; Barney, K.A.; Degen, J.L. Tumor cell-associated tissue factor and circulating hemostatic factors cooperate to increase metastatic potential through natural killer cell-dependent and-independent mechanisms. Blood 2007, 110, 133–141. [Google Scholar] [CrossRef] [Green Version]
  90. Palumbo, J.S.; Talmage, K.E.; Massari, J.V.; La Jeunesse, C.M.; Flick, M.J.; Kombrinck, K.W.; Jirouskova, M.; Degen, J.L. Platelets and fibrin(ogen) increase metastatic potential by impeding natural killer cell-mediated elimination of tumor cells. Blood 2005, 105, 178–185. [Google Scholar] [CrossRef] [Green Version]
  91. Kopp, H.G.; Placke, T.; Salih, H.R. Platelet-derived transforming growth factor-beta down-regulates NKG2D thereby inhibiting natural killer cell antitumor reactivity. Cancer Res. 2009, 69, 7775–7783. [Google Scholar] [CrossRef] [PubMed]
  92. Placke, T.; Orgel, M.; Schaller, M.; Jung, G.; Rammensee, H.G.; Kopp, H.G.; Salih, H.R. Platelet-derived MHC class I confers a pseudonormal phenotype to cancer cells that subverts the antitumor reactivity of natural killer immune cells. Cancer Res. 2012, 72, 440–448. [Google Scholar] [CrossRef] [PubMed]
  93. Maurer, S.; Kropp, K.N.; Klein, G.; Steinle, A.; Haen, S.P.; Walz, J.S.; Hinterleitner, C.; Marklin, M.; Kopp, H.G.; Salih, H.R. Platelet-mediated shedding of NKG2D ligands impairs NK cell immune-surveillance of tumor cells. Oncoimmunology 2018, 7, e1364827. [Google Scholar] [CrossRef] [PubMed]
  94. Placke, T.; Salih, H.R.; Kopp, H.G. GITR ligand provided by thrombopoietic cells inhibits NK cell antitumor activity. J. Immunol. 2012, 189, 154–160. [Google Scholar] [CrossRef] [PubMed]
  95. Vuletic, A.; Jovanic, I.; Jurisic, V.; Milovanovic, Z.; Nikolic, S.; Spurnic, I.; Konjevic, G. Decreased Interferon gamma Production in CD3+ and CD3− CD56+ Lymphocyte Subsets in Metastatic Regional Lymph Nodes of Melanoma Patients. Pathol. Oncol. Res. 2015, 21, 1109–1114. [Google Scholar] [CrossRef] [PubMed]
  96. Schuster, I.S.; Coudert, J.D.; Andoniou, C.E.; Degli-Esposti, M.A. “Natural Regulators”: NK Cells as Modulators of T Cell Immunity. Front. Immunol. 2016, 7, 235. [Google Scholar] [CrossRef] [PubMed]
  97. Porrata, L.F.; Inwards, D.J.; Ansell, S.M.; Micallef, I.N.; Johnston, P.B.; Gastineau, D.A.; Litzow, M.R.; Winters, J.L.; Markovic, S.N. Early lymphocyte recovery predicts superior survival after autologous stem cell transplantation in non-Hodgkin lymphoma: A prospective study. Biol. Blood Marrow Transplant. 2008, 14, 807–816. [Google Scholar] [CrossRef] [PubMed]
  98. Rueff, J.; Medinger, M.; Heim, D.; Passweg, J.; Stern, M. Lymphocyte subset recovery and outcome after autologous hematopoietic stem cell transplantation for plasma cell myeloma. Biol. Blood Marrow Transplant. 2014, 20, 896–899. [Google Scholar] [CrossRef]
  99. Parkhurst, M.R.; Riley, J.P.; Dudley, M.E.; Rosenberg, S.A. Adoptive transfer of autologous natural killer cells leads to high levels of circulating natural killer cells but does not mediate tumor regression. Clin. Cancer Res. 2011, 17, 6287–6297. [Google Scholar] [CrossRef]
  100. Sakamoto, N.; Ishikawa, T.; Kokura, S.; Okayama, T.; Oka, K.; Ideno, M.; Sakai, F.; Kato, A.; Tanabe, M.; Enoki, T.; et al. Phase I clinical trial of autologous NK cell therapy using novel expansion method in patients with advanced digestive cancer. J. Transl. Med. 2015, 13, 277. [Google Scholar] [CrossRef] [Green Version]
  101. Rosenberg, S.A.; Lotze, M.T.; Muul, L.M.; Leitman, S.; Chang, A.E.; Ettinghausen, S.E.; Matory, Y.L.; Skibber, J.M.; Shiloni, E.; Vetto, J.T.; et al. Observations on the systemic administration of autologous lymphokine-activated killer cells and recombinant interleukin-2 to patients with metastatic cancer. N. Engl. J. Med. 1985, 313, 1485–1492. [Google Scholar] [CrossRef] [PubMed]
  102. Ruggeri, L.; Mancusi, A.; Burchielli, E.; Capanni, M.; Carotti, A.; Aloisi, T.; Aversa, F.; Martelli, M.F.; Velardi, A. NK cell alloreactivity and allogeneic hematopoietic stem cell transplantation. Blood Cells Mol. Dis. 2008, 40, 84–90. [Google Scholar] [CrossRef] [PubMed]
  103. Qin, Z.; Chen, J.; Zeng, J.; Niu, L.; Xie, S.; Wang, X.; Liang, Y.; Wu, Z.; Zhang, M. Effect of NK cell immunotherapy on immune function in patients with hepatic carcinoma: A preliminary clinical study. Cancer Biol. 2017, 18, 323–330. [Google Scholar] [CrossRef] [PubMed]
  104. Alnaggar, M.; Lin, M.; Mesmar, A.; Liang, S.; Qaid, A.; Xu, K.; Chen, J.; Niu, L.; Yin, Z. Allogenic Natural Killer Cell Immunotherapy Combined with Irreversible Electroporation for Stage IV Hepatocellular Carcinoma: Survival Outcome. Cell Physiol. Biochem. 2018, 48, 1882–1893. [Google Scholar] [CrossRef] [PubMed]
  105. Lin, M.; Liang, S.; Wang, X.; Liang, Y.; Zhang, M.; Chen, J.; Niu, L.; Xu, K. Short-term clinical efficacy of percutaneous irreversible electroporation combined with allogeneic natural killer cell for treating metastatic pancreatic cancer. Immunol. Lett. 2017, 186, 20–27. [Google Scholar] [CrossRef] [PubMed]
  106. Adotevi, O.; Godet, Y.; Galaine, J.; Lakkis, Z.; Idirene, I.; Certoux, J.M.; Jary, M.; Loyon, R.; Laheurte, C.; Kim, S.; et al. In situ delivery of allogeneic natural killer cell (NK) combined with Cetuximab in liver metastases of gastrointestinal carcinoma: A phase I clinical trial. Oncoimmunology 2018, 7, e1424673. [Google Scholar] [CrossRef] [PubMed]
  107. Geller, M.A.; Cooley, S.; Judson, P.L.; Ghebre, R.; Carson, L.F.; Argenta, P.A.; Jonson, A.L.; Panoskaltsis-Mortari, A.; Curtsinger, J.; McKenna, D.; et al. A phase II study of allogeneic natural killer cell therapy to treat patients with recurrent ovarian and breast cancer. Cytotherapy 2011, 13, 98–107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Lin, M.; Liang, S.Z.; Shi, J.; Niu, L.Z.; Chen, J.B.; Zhang, M.J.; Xu, K.C. Circulating tumor cell as a biomarker for evaluating allogenic NK cell immunotherapy on stage IV non-small cell lung cancer. Immunol. Lett. 2017, 191, 10–15. [Google Scholar] [CrossRef]
  109. Fehniger, T.A.; Miller, J.S.; Stuart, R.K.; Cooley, S.; Salhotra, A.; Curtsinger, J.; Westervelt, P.; DiPersio, J.F.; Hillman, T.M.; Silver, N.; et al. A Phase 1 Trial of CNDO-109-Activated Natural Killer Cells in Patients with High-Risk Acute Myeloid Leukemia. Biol. Blood Marrow Transplant. 2018, 24, 1581–1589. [Google Scholar] [CrossRef]
  110. Romee, R.; Rosario, M.; Berrien-Elliott, M.M.; Wagner, J.A.; Jewell, B.A.; Schappe, T.; Leong, J.W.; Abdel-Latif, S.; Schneider, S.E.; Willey, S.; et al. Cytokine-induced memory-like natural killer cells exhibit enhanced responses against myeloid leukemia. Sci. Transl. Med. 2016, 8, 357. [Google Scholar] [CrossRef]
  111. Gill, S.; June, C.H. Going viral: Chimeric antigen receptor T-cell therapy for hematological malignancies. Immunol. Rev. 2015, 263, 68–89. [Google Scholar] [CrossRef]
  112. Kruschinski, A.; Moosmann, A.; Poschke, I.; Norell, H.; Chmielewski, M.; Seliger, B.; Kiessling, R.; Blankenstein, T.; Abken, H.; Charo, J. Engineering antigen-specific primary human NK cells against HER-2 positive carcinomas. Proc. Natl. Acad. Sci. USA 2008, 105, 17481–17486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Schonfeld, K.; Sahm, C.; Zhang, C.; Naundorf, S.; Brendel, C.; Odendahl, M.; Nowakowska, P.; Bonig, H.; Kohl, U.; Kloess, S.; et al. Selective inhibition of tumor growth by clonal NK cells expressing an ErbB2/HER2-specific chimeric antigen receptor. Mol. Ther. 2015, 23, 330–338. [Google Scholar] [CrossRef] [PubMed]
  114. Klingemann, H. Are natural killer cells superior CAR drivers? Oncoimmunology 2014, 3, e28147. [Google Scholar] [CrossRef] [PubMed]
  115. Tang, X.; Yang, L.; Li, Z.; Nalin, A.P.; Dai, H.; Xu, T.; Yin, J.; You, F.; Zhu, M.; Shen, W.; et al. First-in-man clinical trial of CAR NK-92 cells: Safety test of CD33-CAR NK-92 cells in patients with relapsed and refractory acute myeloid leukemia. Am. J. Cancer Res. 2018, 8, 1083–1089. [Google Scholar] [PubMed]
  116. Boissel, L.; Betancur, M.; Wels, W.S.; Tuncer, H.; Klingemann, H. Transfection with mRNA for CD19 specific chimeric antigen receptor restores NK cell mediated killing of CLL cells. Leuk. Res. 2009, 33, 1255–1259. [Google Scholar] [CrossRef] [Green Version]
  117. Uherek, C.; Tonn, T.; Uherek, B.; Becker, S.; Schnierle, B.; Klingemann, H.G.; Wels, W. Retargeting of natural killer-cell cytolytic activity to ErbB2-expressing cancer cells results in efficient and selective tumor cell destruction. Blood 2002, 100, 1265–1273. [Google Scholar]
  118. Hermanson, D.L.; Kaufman, D.S. Utilizing chimeric antigen receptors to direct natural killer cell activity. Front. Immunol. 2015, 6, 195. [Google Scholar] [CrossRef]
  119. Tonn, T.; Schwabe, D.; Klingemann, H.G.; Becker, S.; Esser, R.; Koehl, U.; Suttorp, M.; Seifried, E.; Ottmann, O.G.; Bug, G. Treatment of patients with advanced cancer with the natural killer cell line NK-92. Cytotherapy 2013, 15, 1563–1570. [Google Scholar] [CrossRef]
  120. Genssler, S.; Burger, M.C.; Zhang, C.; Oelsner, S.; Mildenberger, I.; Wagner, M.; Steinbach, J.P.; Wels, W.S. Dual targeting of glioblastoma with chimeric antigen receptor-engineered natural killer cells overcomes heterogeneity of target antigen expression and enhances antitumor activity and survival. Oncoimmunology 2016, 5, e1119354. [Google Scholar] [CrossRef]
  121. Han, J.; Chu, J.; Keung Chan, W.; Zhang, J.; Wang, Y.; Cohen, J.B.; Victor, A.; Meisen, W.H.; Kim, S.H.; Grandi, P.; et al. CAR-Engineered NK Cells Targeting Wild-Type EGFR and EGFRvIII Enhance Killing of Glioblastoma and Patient-Derived Glioblastoma Stem Cells. Sci. Rep. 2015, 5, 11483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Zhang, C.; Burger, M.C.; Jennewein, L.; Genssler, S.; Schonfeld, K.; Zeiner, P.; Hattingen, E.; Harter, P.N.; Mittelbronn, M.; Tonn, T.; et al. ErbB2/HER2-Specific NK Cells for Targeted Therapy of Glioblastoma. J. Natl. Cancer Inst. 2016, 108. [Google Scholar] [CrossRef] [PubMed]
  123. Chen, X.; Han, J.; Chu, J.; Zhang, L.; Zhang, J.; Chen, C.; Chen, L.; Wang, Y.; Wang, H.; Yi, L.; et al. A combinational therapy of EGFR-CAR NK cells and oncolytic herpes simplex virus 1 for breast cancer brain metastases. Oncotarget 2016, 7, 27764–27777. [Google Scholar] [CrossRef] [Green Version]
  124. Jiang, T.; Zhou, C.; Ren, S. Role of IL-2 in cancer immunotherapy. Oncoimmunology 2016, 5, e1163462. [Google Scholar] [CrossRef] [Green Version]
  125. Rosenberg, S.A. IL-2: The first effective immunotherapy for human cancer. J. Immunol. 2014, 192, 5451–5458. [Google Scholar] [CrossRef] [PubMed]
  126. Ahmadzadeh, M.; Rosenberg, S.A. IL-2 administration increases CD4+ CD25(hi) Foxp3+ regulatory T cells in cancer patients. Blood 2006, 107, 2409–2414. [Google Scholar] [CrossRef] [PubMed]
  127. Berntsen, A.; Brimnes, M.K.; Thor Straten, P.; Svane, I.M. Increase of circulating CD4+CD25highFoxp3+ regulatory T cells in patients with metastatic renal cell carcinoma during treatment with dendritic cell vaccination and low-dose interleukin-2. J. Immunother. 2010, 33, 425–434. [Google Scholar] [CrossRef]
  128. Sim, G.C.; Martin-Orozco, N.; Jin, L.; Yang, Y.; Wu, S.; Washington, E.; Sanders, D.; Lacey, C.; Wang, Y.; Vence, L.; et al. IL-2 therapy promotes suppressive ICOS+ Treg expansion in melanoma patients. J. Clin. Investig. 2014, 124, 99–110. [Google Scholar] [CrossRef]
  129. Rosenberg, S.A.; Lotze, M.T.; Yang, J.C.; Aebersold, P.M.; Linehan, W.M.; Seipp, C.A.; White, D.E. Experience with the use of high-dose interleukin-2 in the treatment of 652 cancer patients. Ann. Surg. 1989, 210, 474–484. [Google Scholar] [CrossRef]
  130. Smith, F.O.; Downey, S.G.; Klapper, J.A.; Yang, J.C.; Sherry, R.M.; Royal, R.E.; Kammula, U.S.; Hughes, M.S.; Restifo, N.P.; Levy, C.L.; et al. Treatment of metastatic melanoma using interleukin-2 alone or in conjunction with vaccines. Clin. Cancer Res. 2008, 14, 5610–5618. [Google Scholar] [CrossRef]
  131. Dudley, M.E.; Wunderlich, J.R.; Yang, J.C.; Sherry, R.M.; Topalian, S.L.; Restifo, N.P.; Royal, R.E.; Kammula, U.; White, D.E.; Mavroukakis, S.A.; et al. Adoptive cell transfer therapy following non-myeloablative but lymphodepleting chemotherapy for the treatment of patients with refractory metastatic melanoma. J. Clin. Oncol. 2005, 23, 2346–2357. [Google Scholar] [CrossRef] [PubMed]
  132. Dudley, M.E.; Yang, J.C.; Sherry, R.; Hughes, M.S.; Royal, R.; Kammula, U.; Robbins, P.F.; Huang, J.; Citrin, D.E.; Leitman, S.F.; et al. Adoptive cell therapy for patients with metastatic melanoma: Evaluation of intensive myeloablative chemoradiation preparative regimens. J. Clin. Oncol. 2008, 26, 5233–5239. [Google Scholar] [CrossRef] [PubMed]
  133. Romee, R.; Leong, J.W.; Fehniger, T.A. Utilizing cytokines to function-enable human NK cells for the immunotherapy of cancer. Scientifica 2014, 2014, 205796. [Google Scholar] [CrossRef] [PubMed]
  134. Robinson, T.O.; Schluns, K.S. The potential and promise of IL-15 in immuno-oncogenic therapies. Immunol. Lett. 2017, 190, 159–168. [Google Scholar] [CrossRef] [PubMed]
  135. Marks-Konczalik, J.; Dubois, S.; Losi, J.M.; Sabzevari, H.; Yamada, N.; Feigenbaum, L.; Waldmann, T.A.; Tagaya, Y. IL-2-induced activation-induced cell death is inhibited in IL-15 transgenic mice. Proc. Natl. Acad. Sci. USA 2000, 97, 11445–11450. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Waldmann, T.A.; Lugli, E.; Roederer, M.; Perera, L.P.; Smedley, J.V.; Macallister, R.P.; Goldman, C.K.; Bryant, B.R.; Decker, J.M.; Fleisher, T.A.; et al. Safety (toxicity), pharmacokinetics, immunogenicity, and impact on elements of the normal immune system of recombinant human IL-15 in rhesus macaques. Blood 2011, 117, 4787–4795. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Tang, F.; Zhao, L.T.; Jiang, Y.; Ba de, N.; Cui, L.X.; He, W. Activity of recombinant human interleukin-15 against tumor recurrence and metastasis in mice. Cell Mol. Immunol. 2008, 5, 189–196. [Google Scholar] [CrossRef]
  138. Klebanoff, C.A.; Finkelstein, S.E.; Surman, D.R.; Lichtman, M.K.; Gattinoni, L.; Theoret, M.R.; Grewal, N.; Spiess, P.J.; Antony, P.A.; Palmer, D.C.; et al. IL-15 enhances the in vivo antitumor activity of tumor-reactive CD8+ T cells. Proc. Natl. Acad. Sci. USA 2004, 101, 1969–1974. [Google Scholar] [CrossRef]
  139. Yu, P.; Steel, J.C.; Zhang, M.; Morris, J.C.; Waldmann, T.A. Simultaneous blockade of multiple immune system inhibitory checkpoints enhances antitumor activity mediated by interleukin-15 in a murine metastatic colon carcinoma model. Clin. Cancer Res. 2010, 16, 6019–6028. [Google Scholar] [CrossRef]
  140. Kobayashi, H.; Dubois, S.; Sato, N.; Sabzevari, H.; Sakai, Y.; Waldmann, T.A.; Tagaya, Y. Role of trans-cellular IL-15 presentation in the activation of NK cell-mediated killing, which leads to enhanced tumor immunosurveillance. Blood 2005, 105, 721–727. [Google Scholar] [CrossRef] [Green Version]
  141. Conlon, K.C.; Lugli, E.; Welles, H.C.; Rosenberg, S.A.; Fojo, A.T.; Morris, J.C.; Fleisher, T.A.; Dubois, S.P.; Perera, L.P.; Stewart, D.M.; et al. Redistribution, hyperproliferation, activation of natural killer cells and CD8 T cells, and cytokine production during first-in-human clinical trial of recombinant human interleukin-15 in patients with cancer. J. Clin. Oncol. 2015, 33, 74–82. [Google Scholar] [CrossRef] [PubMed]
  142. Perez-Martinez, A.; Fernandez, L.; Valentin, J.; Martinez-Romera, I.; Corral, M.D.; Ramirez, M.; Abad, L.; Santamaria, S.; Gonzalez-Vicent, M.; Sirvent, S.; et al. A phase I/II trial of interleukin-15—Stimulated natural killer cell infusion after haplo-identical stem cell transplantation for pediatric refractory solid tumors. Cytotherapy 2015, 17, 1594–1603. [Google Scholar] [CrossRef] [PubMed]
  143. Ni, J.; Miller, M.; Stojanovic, A.; Garbi, N.; Cerwenka, A. Sustained effector function of IL-12/15/18-preactivated NK cells against established tumors. J. Exp. Med. 2012, 209, 2351–2365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Parrish-Novak, J.; Dillon, S.R.; Nelson, A.; Hammond, A.; Sprecher, C.; Gross, J.A.; Johnston, J.; Madden, K.; Xu, W.; West, J.; et al. Interleukin 21 and its receptor are involved in NK cell expansion and regulation of lymphocyte function. Nature 2000, 408, 57–63. [Google Scholar] [CrossRef] [PubMed]
  145. Moroz, A.; Eppolito, C.; Li, Q.; Tao, J.; Clegg, C.H.; Shrikant, P.A. IL-21 enhances and sustains CD8+ T cell responses to achieve durable tumor immunity: comparative evaluation of IL-2, IL-15, and IL-21. J. Immunol. 2004, 173, 900–909. [Google Scholar] [CrossRef]
  146. Di Carlo, E.; Comes, A.; Orengo, A.M.; Rosso, O.; Meazza, R.; Musiani, P.; Colombo, M.P.; Ferrini, S. IL-21 induces tumor rejection by specific CTL and IFN-gamma-dependent CXC chemokines in syngeneic mice. J. Immunol. 2004, 172, 1540–1547. [Google Scholar] [CrossRef] [PubMed]
  147. Thompson, J.A.; Curti, B.D.; Redman, B.G.; Bhatia, S.; Weber, J.S.; Agarwala, S.S.; Sievers, E.L.; Hughes, S.D.; DeVries, T.A.; Hausman, D.F. Phase I study of recombinant interleukin-21 in patients with metastatic melanoma and renal cell carcinoma. J. Clin. Oncol. 2008, 26, 2034–2039. [Google Scholar] [CrossRef] [PubMed]
  148. Wang, G.; Tschoi, M.; Spolski, R.; Lou, Y.; Ozaki, K.; Feng, C.; Kim, G.; Leonard, W.J.; Hwu, P. In vivo antitumor activity of interleukin 21 mediated by natural killer cells. Cancer Res. 2003, 63, 9016–9022. [Google Scholar] [PubMed]
  149. Ugai, S.; Shimozato, O.; Kawamura, K.; Wang, Y.Q.; Yamaguchi, T.; Saisho, H.; Sakiyama, S.; Tagawa, M. Expression of the interleukin-21 gene in murine colon carcinoma cells generates systemic immunity in the inoculated hosts. Cancer Gene 2003, 10, 187–192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Petrella, T.M.; Tozer, R.; Belanger, K.; Savage, K.J.; Wong, R.; Smylie, M.; Kamel-Reid, S.; Tron, V.; Chen, B.E.; Hunder, N.N.; et al. Interleukin-21 has activity in patients with metastatic melanoma: A phase II study. J. Clin. Oncol. 2012, 30, 3396–3401. [Google Scholar] [CrossRef]
  151. Stolfi, C.; Pallone, F.; Macdonald, T.T.; Monteleone, G. Interleukin-21 in cancer immunotherapy: Friend or foe? Oncoimmunology 2012, 1, 351–354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Clynes, R.A.; Towers, T.L.; Presta, L.G.; Ravetch, J.V. Inhibitory Fc receptors modulate in vivo cytotoxicity against tumor targets. Nat. Med. 2000, 6, 443–446. [Google Scholar] [CrossRef] [PubMed]
  153. Weng, W.K.; Levy, R. Two immunoglobulin G fragment C receptor polymorphisms independently predict response to rituximab in patients with follicular lymphoma. J. Clin. Oncol. 2003, 21, 3940–3947. [Google Scholar] [CrossRef] [PubMed]
  154. Rodriguez, J.; Zarate, R.; Bandres, E.; Boni, V.; Hernandez, A.; Sola, J.J.; Honorato, B.; Bitarte, N.; Garcia-Foncillas, J. Fc gamma receptor polymorphisms as predictive markers of Cetuximab efficacy in epidermal growth factor receptor downstream-mutated metastatic colorectal cancer. Eur. J. Cancer 2012, 48, 1774–1780. [Google Scholar] [CrossRef] [PubMed]
  155. Romond, E.H.; Perez, E.A.; Bryant, J.; Suman, V.J.; Geyer, C.E., Jr.; Davidson, N.E.; Tan-Chiu, E.; Martino, S.; Paik, S.; Kaufman, P.A.; et al. Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer. N. Engl. J. Med. 2005, 353, 1673–1684. [Google Scholar] [CrossRef] [PubMed]
  156. Vogel, C.L.; Cobleigh, M.A.; Tripathy, D.; Gutheil, J.C.; Harris, L.N.; Fehrenbacher, L.; Slamon, D.J.; Murphy, M.; Novotny, W.F.; Burchmore, M.; et al. Efficacy and safety of trastuzumab as a single agent in first-line treatment of HER2-overexpressing metastatic breast cancer. J. Clin. Oncol. 2002, 20, 719–726. [Google Scholar] [CrossRef] [PubMed]
  157. Seimetz, D.; Lindhofer, H.; Bokemeyer, C. Development and approval of the trifunctional antibody catumaxomab (anti-EpCAM x anti-CD3) as a targeted cancer immunotherapy. Cancer Treat. Rev. 2010, 36, 458–467. [Google Scholar] [CrossRef]
  158. Ferrari de Andrade, L.; Tay, R.E.; Pan, D.; Luoma, A.M.; Ito, Y.; Badrinath, S.; Tsoucas, D.; Franz, B.; May, K.F., Jr.; Harvey, C.J.; et al. Antibody-mediated inhibition of MICA and MICB shedding promotes NK cell-driven tumor immunity. Science 2018, 359, 1537–1542. [Google Scholar] [CrossRef]
  159. Muntasell, A.; Ochoa, M.C.; Cordeiro, L.; Berraondo, P.; Lopez-Diaz de Cerio, A.; Cabo, M.; Lopez-Botet, M.; Melero, I. Targeting NK-cell checkpoints for cancer immunotherapy. Curr. Opin. Immunol. 2017, 45, 73–81. [Google Scholar] [CrossRef]
  160. Lesokhin, A.M.; Callahan, M.K.; Postow, M.A.; Wolchok, J.D. On being less tolerant: enhanced cancer immunosurveillance enabled by targeting checkpoints and agonists of T cell activation. Sci. Transl. Med. 2015, 7, 280sr1. [Google Scholar] [CrossRef]
  161. Larkin, J.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.J.; Cowey, C.L.; Lao, C.D.; Schadendorf, D.; Dummer, R.; Smylie, M.; Rutkowski, P.; et al. Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. N. Engl. J. Med. 2015, 373, 23–34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Hsu, J.; Hodgins, J.J.; Marathe, M.; Nicolai, C.J.; Bourgeois-Daigneault, M.C.; Trevino, T.N.; Azimi, C.S.; Scheer, A.K.; Randolph, H.E.; Thompson, T.W.; et al. Contribution of NK cells to immunotherapy mediated by PD-1/PD-L1 blockade. J. Clin. Investig. 2018, 128, 4654–4668. [Google Scholar] [CrossRef] [PubMed]
  163. Bottcher, J.P.; Bonavita, E.; Chakravarty, P.; Blees, H.; Cabeza-Cabrerizo, M.; Sammicheli, S.; Rogers, N.C.; Sahai, E.; Zelenay, S.; Reis e Sousa, C. NK Cells Stimulate Recruitment of cDC1 into the Tumor Microenvironment Promoting Cancer Immune Control. Cell 2018, 172, 1022.e14–1037.e14. [Google Scholar] [CrossRef] [PubMed]
  164. Romagne, F.; Andre, P.; Spee, P.; Zahn, S.; Anfossi, N.; Gauthier, L.; Capanni, M.; Ruggeri, L.; Benson, D.M., Jr.; Blaser, B.W.; et al. Preclinical characterization of 1-7F9, a novel human anti-KIR receptor therapeutic antibody that augments natural killer-mediated killing of tumor cells. Blood 2009, 114, 2667–2677. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Carlsten, M.; Korde, N.; Kotecha, R.; Reger, R.; Bor, S.; Kazandjian, D.; Landgren, O.; Childs, R.W. Checkpoint Inhibition of KIR2D with the Monoclonal Antibody IPH2101 Induces Contraction and Hyporesponsiveness of NK Cells in Patients with Myeloma. Clin. Cancer Res. 2016, 22, 5211–5222. [Google Scholar] [CrossRef] [PubMed]
  166. McWilliams, E.M.; Mele, J.M.; Cheney, C.; Timmerman, E.A.; Fiazuddin, F.; Strattan, E.J.; Mo, X.; Byrd, J.C.; Muthusamy, N.; Awan, F.T. Therapeutic CD94/NKG2A blockade improves natural killer cell dysfunction in chronic lymphocytic leukemia. Oncoimmunology 2016, 5, e1226720. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Zhang, Q.; Bi, J.; Zheng, X.; Chen, Y.; Wang, H.; Wu, W.; Wang, Z.; Wu, Q.; Peng, H.; Wei, H.; et al. Blockade of the checkpoint receptor TIGIT prevents NK cell exhaustion and elicits potent anti-tumor immunity. Nat. Immunol. 2018. [Google Scholar] [CrossRef]
  168. Blake, S.J.; Stannard, K.; Liu, J.; Allen, S.; Yong, M.C.; Mittal, D.; Aguilera, A.R.; Miles, J.J.; Lutzky, V.P.; de Andrade, L.F.; et al. Suppression of Metastases Using a New Lymphocyte Checkpoint Target for Cancer Immunotherapy. Cancer Discov. 2016, 6, 446–459. [Google Scholar] [CrossRef] [Green Version]
  169. Xu, L.; Huang, Y.; Tan, L.; Yu, W.; Chen, D.; Lu, C.; He, J.; Wu, G.; Liu, X.; Zhang, Y. 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. [Google Scholar] [CrossRef]
  170. Wang, Z.; Zhu, J.; Gu, H.; Yuan, Y.; Zhang, B.; Zhu, D.; Zhou, J.; Zhu, Y.; Chen, W. The Clinical Significance of Abnormal Tim-3 Expression on NK Cells from Patients with Gastric Cancer. Immunol. Investig. 2015, 44, 578–589. [Google Scholar] [CrossRef]
  171. Da Silva, I.P.; Gallois, A.; Jimenez-Baranda, S.; Khan, S.; Anderson, A.C.; Kuchroo, V.K.; Osman, I.; Bhardwaj, N. Reversal of NK-cell exhaustion in advanced melanoma by Tim-3 blockade. Cancer Immunol. Res. 2014, 2, 410–422. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Therapeutic approaches involving Natural Killer (NK) cells to treat metastatic cancer. (1) The transfer of expanded and activated NK cells is increasingly used to improve NK cell responses. Autologous NK cells, allogenic NK cells, NK cell lines, such as NK-92 cells, or Chimeric Antigen Receptor (CAR) NK cells may be harnessed as a source of NK cells for adoptive transfer. (2) The activity of NK cells may also be stimulated by cytokine or drug treatment. IL-2, IL-15, and IL-21 are the most interesting agents that potentiate NK cell activity. (3) Therapeutic approaches that engage NK cell activating receptors are widely used in clinics, particularly, mAbs that engage CD16 receptors and induce Antibody-Dependent Cellular Cytotoxicity (ADCC) activity. Trastuzumab or cetuximab are the most widely employed therapeutic strategies involving NK cells used in clinics. The so-called bispecific and trispecific antibodies may improve ADCC activity by redirecting NK cells and T cells to tumor cells. Targeting other activating NK cell receptors, such as NKG2D, or their ligands, such as MHC class I polypeptide-related sequence A (MICA), are innovative strategies that need to be evaluated in clinical trials. (4) Targeting inhibitory receptors and immunosubversive mechanisms developed by cancer cells may release the antitumor potential of NK cells. Blocking antibodies directed against MICA boosts NK cell activity by preventing the shedding of soluble MICA and NKG2D downregulation. Blocking antibodies against inhibitory NK cell receptors or checkpoint proteins, including, but not limited to, Killer cell Immunoglobulin-like Receptors (KIRs) or Natural Killer Group 2A (NKG2A), Programmed Death-1 (PD-1)/PD-L1, TIM-3, CD96, or T cell immunoreceptor with Ig and ITIM domains (TIGIT), have great clinical potential.
Figure 1. Therapeutic approaches involving Natural Killer (NK) cells to treat metastatic cancer. (1) The transfer of expanded and activated NK cells is increasingly used to improve NK cell responses. Autologous NK cells, allogenic NK cells, NK cell lines, such as NK-92 cells, or Chimeric Antigen Receptor (CAR) NK cells may be harnessed as a source of NK cells for adoptive transfer. (2) The activity of NK cells may also be stimulated by cytokine or drug treatment. IL-2, IL-15, and IL-21 are the most interesting agents that potentiate NK cell activity. (3) Therapeutic approaches that engage NK cell activating receptors are widely used in clinics, particularly, mAbs that engage CD16 receptors and induce Antibody-Dependent Cellular Cytotoxicity (ADCC) activity. Trastuzumab or cetuximab are the most widely employed therapeutic strategies involving NK cells used in clinics. The so-called bispecific and trispecific antibodies may improve ADCC activity by redirecting NK cells and T cells to tumor cells. Targeting other activating NK cell receptors, such as NKG2D, or their ligands, such as MHC class I polypeptide-related sequence A (MICA), are innovative strategies that need to be evaluated in clinical trials. (4) Targeting inhibitory receptors and immunosubversive mechanisms developed by cancer cells may release the antitumor potential of NK cells. Blocking antibodies directed against MICA boosts NK cell activity by preventing the shedding of soluble MICA and NKG2D downregulation. Blocking antibodies against inhibitory NK cell receptors or checkpoint proteins, including, but not limited to, Killer cell Immunoglobulin-like Receptors (KIRs) or Natural Killer Group 2A (NKG2A), Programmed Death-1 (PD-1)/PD-L1, TIM-3, CD96, or T cell immunoreceptor with Ig and ITIM domains (TIGIT), have great clinical potential.
Cancers 11 00029 g001
Table 1. The most innovative NK cell immunotherapies for targeting metastasis.
Table 1. The most innovative NK cell immunotherapies for targeting metastasis.
TherapyAdvantagesDisadvantagesReferences
Adoptive transfer
Autologous NK cellsUniversal use. SafeLow efficacy[97,98,99,100,101]
Allogenic NK cellsHighly effective against some KIR-ligand mismatch malignanciesIn clinical evaluation for metastatic cancer; no standard protocols or products[102,103,104,105,106,107,108,109,110]
CAR NK cellsHighly potentiate NK cell antitumor activity; likely to be more efficient and safer than CAR T cellsDifficult to manipulate and expand
In clinical evaluation for metastatic cancer
[111,112,113,114,115,116]
NK cell linesUnlimited, homogeneous, well-defined, and highly active population of NK cells; low costLow efficacy; safety concerns; need to be irradiated[117,118,119,120,121,122,123]
Cytokine-based therapy
IL-2Boost NK cell and T cell activity; significant efficacy in a proportion of melanoma and renal metastatic patientsToxic at high doses; activation of Tregs[124,125,126,127,128,129,130,131,132]
IL-15Similar antitumor activity to IL-2 without activating Tregs and with better toxic profileIn clinical evaluation for metastatic cancer[133,134,135,136,137,138,139,140,141,142,143]
IL-21Boost NK cell and T cell activity without activating Tregs, potential combination with MAbsLow knowledge of IL-21 biology; possible unexpected effects[144,145,146,147,148,149,150,151]
Modulation of receptors or ligands
Tumor-targeting MAbsRedirect NK cell activity against specific tumors Promote ADCC; improve survival of metastatic patientsNeeds combination to improve efficacy[152,153,154,155,156]
Bispecific, trispecific AbsMultiple targets; redirect NK cells and T cells against specific tumors; improve anti-tumor capacityPossible off-target effects[157]
mAb to NKG2D (IPH4301)Prevent immune evasionLack of clinical experience
mAb to MICAPrevent soluble MICA shedding and NKG2D downregulationLack of clinical experience[158]
Targeting inhibitory receptors
PD1/PD-L1 blockadeImpressive clinical results in some metastatic patients; manageable toxicityLack of knowledge of the role of NK cells and the clinical benefits[159,160,161,162,163]
mAbs to KIRsSafe; boost NK cell activity against tumor cellsLow efficacy; need to be combined[164,165]
mAbs to NKG2A
(monalizumab)
Boost NK cell activity against tumor cellsIn clinical evaluation[166]
TIGIT/CD96 blockadeBoost NK cell activity against tumor cells and metastasisIn preclinical studies[167,168]
TIM-3 blockadeBoosts NK cell activity against tumor cells and metastasisIn preclinical studies[169,170,171]
NK: Natural Killer, CAR: Chimeric Antigen Receptor, IL: Interleukin, ADCC: Antibody-Dependent Cellular Cytotoxicity, NKG2D: Natural killer group 2D, MICA: MHC class I polypeptide-related sequence A, KIR: Killer cell Immunoglobulin-like Receptor, TIGIT: T cell immunoreceptor with Ig and ITIM domains, PD1: Programmed Death 1, TIM-3: T-cell Immunoglobulin and Mucin-domain containing-3.

Share and Cite

MDPI and ACS Style

Lorenzo-Herrero, S.; López-Soto, A.; Sordo-Bahamonde, C.; Gonzalez-Rodriguez, A.P.; Vitale, M.; Gonzalez, S. NK Cell-Based Immunotherapy in Cancer Metastasis. Cancers 2019, 11, 29. https://doi.org/10.3390/cancers11010029

AMA Style

Lorenzo-Herrero S, López-Soto A, Sordo-Bahamonde C, Gonzalez-Rodriguez AP, Vitale M, Gonzalez S. NK Cell-Based Immunotherapy in Cancer Metastasis. Cancers. 2019; 11(1):29. https://doi.org/10.3390/cancers11010029

Chicago/Turabian Style

Lorenzo-Herrero, Seila, Alejandro López-Soto, Christian Sordo-Bahamonde, Ana P Gonzalez-Rodriguez, Massimo Vitale, and Segundo Gonzalez. 2019. "NK Cell-Based Immunotherapy in Cancer Metastasis" Cancers 11, no. 1: 29. https://doi.org/10.3390/cancers11010029

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

Article Metrics

Back to TopTop