Next Article in Journal
Overview of Trop-2 in Cancer: From Pre-Clinical Studies to Future Directions in Clinical Settings
Previous Article in Journal
Refining the Characterization and Outcome of Pathological Complete Responders after Neoadjuvant Chemotherapy for Muscle-Invasive Bladder Cancer: Lessons from the Randomized Phase III VESPER (GETUG-AFU V05) Trial
Previous Article in Special Issue
Alterations in Natural Killer Cells in Colorectal Cancer Patients with Stroma AReactive Invasion Front Areas (SARIFA)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Addressing Natural Killer Cell Dysfunction and Plasticity in Cell-Based Cancer Therapeutics

by
Kassandra M. Coyle
1,
Lindsey G. Hawke
2 and
Mark L. Ormiston
1,2,*
1
Department of Medicine, Queen’s University, Kingston, ON K7L3N6, Canada
2
Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L3N6, Canada
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(6), 1743; https://doi.org/10.3390/cancers15061743
Submission received: 2 February 2023 / Revised: 2 March 2023 / Accepted: 8 March 2023 / Published: 13 March 2023
(This article belongs to the Special Issue Natural Killer Cells in Cancer Biology and Therapy)

Abstract

:

Simple Summary

The natural killer (NK) cells of the immune system identify and remove stressed, infected, or cancerous cells in the body. This anti-tumor functionality has been harnessed through promising cell-based therapies that involve the isolation, expansion, activation, and delivery of NK cells for the treatment of several cancers. A variety of techniques have been developed to genetically modify or otherwise improve the activity of these therapeutic NK cells. However, certain elements of the tumor microenvironment are known to alter NK cell functions, suppressing their killing capacity and enhancing their ability to stimulate the formation of blood vessels that support tumor growth. This review summarizes current NK cell-based cancer therapies and discusses improvements that are being pursued to address these mechanisms of tumor-mediated NK cell suppression.

Abstract

Natural killer (NK) cells are cytotoxic group 1 innate lymphoid cells (ILC), known for their role as killers of stressed, cancerous, and virally infected cells. Beyond this cytotoxic function, NK cell subsets can influence broader immune responses through cytokine production and have been linked to central roles in non-immune processes, such as the regulation of vascular remodeling in pregnancy and cancer. Attempts to exploit the anti-tumor functions of NK cells have driven the development of various NK cell-based therapies, which have shown promise in both pre-clinical disease models and early clinical trials. However, certain elements of the tumor microenvironment, such as elevated transforming growth factor (TGF)-β, hypoxia, and indoalemine-2,3-dioxygenase (IDO), are known to suppress NK cell function, potentially limiting the longevity and activity of these approaches. Recent studies have also identified these factors as contributors to NK cell plasticity, defined by the conversion of classical cytotoxic NK cells into poorly cytotoxic, tissue-resident, or ILC1-like phenotypes. This review summarizes the current approaches for NK cell-based cancer therapies and examines the challenges presented by tumor-linked NK cell suppression and plasticity. Ongoing efforts to overcome these challenges are discussed, along with the potential utility of NK cell therapies to applications outside cancer.

1. Introduction

In humans, conventional NK cells can be divided into two distinct subsets: highly cytotoxic CD56dimCD16+ NK cells, which predominate in the peripheral circulation, and CD56brightCD16+/− NK cells, which exhibit reduced cytotoxic function, are primarily localized to tissues and secondary lymphoid organs [1,2,3], and are more commonly associated with the regulation of broad immune responses through the secretion of various cytokines (IFNγ, TNFα, GM-CSF, IL-10, IL-5, and IL-13) and chemokines (MIP-1a, MIP-1β, IL-8, and RANTES) [3,4]. CD56dimCD16+ NK cells make up 90% of peripheral blood and splenic NK cells and exhibit a potent cytolytic activity via the integration of signals from an array of activating and inhibitory receptors [5,6]. In this process, deactivating or inhibitory receptors recognize “self” MHC class-I (MHC-I) molecules that are expressed on the surface of healthy cells and contribute to NK cell self-tolerance. In contrast, virally infected or tumor cells that downregulate surface MHC-I to avoid immune recognition exhibit a “missing self” phenotype, resulting in decreased inhibitory signaling, NK cell degranulation, and lysis [6].
Alternatively, the recognition of antibody-bound target cells by NK cell Fcγ receptors can induce lysis via antibody-dependent cell cytotoxicity (ADCC) [7]. NK cells also express several death ligands with potential cytotoxic activity, such as TNFα, Fas ligand (FasL), and TRAIL. However, only FasL and TRAIL have been shown to induce killing through death receptor-induced apoptosis [8]. In addition to the direct lysis of target cells, the secretion of IFNγ by activated NK cells enhances antiviral, antibacterial, and anti-tumor activity through a variety of mechanisms, including the promotion of macrophage activation, improved dendritic cell antigen presentation, and enhanced lymphocyte endothelium interactions, as well as the regulation of cellular proliferation and apoptosis [9,10,11,12,13,14].

2. NK Cell Therapies

2.1. Primary NK Cell Sources

To date, therapies that aim to harness the anti-tumor function of NK cells have involved either the adoptive transfer of ex vivo expanded NK cells, with or without genetic modification [15,16,17,18], or the promotion of in vivo NK cell activation through the administration of recombinant proteins, such as interleukin-15 (IL-15) superagonists [19,20]. For cell-based approaches, several sources have been explored, including primary NK cells from autologous or allogeneic peripheral blood, apheresis products, or umbilical cord blood (UCB) (Figure 1). Primary NK cells comprise only 10% of all lymphocytes in peripheral blood, limiting the number of cells that can be harvested for therapeutic applications and necessitating extensive purification methods, such as the magnetic depletion of undesired cell types [21]. NK cells are comparatively enriched in UCB, comprising 15–30% of total lymphocytes, but are immature and may be less cytotoxic compared to other sources [22,23].
Alternatively, NK cells can be safely generated from CD34+ hematopoietic progenitors, induced pluripotent stem cells (iPSC), or embryonic stem cells [24,25]. While iPSC-derived NK cells offer a potentially infinite source of homogenously differentiated NK cells and can be functionally enhanced by genetic modification with relative ease, few groups have successfully created functional NK cells from this cellular source [26]. Woan and colleagues reported the use of a clonal iPSC-NK cell line, engineered to express both a high-affinity, non-cleavable version of the CD16a Fc receptor and a membrane-bound IL-15/IL-15 receptor-α fusion protein [27]. These cells demonstrated potent anti-tumor activity in vitro, as well as in myeloma and acute myeloid leukemia (AML) xenograft models. Clinical trials are currently underway [28]. The use of UCB-derived CD34+ cells as a source for NK cell therapies is also being explored by Deverra Therapeutics, through their non-engineered product DVX201 [29].

2.2. Immortalized NK Cell Lines

Immortalized NK cell lines also offer an alternative to primary NK cells, as they can be cultured indefinitely and are amenable to genetic manipulation. Several NK cell lines have been explored for therapeutic use, including NKL, NKG, NK-YS, YT, YTS, NK-92 cells, and high-affinity NK cells (haNKs). Originally isolated from an individual with non-Hodgkin’s lymphoma, NK-92 cells display high levels of anti-tumor cytotoxicity more consistently and reproducibly than other cell lines [30,31], are easily expandable to the clinical grade levels required for treatment [32], and have been used in recent clinical trials in patients with advanced, treatment-refractory malignancies [33,34]. HaNKs are NK-92 cells that have been engineered to express a high-affinity CD16 receptor for increased ADCC and endogenous production of IL-2 to maintain cytotoxic function [35,36]. While advantageous, the clinical application of these cells is not without limitation. NK-92 cells must be irradiated to protect from malignant expansion, potentially limiting their in vivo efficacy and persistence.

2.3. NK Cell Expansion and Activation

Regardless of their source, NK cells must be expanded to generate sufficient numbers of highly functional cells for therapeutic applications (Figure 1). Clinical scale expansion can be achieved using mitotically inactivated feeder cells, such as peripheral blood mononuclear cells (PBMCs), K562 or Jurkat cells, with or without genetic modification [37,38,39]. Relative to freshly isolated NK cells, expanded populations adopt an activated phenotype with increased expression of activating receptors NKG2D, NKG2C, NKp30, NKp44, DNAM-1 and key effector molecules, such as TRAIL, FasL and granzymes [40].
NK cells have also been successfully expanded in feeder-free systems using cell culture flasks, bags, or bioreactors in the presence of high doses of cytokines, such as IL-15 and IL-21 [41,42], cytokine-conjugated magnetic beads [43,44], or plasma membrane-derived particles [45]. However, these methods typically yield lower cell numbers than what is achieved with feeder cell-based expansion. More recently, advanced systems involving dissolvable polymer-based microspheres [46] or streamlined expansion protocols [47] have allowed for improved yields and the incorporation of nonviral genome editing into feeder-free expansion systems.
Although growing evidence supports the safety and efficacy of the majority of these expansion techniques [48], ex vivo NK cell expansion introduces the potential for senescence and exhaustion [49], resulting in the need for expanded NK cells to be activated or “primed” with cytokines like IL-2, IL-12, IL-15, and IL-18 to achieve maximal efficacy on tumors [50,51]. These cytokines, along with IL-21 and type I interferons, are central to the maturation, activation and survival of NK cells. IL-2 is critical to NK activation, can rejuvenate exhausted NK cells, and restores or preserves their cytotoxic potential in response to various stressors or following exposure to multiple myeloma [52,53,54]. Similarly, the stimulation of NK cells with various combinations of IL-2, IL-12, IL-15 and IL-18 increases the production of critical effector cytokines like IFNγ, IL-8, and TNF-α [55], enhances the responsiveness of the cells to the integrin, LFA-1, and improves subsequent receptor stimulation [56].

2.4. Alternative Approaches for NK Cell Activation

In addition to cytokine-based approaches, genetic manipulations, such as transgenic expression of the Chimeric Antigen Receptor (CAR), have also been explored as a means to enhance NK cell activation for therapeutic applications (Figure 1) [57]. CAR is a recombinant protein that can be introduced into cytotoxic lymphocytes to enable tumor antigen recognition and trigger activation [58]. CAR expression allows NK cells to specifically target cancer cells through recognition of tumor associated antigens. The generation of CAR-NK cells involves transducing isolated NK cells with CAR-encoding genes and expanding these cells prior to adoptive transfer into the patient [59]. Recently, UCB-derived CAR-NK cells were used to treat patients with relapsed or refractory CD19+ cancers, resulting in complete remission in the majority of patients without significant toxic effects [60]. Multiple clinical trials are currently underway [61], and ongoing studies are investigating the use of CAR-NK cells for the treatment of non-malignant targets, such as HIV-infected cells [62,63].
Despite these advantages, the complex CAR-NK cell manufacturing process is hindered by inefficient transduction methods [64], as well as the poor in vivo persistence shared by many NK cell therapy platforms. The personalized approach required for CAR-NK cells is also extremely expensive, time consuming, and difficult to apply large-scale. These limitations, which are not exclusive to CAR-NK approaches (Table 1), demonstrate a clear need for effective “off-the-shelf” therapies. One such approach is bi- and tri-specific antibodies (BiKEs and TriKEs), which trigger ADCC by binding both CD16 on NK cells and specific tumor antigens, creating a connection-like bridge between these cells and allowing for NK cell activation [65]. TriKEs have also been modified to incorporate cytokines like IL-15 to increase in vivo persistence and activation of NK cells [66]. In vitro, TriKEs have demonstrated enhanced NK cell activation and killing of both AML cell lines and patient-derived AML blasts [67]. A clinical trial in the treatment of high-risk hematological malignancies is currently underway [68].
The NK cell engager platform (ANKET) has also been investigated as a means to harness NK cells as next-generation cancer immunotherapies. These NK cell engagers link a monoclonal antibody targeting the activating NK cell receptor NKp46 (or NKp30), an Fc fragment to promote ADCC via CD16, and an antibody targeting a tumor associated antigen, to enable the tumor-localized activation of host NK cells [76]. Work with these engagers has shown promise in targeting various cancer cell lines [76,77,78]. Recently, Demaria and colleagues reported a tetraspecific CD20-ANKET, which engages NKp46, CD16a, the beta chain of the IL-2 receptor, and a tumor associated antigen to induce preferential NK cell activation and target cell killing [79]. This tetraspecific CD20-ANKET induced the local control of tumors in non-human primates, without significant adverse side effects [79].

3. NK Cell Impairment in Cancer

Even though extensive progress has been made towards the efficient isolation, expansion and functional enhancement of NK cells for therapeutic applications, these approaches are still limited by disease-associated environmental challenges that suppress or modify NK cell function in vivo. NK cell dysfunction is a major component of the development, progression, and survival of many cancer models and is strongly correlated with worsened survival outcomes in patients [80,81]. A decrease in circulating NK cells is often accompanied by poor infiltration of NK cells into the tumor microenvironment [3,82,83,84,85]. NK cells isolated from tumors also produce less IFNγ, CD107a, granzyme B, and perforin when compared with NK cells from peritumor regions or peripheral blood [83,85,86] and exhibit both a downregulation of NK activating receptors, including NKG2D, CD16, NCRs (NKp30, NKp44 and NKp46), CD226, and 2B4 [83,87,88,89], and elevated inhibitory receptors, such as NKG2A [86].
These functional impairments have been attributed to several cellular processes, including exhaustion, where persistent NK activation results in decreased effector functions and poor control of malignancies and infections [90], as well as anergy, which is defined as a hypo-reactive state that is attributed to either excessive inhibitory signaling [91] or NK activation in the absence of proper licensing via inhibitory receptor binding [92,93]. NK cell dysfunction can also arise from the deprivation of survival signaling from cytokines like IL-15 [94], or as a result of suppressive signals in the tumor microenvironment, such as TGFβ, hypoxia and IDO, which are discussed in detail below (Figure 2). In addition to the more classical forms of NK functional impairment, recent work has also identified substantial potential for plasticity between ILC1 subsets, through which tumor-associated factors can drive the conversion of conventional cytotoxic NK cells towards tissue-resident or ILC1-like phenotypes that are either poorly cytotoxic or may even aid in tumor growth via the promotion of enhanced tumor vascularization [95,96,97,98,99]. Together, these mechanisms of impairment demonstrate how the tumor microenvironment can reduce the efficacy of conventional NK cell-based therapeutics and highlight the need to both understand and specifically target disease-mediated NK cell suppression to increase the performance of next-generation therapeutic approaches.

NK Cell Plasticity

Since the first identification of distinct ILC populations, group 1 ILCs have been classified into two subgroups: conventional NK cells, which express cytotoxic factors like IFNγ, perforin, and granzyme B, as well as the transcription factors T-bet and Eomes, and ILC1 cells, which are Eomes-negative, poorly cytotoxic and largely reside in non-lymphoid tissues, such as the skin, liver and uterus [100,101]. Although these subsets were originally believed to be the developmentally distinct progeny of discrete lymphoid precursors, later work identified the potential for factors such as TGFβ to induce the conversion of circulating NK cells into intermediate ILC1 or ILC1-like phenotypes, characterized by a reduction in cytotoxic capacity, the acquisition of ILC1-linked surface markers, and, for ILC1-like cells, a reduction in Eomes expression [102]. Importantly, converted ILC1s and intermediate ILC1s are unable to control tumor growth and metastasis, providing a direct mechanism by which this plasticity can facilitate the escape of tumors from NK cell-mediated immunosurveillance. This process of conversion is not dependent upon canonical TGFβ signaling via the Smad family of signal transducers [103], but instead relies on non-canonical TAK1-mediated activation of p38 MAP Kinase via a process that is synergistically enhanced by IL-15 [104]. The spontaneous conversion of NK cell to an ILC1-like phenotype has been demonstrated in mice harboring a conditional deletion of the Smad4 gene in their Group 1 ILCs [103], confirming that canonical signaling via Smad4 actually works to prevent conversion via non-canonical TGFβ pathways. Importantly, these Smad4-deficient mice are also unable to control tumor metastasis or viral infection, highlighting the link between NK plasticity and cancer.

4. TGFβ-Mediated NK Cell Impairment

Beyond its specific actions on NK cell plasticity, TGFβ is a major immunosuppressive cytokine that correlates with poor prognosis and reduced NK cell activity in multiple cancer types [105,106]. In pancreatic cancer, membrane-bound TGFβ1 on cancer-expanded myeloid-derived suppressor cells (MDSC) was found to induce NK cell dysfunction through diminished NKG2D expression and impaired NK cytotoxicity [107,108]. TGFβ downregulates the expression of NKG2D on NK cells and CD8+ T cells in several cancer models [105,106,107,109], and has been shown to suppress IL-15 and STAT5-mediated NK cell activation through the blockade of mTOR activation [110]. Importantly, NK cell-specific deletion of the TGFβ type II receptor (TGFβRII) causes enhanced anti-tumor activity in multiple mouse models of metastasis, whereas constitutive TGFβ signaling arrests NK cell development and drives increased tumor growth [110]. These effects of TGFβ were linked to a direct inhibition of mTOR-dependent metabolic activity in NK cells stimulated by IL-15, and point to the stimulation of NK cell metabolic activity as a potential strategy to promote or enhance NK cell-based cancer therapies.
As detailed above, TGFβ is also known to impair the cytotoxic function of NK cells by inducing the conversion of cytotoxic circulating NK cells to a poorly cytotoxic tissue-resident or ILC1-like cell type [104,111]. In mice, CD49a+CD49b tissue-resident NK cells in tissues such as the liver, skin and uterus can be distinguished from circulating NK cells, which are CD49aCD49b+. Uterine NK (uNK) cells are among the best-defined of these tissue-resident NK cell subsets [112,113]. During the initial stages of pregnancy, uNKs increase in number at the site of embryo implantation [114,115,116,117,118] to accompany extensive uterine vascular remodelling that allows for a 10-fold increase in blood supply to the intervillous space [119]. In pregnancy, uNK-derived IFNγ, matrix metalloproteinases (MMPs), and angiogenic growth factors such as vascular endothelial growth factor (VEGF), placental growth factor (PIGF) and angiopoietin (ANG) 1 and 2, contribute to uterine vascular remodeling and immunoregulation during normal pregnancy [112,120].
In addition to TGFβ, which can drive the conversion of circulating cytotoxic NK cells to poorly cytotoxic ILC1-like cells with impaired anti-tumor function and tissue-resident surface marker expression [103,121], exposing circulating NK cells to conditioned medium from decidual stromal cells or a combination of TGFβ, hypoxia, IDO, and demethylating agents, has also been shown to promote the acquisition of uNK phenotypic and functional attributes by peripheral blood NK cells [104,122,123,124]. These attributes include reduced cytotoxicity and the expression of adhesion molecules, such as CD9 and CD103, which are directly linked to tissue residency. While several studies have associated this conversion with the enhanced production of VEGF [125] and have shown that NK-derived VEGF is a critical regulator of tumor vascularization and growth in mouse models [126], in vitro studies examining the discrete effects of TGFβ and hypoxia on cultured human peripheral NK cells have shown that VEGF production is largely hypoxia dependent and is not enhanced by the process of TGFβ-mediated NK cell conversion [124]. These findings do leave open the possibility of a two-stage model, whereby TGFβ promotes the movement of NK cells into tissues, and the resultant reduction in oxygen tension drives VEGF production (Figure 2A).

Current Work Targeting the TGFβ-NK Cell Axis

Multiple studies have targeted TGFβ-mediated immunosuppression in cancer through the use of TGFβ ligand traps or receptor kinase inhibitors [127,128,129,130,131,132]. While many of these works demonstrated beneficial effects via the preservation of NK cell function [133], these strategies, which are based on the global blockade of TGFβ signaling, are unlikely to translate into effective therapies due to unwanted side-effects, including autoimmune inflammation and cardiovascular toxicity [134,135]. As an alternative, more targeted approaches that directly address the impact of TGFβ on NK cell phenotype and function are currently being explored and have shown promise in early pre-clinical studies (Figure 3A). UCB-derived NK cells, modified to express a dominant-negative TGFβ receptor that is coupled to an NK-specific activating signal, demonstrated higher cytotoxic activity in TGFβ-rich environments, both in vitro and in vivo [136]. These TGFβ receptor-modified NK cells are phenotypically and functionally similar to unmodified cells, with an added protection against exogenous TGFβ in a xenograft model of neuroblastoma. Similarly, the genetic modification of NK-92 cells to express a chimeric receptor with a TGFβRII extracellular and transmembrane domain, linked to an NK-activating NKG2D intracellular domain, exhibited resistance to TGFβ, a higher killing capacity, and elevated IFNγ production, as well as improved inhibition of tumor growth in vivo [137]. NK-92 cells modified to express a dominant negative TGFβRII also demonstrated insensitivity to TGFβ-mediated suppression in vitro and a capacity to decrease tumor proliferation, reduce lung metastasis, and enhance the survival of mice in a lung cancer model [138].

5. Hypoxia-Mediated NK Cell Impairment

In certain tumors, the absence of a functional circulation can give rise to a hypoxic microenvironment, altering the function of multiple immune cell types and favoring disease progression (Figure 2B). NK cells are sensitive to hypoxia, as their cytolytic function is impaired and expression of the activating receptors, NKp46, NKp30, NKp44 and NKG2D, is reduced [139,140,141,142]. In response to hypoxia, NK cells upregulate hypoxia-inducible factor 1α (HIF-1α), a transcription factor that regulates oxidative and glycolytic cellular metabolism, increasing anaerobic metabolism, decreasing mitochondrial oxygen consumption, and driving alterations in the NK cell transcriptome that govern the adaptation to oxygen-depleted environments [143,144]. IL-2 primed NK cells subjected to short-term (16 h) and prolonged (96 h) hypoxia are functionally reprogrammed, exhibiting differential expression of a large number of genes, including proinflammatory cytokines, chemokines, and chemokine-receptors [145]. These changes result in increased NK cell migration and decreased cytotoxicity [146], as well as reduced secretion of IFNγ, TNF-α, GM-CSF, and members of the CC chemokine family (CC3 and CC5) [145].
Additionally, increased HIF-1α expression by tumor cells can decrease NK cell-mediated killing by downregulating tumor-derived MHC class I chain-related genes [147] or by promoting the shedding of MICA, a ligand that triggers the cytolytic action of immune effectors, from the surface of tumor cells [148,149]. This HIF-1α-mediated NK cell evasion is thought to be linked to increased expression of the metalloproteinase ADAM10, which is required for the hypoxia-induced shedding of MICA [150].
As detailed above, exposure to hypoxia can promote NK cell-mediated angiogenesis via HIF-1α-induced VEGF production [123,145]. Targeting this process may have therapeutic potential, as mice bearing an NK-specific deletion of VEGF or HIF-1α exhibit reduced growth of solid tumors and impaired tumor vascularization [151,152]. Of note, tumors from mice lacking HIF-1α in their NK cells exhibited high-density networks of immature, non-functional vessels that were attributed to the reduced infiltration of sVEGFR1-expressing NK cells into hypoxic regions of the tumor, increased VEGF bioavailability, and non-productive angiogenesis [152].

Current Work Targeting Hypoxic NK Cells

Inhibition of HIF-1α with the small molecule KC7F2 has been explored as a potential avenue for cancer therapy. Single-cell RNA sequencing of tumor infiltrating NK cells revealed that HIF-1α inhibition maintained the expression of activation markers and effector molecules, while preserving cytotoxic activity in hypoxic conditions [153]. Moreover, the conditional deletion of HIF-1α in NK cells decreased tumor burden and enhanced survival in several cancer models, including RMA-S lymphoma, Lewis lung carcinoma and B16-Rae1 melanoma [153].
Downregulation of HIF-1a signaling within the tumor itself can also allow for enhanced immune activity. Intratumoral gene transfer of an antisense HIF-1α plasmid has been shown to downregulate VEGF and decrease tumor microvessel density in an EL-4 mouse lymphoma model, resulting in the complete and permanent NK cell rejection of small tumors (0.1 cm in diameter) [154]. When synergized with T cell co-stimulatory B7-1-mediated immunotherapy, this antisense HIF-1α therapy caused the NK cell-dependent rejection of larger EL-4 tumors up to 0.4 cm in diameter that were refractory to other monotherapies. Further, mice exposed to this combination therapy resisted a rechallenge with parental tumor cells, indicating systemic anti-tumor immunity [154].
Beyond the direct targeting of HIF-1α, IL-2 activation can also restore the killing potential of NK cells that have been exposed to hypoxia, allowing for maintained degranulation in response to target multiple myeloma cells, despite hypoxic conditions [54]. Although off-target effects hinder clinical feasibility of systemic recombinant IL-2 treatment, targeted approaches, such as haNK cells, which express a high-affinity CD16 receptor and internal IL-2, offer the potential of sustained killing under hypoxic conditions, when compared to healthy donor NK cells whose cytolytic abilities are impaired (Figure 3B) [155].

6. IDO-Mediated NK Cell Impairment

IDO is an intracellular enzyme that catabolizes tryptophan to N-formylkynurenine, which is then converted into extracellular messengers that are collectively known as kynurenines [156]. In addition to its broad immunosuppressive activity in autoimmune diseases [157,158,159], chronic infections [160], and cancer [161], IDO-derived kynurenine exposure induces NK cell apoptosis through a reactive oxygen species-mediated pathway and impairs NK cell function by suppressing surface expression of the NK activating receptors NKp46 and NKG2D (Figure 2C) [162,163,164]. Short hairpin RNA silencing of IDO in ovarian cancer cells has been shown to reinforce cancer cell sensitivity to NK cells in vitro, while also reducing tumor growth, decreasing peritoneal dissemination, and promoting NK cell accumulation in the tumor stroma in vivo [165]. Mechanistic studies examining the impairment of NK cells by IDO have shown that high IDO expression significantly reduces NK cell cytotoxicity via the miR-18a/NKG2D/NKGD2L regulatory axis, providing for a useful target for specific future manipulations [166].

Current Work Targeting the IDO-NK Cell Pathway

Based on its influence on NK cells, as well as other immune cells, IDO inhibition has arisen as a potential target for anti-cancer therapeutics. Several IDO inhibitors are being tested in both pre-clinical and clinical trials (Figure 3C) [167]. Most inhibitors bind the IDO enzyme, preventing the conversion of tryptophan into the kynurenines that enact toxic effects on NK cells and other immune substrates. Global IDO inhibition using the competitive IDO inhibitor, 1-methyl-L-tryptophan (1MT) slowed tumor growth and enhanced tumor rejection in mice injected with IDO-expressing P815B cells [161]. IDO inhibition by 1MT also cooperates with several chemotherapeutic agents to effectively promote regression of established breast cancer tumors that were previously resistant to chemotherapy, suggesting that, while minimally effective on its own, 1MT intervention may be best used in combination with other anti-cancer therapies [168]. In a breast cancer model, the direct administration of 1MT directly into tumor grafts slowed tumor outgrowth, whereas the combination of 1MT with paclitaxel resulted in a 30% decrease in tumor volume within 2 weeks of initiating therapy [168]. Similarly, combining 1MT with the nicotinamide phosphoribosyl transferase (NAMPT) inhibitor, APO866, resulted in a greater effect on both murine gastric and bladder tumor models than either treatment alone [169].
Despite these successes, there is also significant controversy regarding the efficacy of these IDO inhibitors. Many of the current inhibitors have low in vitro activity. 1MT and Norharmane inhibit IDO on micromolar levels, while Epacadostat and Navoximod exert inhibitory effects in the nanomolar range [170]. As such, the in vivo efficacy of these inhibitors has been brought into question. 1MT also exists as two stereoisomers, with potentially different biological properties. It is currently unclear which isomer might be preferable for therapeutic development [171,172]. Beyond these limitations, there is also limited data available on the direct actions of these therapeutics on NK cell function, both in vitro and in vivo.

7. Conclusions and Future Perspectives

The NK cell is a highly dynamic member of the immune system that can adapt its phenotype in response to microenvironmental conditions that vary between the circulation and healthy or diseased tissues. Extracellular influences, such as TGFβ and hypoxia, can promote the phenotypic shift in NK cells to a more tissue-resident or ILC1-like phenotype, diminishing their powerful cytotoxic functions in favor of a cytokine-producing cell type with the potential to enhance tumor angiogenesis and growth. While the recent success of CAR-T cell therapies highlights the feasibility of cell-based approaches for enhancing anti-tumor immunity in human subjects, these mechanisms of in vivo suppression represent potentially major limitations on the persistence, efficacy, and, ultimately, the clinical translatability of NK cell-based therapeutics for the treatment of cancer. Ongoing pre-clinical studies exploring the specific targeting of these mechanisms of NK cell suppression, either via the genetic modification of primary or immortalized NK cells, the systemic administration of therapeutic agents, or the targeting of shared mechanisms, such as reduced NK cell bioenergetic metabolism, not only show promise for the treatment of individual cancers, but may one day benefit patients with other diseases linked to NK cell impairment.
NK cell-based therapies are already being investigated [54,155,156,157] in the treatment of HIV [173,174]. Additionally, conditions such as endometriosis and pulmonary arterial hypertension (PAH), a disease of obstructive remodeling in the pulmonary circulation, are also linked to NK cell impairment associated with excessive TGFβ, IDO and HIF activity. It is therefore possible that future NK-cell-based therapies with improved selectivity and efficacy may eventually be explored for these conditions. In endometriosis, crosstalk between endometrial stromal cells and macrophages, and their secretion of IL-10/TGFβ impairs NK cell cytotoxicity, viability, and phenotype via a process that can be partially reversed by anti-TGFβ therapy [175]. Peritoneal fluid analysis from women with endometriosis also identified high levels of IDO, which reduced expression of the activating receptors NKp46 and NKG2D [176].
In PAH, NK cell dysfunction is a feature of both human patients and animal models of disease [177]. NK cells from PAH patients exhibit excessive TGFβ signaling, linked to the reduced expression of activating and inhibitory receptors. Interestingly, pulmonary hypertension develops spontaneously in NK cell-deficient mice [178], suggesting a potentially causal role for NK cells in the development of this disease. Together, these findings point to a future for NK cell-based therapeutics that may extend beyond cancer. With the recent initiation of multiple ongoing clinical trials, and the rise of advanced methods for the expansion, activation, and genetic modification of NK cells for therapeutic applications, the next decade of NK cell therapies offers great promise for translational advances across a wide range of applications.

Author Contributions

Conceptualization, K.M.C. and M.L.O.; writing K.M.C. and MLO; review and editing, K.M.C., L.G.H. and M.L.O.; supervision, M.L.O.; funding acquisition, M.L.O. All authors have read and agreed to the published version of the manuscript.

Funding

This manuscript was funded by the Canadian Institutes of Health Research, grant number PJT-180356.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Caligiuri, M.A. Human Natural Killer Cells. Blood J. Am. Soc. Hematol. 2008, 112, 461–469. [Google Scholar] [CrossRef]
  2. Moretta, A.; Marcenaro, E.; Parolini, S.; Ferlazzo, G.; Moretta, L. NK Cells at the Interface between Innate and Adaptive Immunity. Cell Death Differ. 2008, 15, 226–233. [Google Scholar] [CrossRef] [Green Version]
  3. Fauriat, C.; Long, E.O.; Ljunggren, H.-G.; Bryceson, Y.T. Regulation of Human NK-Cell Cytokine and Chemokine Production by Target Cell Recognition. Blood J. Am. Soc. Hematol. 2010, 115, 2167–2176. [Google Scholar] [CrossRef] [Green Version]
  4. Bluman, E.M.; Bartynski, K.J.; Avalos, B.R.; Caligiuri, M.A. Human Natural Killer Cells Produce Abundant Macrophage Inflammatory Protein-1 Alpha in Response to Monocyte-Derived Cytokines. J. Clin. Investig. 1996, 97, 2722–2727. [Google Scholar] [CrossRef]
  5. Ljunggren, H.-G.; Kärre, K. In Search of the ‘Missing Self’: MHC Molecules and NK Cell Recognition. Immunol. Today 1990, 11, 237–244. [Google Scholar] [CrossRef]
  6. Kärre, K. NK Cells, MHC Class I Molecules and the Missing Self. Scand. J. Immunol. 2002, 55, 221–228. [Google Scholar] [CrossRef]
  7. Vivier, E.; Tomasello, E.; Baratin, M.; Walzer, T.; Ugolini, S. Functions of Natural Killer Cells. Nat. Immunol. 2008, 9, 503–510. [Google Scholar] [CrossRef]
  8. Ramírez-Labrada, A.; Pesini, C.; Santiago, L.; Hidalgo, S.; Calvo-Pérez, A.; Oñate, C.; Andrés-Tovar, A.; Garzón-Tituaña, M.; Uranga-Murillo, I.; Arias, M.A.; et al. All About (NK Cell-Mediated) Death in Two Acts and an Unexpected Encore: Initiation, Execution and Activation of Adaptive Immunity. Front. Immunol. 2022, 13, 896228. [Google Scholar] [CrossRef]
  9. Paul, S.; Kulkarni, N.; Shilpi; Lal, G. Intratumoral Natural Killer Cells Show Reduced Effector and Cytolytic Properties and Control the Differentiation of Effector Th1 Cells. Oncoimmunology 2016, 5, e1235106. [Google Scholar] [CrossRef] [Green Version]
  10. Chaix, J.; Tessmer, M.S.; Hoebe, K.; Fuséri, N.; Ryffel, B.; Dalod, M.; Alexopoulou, L.; Beutler, B.; Brossay, L.; Vivier, E. Cutting Edge: Priming of NK Cells by IL-18. J. Immunol. 2008, 181, 1627–1631. [Google Scholar] [CrossRef] [Green Version]
  11. Ortaldo, J.R.; Winkler-Pickett, R.; Wigginton, J.; Horner, M.; Bere, E.W.; Mason, A.T.; Bhat, N.; Cherry, J.; Sanford, M.; Hodge, D.L. Regulation of ITAM-Positive Receptors: Role of IL-12 and IL-18. Blood 2006, 107, 1468–1475. [Google Scholar] [CrossRef] [Green Version]
  12. Cai, G.; Kastelein, R.A.; Hunter, C.A. IL-10 Enhances NK Cell Proliferation, Cytotoxicity and Production of IFN-γ When Combined with IL-18. Eur. J. Immunol. 1999, 29, 2658–2665. [Google Scholar] [CrossRef]
  13. Billiau, A. Interferon-γ: Biology and Role in Pathogenesis. In Advances in Immunology; Dixon, F.J., Ed.; Academic Press: Cambridge, MA, USA, 1996; Volume 62, pp. 61–130. ISBN 0065-2776. [Google Scholar]
  14. Boehm, U.; Klamp, T.; Groot, M.; Howard, J.C. CELLULAR RESPONSES TO INTERFERON-γ. Annu. Rev. Immunol. 1997, 15, 749–795. [Google Scholar] [CrossRef]
  15. Hercend, T.; Farace, F.; Baume, D.; Charpentier, F.; Droz, J.-P.; Triebel, F.; Escudier, B. Immunotherapy with Lymphokine-Activated Natural Killer Cells and Recombinant Interleukin-2: A Feasibility Trial in Metastatic Renal Cell Carcinoma. J. Biol. Response Mod. 1990, 9, 546–555. [Google Scholar]
  16. Miller, J.S.; Tessmer-Tuck, J.; Pierson, B.A.; Weisdorf, D.; McGlave, P.; Blazar, B.R.; Katsanis, E.; Verfaillie, C.; Lebkowski, J.; Radford, J., Jr. Low Dose Subcutaneous Interleukin-2 after Autologous Transplantation Generates Sustained In Vivo Natural Killer Cell Activity. Biol. Blood Marrow Transpl. 1997, 3, 34–44. [Google Scholar]
  17. Burns, L.J.; Weisdorf, D.J.; DeFor, T.E.; Vesole, D.H.; Repka, T.L.; Blazar, B.R.; Burger, S.R.; Panoskaltsis-Mortari, A.; Keever-Taylor, C.A.; Zhang, M.J. IL-2-Based Immunotherapy after Autologous Transplantation for Lymphoma and Breast Cancer Induces Immune Activation and Cytokine Release: A Phase I/II Trial. Bone Marrow Transpl. 2003, 32, 177–186. [Google Scholar] [CrossRef] [Green Version]
  18. Liang, S.; Xu, K.; Niu, L.; Wang, X.; Liang, Y.; Zhang, M.; Chen, J.; Lin, M. Comparison of Autogeneic and Allogeneic Natural Killer Cells Immunotherapy on the Clinical Outcome of Recurrent Breast Cancer. Onco Targets Ther. 2017, 10, 4273. [Google Scholar] [CrossRef] [Green Version]
  19. Romee, R.; Cooley, S.; Berrien-Elliott, M.M.; Westervelt, P.; Verneris, M.R.; Wagner, J.E.; Weisdorf, D.J.; Blazar, B.R.; Ustun, C.; Defor, T.E.; et al. First-in-Human Phase 1 Clinical Study of the IL-15 Superagonist Complex ALT-803 to Treat Relapse after Transplantation. Blood J. Am. Soc. Hematol. 2018, 131, 2515–2527. [Google Scholar] [CrossRef] [Green Version]
  20. Rhode, P.R.; Egan, J.O.; Xu, W.; Hong, H.; Webb, G.M.; Chen, X.; Liu, B.; Zhu, X.; Wen, J.; You, L.; et al. Comparison of the Superagonist Complex, ALT-803, to IL15 as Cancer Immunotherapeutics in Animal Models. Cancer Immunol. Res. 2016, 4, 49–60. [Google Scholar] [CrossRef] [Green Version]
  21. Koehl, U.; Brehm, C.; Huenecke, S.; Kloess, S.; Bremm, M.; Zimmermann, S.Y.; Ullrich, E.; Soerensen, J.; Quaiser, A.; Erben, S.; et al. Clinical Grade Purification and Expansion of NK Cell Products for an Optimized Manufacturing Protocol. Front. Oncol. 2013, 3, 118. [Google Scholar] [CrossRef] [Green Version]
  22. Harris, D.T.; Schumacher, M.J.; Locascio, J.; Besencon, F.J.; Olson, G.B.; DeLuca, D.; Shenker, L.; Bard, J.; Boyse, E.A. Phenotypic and Functional Immaturity of Human Umbilical Cord Blood T Lymphocytes. Proc. Natl. Acad. Sci. 1992, 89, 10006–10010. [Google Scholar] [CrossRef] [Green Version]
  23. Luevano, M.; Daryouzeh, M.; Alnabhan, R.; Querol, S.; Khakoo, S.; Madrigal, A.; Saudemont, A. The Unique Profile of Cord Blood Natural Killer Cells Balances Incomplete Maturation and Effective Killing Function upon Activation. Hum. Immunol. 2012, 73, 248–257. [Google Scholar] [CrossRef]
  24. Dolstra, H.; Roeven, M.W.H.; Spanholtz, J.; Hangalapura, B.N.; Tordoir, M.; Maas, F.; Leenders, M.; Bohme, F.; Kok, N.; Trilsbeek, C. Successful Transfer of Umbilical Cord Blood CD34+ Hematopoietic Stem and Progenitor-Derived NK Cells in Older Acute Myeloid Leukemia PatientsHSPC-NK Cell Adoptive Transfer in Older AML Patients. Clin. Cancer Res. 2017, 23, 4107–4118. [Google Scholar] [CrossRef] [Green Version]
  25. Spanholtz, J.; Tordoir, M.; Eissens, D.; Preijers, F.; van der Meer, A.; Joosten, I.; Schaap, N.; de Witte, T.M.; Dolstra, H. High Log-Scale Expansion of Functional Human Natural Killer Cells from Umbilical Cord Blood CD34-Positive Cells for Adoptive Cancer Immunotherapy. PLoS ONE 2010, 5, e9221. [Google Scholar] [CrossRef]
  26. Knorr, D.A.; Ni, Z.; Hermanson, D.; Hexum, M.K.; Bendzick, L.; Cooper, L.J.N.; Lee, D.A.; Kaufman, D.S. Clinical-Scale Derivation of Natural Killer Cells from Human Pluripotent Stem Cells for Cancer Therapy. Stem Cells Transl. Med. 2013, 2, 274–283. [Google Scholar] [CrossRef]
  27. Woan, K.V.; Kim, H.; Bjordahl, R.; Davis, Z.B.; Gaidarova, S.; Goulding, J.; Hancock, B.; Mahmood, S.; Abujarour, R.; Wang, H. Harnessing Features of Adaptive NK Cells to Generate IPSC-Derived NK Cells for Enhanced Immunotherapy. Cell Stem Cell 2021, 28, 2062–2075. [Google Scholar] [CrossRef]
  28. FT538 in Subjects with Advanced Hematologic Malignancies—Full Text View ClinicalTrials.gov. (n.d.). Available online: https://clinicaltrials.gov/ct2/show/NCT04614636. (accessed on 10 March 2023).
  29. Safety and Efficacy of Allogeneic NK Cell Infusions in Patients with Relapsed/Refractory AML and High Risk MDS—Full Text View ClinicalTrials.gov. (n.d.). Available online: https://clinicaltrials.gov/ct2/show/NCT04901416 (accessed on 10 March 2023).
  30. Gong, J.H.; Maki, G.; Klingemann, H.G. Characterization of a Human Cell Line (NK-92) with Phenotypical and Functional Characteristics of Activated Natural Killer Cells. Leukemia 1994, 8, 652–658. [Google Scholar]
  31. Klingemann, H.; Boissel, L.; Toneguzzo, F. Natural Killer Cells for Immunotherapy—Advantages of the NK-92 Cell Line over Blood NK Cells. Front. Immunol. 2016, 7, 91. [Google Scholar] [CrossRef] [Green Version]
  32. Tarn, Y.K.; Martinson, J.A.; Doligosa, K.; Klingernann, H.-G. Ex Vivo Expansion of the Highly Cytotoxic Human Natural Killer Cell Line NK-92 under Current Good Manufacturing Practice Conditions for Clinical Adoptive Cellular Immunotherapy. Cytotherapy 2003, 5, 259–272. [Google Scholar] [CrossRef]
  33. Boyiadzis, M.; Agha, M.; Redner, R.L.; Sehgal, A.; Im, A.; Hou, J.-Z.; Farah, R.; Dorritie, K.A.; Raptis, A.; Lim, S.H.; et al. Phase 1 Clinical Trial of Adoptive Immunotherapy Using “off-the-Shelf” Activated Natural Killer Cells in Patients with Refractory and Relapsed Acute Myeloid Leukemia. Cytotherapy 2017, 19, 1225–1232. [Google Scholar] [CrossRef]
  34. 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]
  35. Jochems, C.; Hodge, J.W.; Fantini, M.; Fujii, R.; Maurice Morillon, Y.I.; Greiner, J.W.; Padget, M.R.; Tritsch, S.R.; Yok Tsang, K.; Campbell, K.S.; et al. An NK Cell Line (HaNK) Expressing High Levels of Granzyme and Engineered to Express the High Affinity CD16 Allele. Oncotarget 2016, 7, 86359. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Fabian, K.P.; Hodge, J.W. The Emerging Role of Off-the-Shelf Engineered Natural Killer Cells in Targeted Cancer Immunotherapy. Mol. Ther. Oncolytics 2021, 23, 266–276. [Google Scholar] [CrossRef] [PubMed]
  37. Bae, D.S.; Lee, J.K. Development of NK Cell Expansion Methods Using Feeder Cells from Human Myelogenous Leukemia Cell Line. Blood Res. 2014, 49, 154–161. [Google Scholar] [CrossRef] [PubMed]
  38. Kweon, S.; Phan, M.T.T.; Chun, S.; Yu, H.B.; Kim, J.; Kim, S.; Lee, J.; Ali, A.K.; Lee, S.H.; Kim, S.K.; et al. Expansion of Human NK Cells Using K562 Cells Expressing OX40 Ligand and Short Exposure to IL-21. Front. Immunol. 2019, 10, 879. [Google Scholar] [CrossRef] [Green Version]
  39. Lim, S.A.; Kim, T.J.; Lee, J.E.; Sonn, C.H.; Kim, K.; Kim, J.; Choi, J.G.; Choi, I.K.; Yun, C.O.; Kim, J.H.; et al. Ex Vivo Expansion of Highly Cytotoxic Human NK Cells by Cocultivation with Irradiated Tumor Cells for Adoptive Immunotherapy. Cancer Res. 2013, 73, 2598–2607. [Google Scholar] [CrossRef] [Green Version]
  40. Gurney, M.; Kundu, S.; Pandey, S.; O’Dwyer, M. Feeder Cells at the Interface of Natural Killer Cell Activation, Expansion and Gene Editing. Front. Immunol. 2022, 13, 802906. [Google Scholar] [CrossRef]
  41. Sutlu, T.; Stellan, B.; Gilljam, M.; Quezada, H.C.; Nahi, H.; Gahrton, G.; Alici, E. Clinical-Grade, Large-Scale, Feeder-Free Expansion of Highly Active Human Natural Killer Cells for Adoptive Immunotherapy Using an Automated Bioreactor. Cytotherapy 2010, 12, 1044–1055. [Google Scholar] [CrossRef] [Green Version]
  42. Wagner, J.; Pfannenstiel, V.; Waldmann, A.; Bergs, J.W.J.; Brill, B.; Huenecke, S.; Klingebiel, T.; Rödel, F.; Buchholz, C.J.; Wels, W.S. A Two-Phase Expansion Protocol Combining Interleukin (IL)-15 and IL-21 Improves Natural Killer Cell Proliferation and Cytotoxicity against Rhabdomyosarcoma. Front. Immunol. 2017, 8, 676. [Google Scholar] [CrossRef] [Green Version]
  43. Li, X.; He, C.; Liu, C.; Ma, J.; Ma, P.; Cui, H.; Tao, H.; Gao, B. Expansion of NK Cells from PBMCs Using Immobilized 4-1BBL and Interleukin-21. Int. J. Oncol. 2015, 47, 335–342. [Google Scholar] [CrossRef] [Green Version]
  44. Gras Navarro, A.; Kmiecik, J.; Leiss, L.; Zelkowski, M.; Engelsen, A.; Bruserud, Ø.; Zimmer, J.; Enger, P.Ø.; Chekenya, M. NK Cells with KIR2DS2 Immunogenotype Have a Functional Activation Advantage to Efficiently Kill Glioblastoma and Prolong Animal Survival. J. Immunol. 2014, 193, 6192–6206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Oyer, J.L.; Igarashi, R.Y.; Kulikowski, A.R.; Colosimo, D.A.; Solh, M.M.; Zakari, A.; Khaled, Y.A.; Altomare, D.A.; Copik, A.J. Generation of Highly Cytotoxic Natural Killer Cells for Treatment of Acute Myelogenous Leukemia Using a Feeder-Free, Particle-Based Approach. Biol. Blood Marrow Transplant. 2015, 21, 632–639. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Johnson, C.D.L.; Zale, N.E.; Frary, E.D.; Lomakin, J.A. Feeder-Cell-Free and Serum-Free Expansion of Natural Killer Cells Using Cloudz Microspheres, G-Rex6M, and Human Platelet Lysate. Front. Immunol. 2022, 13, 803380. [Google Scholar] [CrossRef] [PubMed]
  47. Huang, R.-S.; Lai, M.-C.; Shih, H.-A.; Lin, S. A Robust Platform for Expansion and Genome Editing of Primary Human Natural Killer Cells. J. Exp. Med. 2021, 218, e20201529. [Google Scholar] [CrossRef]
  48. Felices, M.; Lenvik, A.J.; McElmurry, R.; Chu, S.; Hinderlie, P.; Bendzick, L.; Geller, M.A.; Tolar, J.; Blazar, B.R.; Miller, J.S. Continuous Treatment with IL-15 Exhausts Human NK Cells via a Metabolic Defect. JCI. Insight. 2018, 3, e96219. [Google Scholar] [CrossRef]
  49. Suen, W.C.-W.; Lee, W.Y.-W.; Leung, K.-T.; Pan, X.-H.; Li, G. Natural Killer Cell-Based Cancer Immunotherapy: A Review on 10 Years Completed Clinical Trials. Cancer Investig. 2018, 36, 431–457. [Google Scholar] [CrossRef]
  50. Miller, J.S.; Soignier, Y.; Panoskaltsis-Mortari, A.; McNearney, S.A.; Yun, G.H.; Fautsch, S.K.; McKenna, D.; Le, C.; Defor, T.E.; Burns, L.J.; et al. Successful Adoptive Transfer and in Vivo Expansion of Human Haploidentical NK Cells in Patients with Cancer. Blood 2005, 105, 3051–3057. [Google Scholar] [CrossRef] [Green Version]
  51. 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]
  52. Lehmann, C.; Zeis, M.; Uharek, L. Activation of Natural Killer Cells with Interleukin 2 (IL-2) and IL-12 Increases Perforin Binding and Subsequent Lysis of Tumour Cells. Br. J. Haematol. 2001, 114, 660–665. [Google Scholar] [CrossRef]
  53. Bhat, R.; Watzl, C. Serial Killing of Tumor Cells by Human Natural Killer Cells—Enhancement by Therapeutic Antibodies. PLoS ONE 2007, 2, e326. [Google Scholar] [CrossRef]
  54. Sarkar, S.; Germeraad, W.T.V.; Rouschop, K.M.A.; Steeghs, E.M.P.; van Gelder, M.; Bos, G.M.J.; Wieten, L. Hypoxia Induced Impairment of NK Cell Cytotoxicity against Multiple Myeloma Can Be Overcome by IL-2 Activation of the NK Cells. PLoS ONE 2013, 8, e64835. [Google Scholar] [CrossRef]
  55. Poznanski, S.M.; Lee, A.J.; Nham, T.; Lusty, E.; Larché, M.J.; Lee, D.A.; Ashkar, A.A. Combined Stimulation with Interleukin-18 and Interleukin-12 Potently Induces Interleukin-8 Production by Natural Killer Cells. J. Innate Immun. 2017, 9, 511–525. [Google Scholar] [CrossRef] [PubMed]
  56. Urlaub, D.; Höfer, K.; Müller, M.-L.; Watzl, C. LFA-1 Activation in NK Cells and Their Subsets: Influence of Receptors, Maturation, and Cytokine Stimulation. J. Immunol. 2017, 198, 1944–1951. [Google Scholar] [CrossRef] [Green Version]
  57. Xie, G.; Dong, H.; Liang, Y.; Ham, J.D.; Rizwan, R.; Chen, J. CAR-NK Cells: A Promising Cellular Immunotherapy for Cancer. EBioMedicine 2020, 59, 102975. [Google Scholar] [CrossRef] [PubMed]
  58. Sadelain, M.; Brentjens, R.; Rivière, I. The Basic Principles of Chimeric Antigen Receptor Design. Cancer Discov. 2013, 3, 388–398. [Google Scholar] [CrossRef] [Green Version]
  59. Khawar, M.B.; Sun, H. CAR-NK Cells: From Natural Basis to Design for Kill. Front. Immunol. 2021, 12, 5157. [Google Scholar] [CrossRef] [PubMed]
  60. Liu, S.; Galat, V.; Galat4, Y.; Lee, Y.K.A.; Wainwright, D.; Wu, J. NK Cell-Based Cancer Immunotherapy: From Basic Biology to Clinical Development. J. Hematol. Oncol. 2021, 14, 7. [Google Scholar] [CrossRef]
  61. Albinger, N.; Hartmann, J.; Ullrich, E. Current Status and Perspective of CAR-T and CAR-NK Cell Therapy Trials in Germany. Gene Ther. 2021, 28, 513–527. [Google Scholar] [CrossRef]
  62. Liu, E.; Marin, D.; Banerjee, P.; Macapinlac, H.A.; Thompson, P.; Basar, R.; Nassif Kerbauy, L.; Overman, B.; Thall, P.; Kaplan, M.; et al. Use of CAR-Transduced Natural Killer Cells in CD19-Positive Lymphoid Tumors. N. Engl. J. Med. 2020, 382, 545–553. [Google Scholar] [CrossRef]
  63. Daher, M.; Melo Garcia, L.; Li, Y.; Rezvani, K. CAR-NK Cells: The next Wave of Cellular Therapy for Cancer. Clin. Transl. Immunology. 2021, 10, e1274. [Google Scholar] [CrossRef]
  64. Yilmaz, A.; Cui, H.; Caligiuri, M.A.; Yu, J. Chimeric Antigen Receptor-Engineered Natural Killer Cells for Cancer Immunotherapy. J. Hematol. Oncol. 2020, 13, 168. [Google Scholar] [CrossRef] [PubMed]
  65. Felices, M.; Lenvik, T.R.; Davis, Z.B.; Miller, J.S.; Vallera, D.A. Generation of BiKEs and TriKEs to Improve NK Cell-Mediated Targeting of Tumor Cells. Methods. Mol. Biol. 2016, 1441, 333–346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Vallera, D.A.; Felices, M.; McElmurry, R.; McCullar, V.; Zhou, X.; Schmohl, J.U.; Zhang, B.; Lenvik, A.J.; Panoskaltsis-Mortari, A.; Verneris, M.R.; et al. IL15 Trispecific Killer Engagers (TriKE) Make Natural Killer Cells Specific to CD33+ Targets While Also Inducing Persistence, in Vivo Expansion, and En-hanced Function. Clin. Cancer. Res. 2016, 22, 3440–3450. [Google Scholar] [CrossRef] [Green Version]
  67. Arvindam, U.S.; van Hauten, P.M.M.; Schirm, D.; Schaap, N.; Hobo, W.; Blazar, B.R.; Vallera, D.A.; Dolstra, H.; Felices, M.; Miller, J.S. A Trispecific Killer Engager Molecule against CLEC12A Effectively Induces NK-Cell Mediated Killing of AML Cells. Leukemia 2021, 35, 1586–1596. [Google Scholar] [CrossRef] [PubMed]
  68. GTB-3550 Tri-Specific Killer Engager (TriKE ®) for High Risk Hematologic Malignancies—Full Text View Clini-calTrials.gov. (n.d.). Available online: https://clinicaltrials.gov/ct2/show/NCT03214666 (accessed on 10 March 2023).
  69. Williams, S.M.; Sumstad, D.; Kadidlo, D.; Curtsinger, J.; Luo, X.; Miller, J.S.; McKenna Jr, D.H. Clinical-scale Production of CGMP Compliant CD3/CD19 Cell-depleted NK Cells in the Evolution of NK Cell Immunotherapy at a Single Institution. Transfusion 2018, 58, 1458–1467. [Google Scholar] [CrossRef]
  70. Passweg, J.R.; Tichelli, A.; Meyer-Monard, S.; Heim, D.; Stern, M.; Kühne, T.; Favre, G.; Gratwohl, A. Purified Donor NK-Lymphocyte Infusion to Consolidate Engraftment after Haploidentical Stem Cell Transplantation. Leukemia 2004, 18, 1835–1838. [Google Scholar] [CrossRef]
  71. Shankar, K.; Capitini, C.M.; Saha, K. Genome Engineering of Induced Pluripotent Stem Cells to Manufacture Natural Killer Cell Therapies. Stem Cell Res. Ther. 2020, 11, 234. [Google Scholar] [CrossRef]
  72. Yoon, S.R.; Lee, Y.S.; Yang, S.H.; Ahn, K.H.; Lee, J.-H.; Lee, J.-H.; Kim, D.Y.; Kang, Y.A.; Jeon, M.; Seol, M. Generation of Donor Natural Killer Cells from CD34+ Progenitor Cells and Subsequent Infusion after HLA-Mismatched Allogeneic Hematopoietic Cell Transplantation: A Feasibility Study. Bone Marrow Transpl. 2010, 45, 1038–1046. [Google Scholar] [CrossRef]
  73. Mark, C.; Czerwinski, T.; Roessner, S.; Mainka, A.; Hörsch, F.; Heublein, L.; Winterl, A.; Sanokowski, S.; Richter, S.; Bauer, N. Cryopreservation Impairs 3-D Migration and Cytotoxicity of Natural Killer Cells. Nat. Commun. 2020, 11, 5224. [Google Scholar] [CrossRef]
  74. Matosevic, S. Viral and Nonviral Engineering of Natural Killer Cells as Emerging Adoptive Cancer Immunotherapies. J. Immunol. Res. 2018, 2018, 4054815. [Google Scholar] [CrossRef]
  75. Somanchi, S.S.; Somanchi, A.; Cooper, L.J.N.; Lee, D.A. Engineering Lymph Node Homing of Ex Vivo–Expanded Human Natural Killer Cells via Trogocytosis of the Chemokine Receptor CCR7. Blood J. Am. Soc. Hematol. 2012, 119, 5164–5172. [Google Scholar] [CrossRef] [PubMed]
  76. Gauthier, L.; Morel, A.; Anceriz, N.; Rossi, B.; Blanchard-Alvarez, A.; Grondin, G.; Trichard, S.; Cesari, C.; Sapet, M.; Bosco, F.; et al. Multifunctional Natural Killer Cell Engagers Targeting NKp46 Trigger Protective Tumor Immunity. Cell 2019, 177, 1701–1713.e16. [Google Scholar] [CrossRef] [PubMed]
  77. Colomar-Carando, N.; Gauthier, L.; Merli, P.; Loiacono, F.; Canevali, P.; Falco, M.; Galaverna, F.; Rossi, B.; Bosco, F.; Caratini, M.; et al. Exploiting Natural Killer Cell Engagers to Control Pediatric B-Cell Precursor Acute Lymphoblastic Leukemia. Cancer Immunol. Res. 2022, 10, 291–302. [Google Scholar] [CrossRef] [PubMed]
  78. Gauthier, L.; Virone-Oddos, A.; Beninga, J.; Rossi, B.; Nicolazzi, C.; Amara, C.; Blanchard-Alvarez, A.; Gourdin, N.; Courta, J.; Basset, A.; et al. Control of Acute Myeloid Leukemia by a Trifunctional NKp46-CD16a-NK Cell Engager Targeting CD123. Nat. Biotechnol. 2023. [Google Scholar] [CrossRef]
  79. Demaria, O.; Gauthier, L.; Vetizou, M.; Blanchard Alvarez, A.; Vagne, C.; Habif, G.; Batista, L.; Baron, W.; Belaïd, N.; Girard-Madoux, M.; et al. Antitumor Immunity Induced by Antibody-Based Natural Killer Cell Engager Therapeutics Armed with Not-Alpha IL-2 Variant. Cell Rep. Med. 2022, 3, 100783. [Google Scholar] [CrossRef]
  80. Chretien, A.-S.; Devillier, R.; Fauriat, C.; Orlanducci, F.; Harbi, S.; Le Roy, A.; Rey, J.; Bouvier Borg, G.; Gautherot, E.; Hamel, J.-F. NKp46 Expression on NK Cells as a Prognostic and Predictive Biomarker for Response to Allo-SCT in Patients with AML. Oncoimmunology 2017, 6, e1307491. [Google Scholar] [CrossRef] [Green Version]
  81. 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. Cytokine-Induced Memory-like Natural Killer Cells Exhibit Enhanced Responses against Myeloid Leukemia. Sci. Transl. Med. 2016, 8, ra123–ra357. [Google Scholar] [CrossRef] [Green Version]
  82. Guillerey, C.; Huntington, N.D.; Smyth, M.J. Targeting Natural Killer Cells in Cancer Immunotherapy. Nat. Immunol. 2016, 17, 1025–1036. [Google Scholar] [CrossRef]
  83. Mamessier, E.; Sylvain, A.; Thibult, M.L.; Houvenaeghel, G.; Jacquemier, J.; Castellano, R.; Gonçalves, A.; André, P.; Romagné, F.; Thibault, G.; et al. Human Breast Cancer Cells Enhance Self Tolerance by Promoting Evasion from NK Cell Antitumor Immunity. J. Clin. Investig. 2011, 121, 3609–3622. [Google Scholar] [CrossRef] [Green Version]
  84. O’Sullivan, T.; Saddawi-Konefka, R.; Gross, E.; Tran, M.; Mayfield, S.P.; Ikeda, H.; Bui, J.D. Interleukin-17D Mediates Tumor Rejection through Recruitment of Natural Killer Cells. Cell Rep. 2014, 7, 989–998. [Google Scholar] [CrossRef] [Green Version]
  85. Platonova, S.; Cherfils-Vicini, J.; Damotte, D.; Crozet, L.; Vieillard, V.; Validire, P.; André, P.; Dieu-Nosjean, M.-C.; Alifano, M.; Régnard, J.-F. Profound Coordinated Alterations of Intratumoral NK Cell Phenotype and Function in Lung Carcinoma. Cancer Res. 2011, 71, 5412–5422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Sun, C.; Xu, J.; Huang, Q.; Huang, M.; Wen, H.; Zhang, C.; Wang, J.; Song, J.; Zheng, M.; Sun, H. High NKG2A Expression Contributes to NK Cell Exhaustion and Predicts a Poor Prognosis of Patients with Liver Cancer. Oncoimmunology 2017, 6, e1264562. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Peng, Y.; Luo, G.; Zhou, J.; Wang, X.; Hu, J.; Cui, Y.; Li, X.C.; Tan, J.; Yang, S.; Zhan, R. CD86 Is an Activation Receptor for NK Cell Cytotoxicity against Tumor Cells. PLoS ONE 2013, 8, e83913. [Google Scholar] [CrossRef]
  88. Sun, C.; Fu, B.; Gao, Y.; Liao, X.; Sun, R.; Tian, Z.; Wei, H. TGF-Β1 down-Regulation of NKG2D/DAP10 and 2B4/SAP Expression on Human NK Cells Contributes to HBV Persistence. PLoS Pathog. 2012, 8, e1002594. [Google Scholar] [CrossRef]
  89. Bi, J.; Tian, Z. NK Cell Exhaustion. Front. Immunol. 2017, 8, 760. [Google Scholar] [CrossRef] [Green Version]
  90. Wherry, E.J. T Cell Exhaustion. Nat. Immunol. 2011, 12, 492–499. [Google Scholar] [CrossRef] [PubMed]
  91. Brodin, P.; Lakshmikanth, T.; Johansson, S.; Kärre, K.; Höglund, P. The Strength of Inhibitory Input during Education Quantitatively Tunes the Functional Responsiveness of Individual Natural Killer Cells. Blood 2009, 113, 2434–2441. [Google Scholar] [CrossRef] [Green Version]
  92. Fernandez, N.C.; Treiner, E.; Vance, R.E.; Jamieson, A.M.; Lemieux, S.; Raulet, D.H. A Subset of Natural Killer Cells Achieves Self-Tolerance without Expressing Inhibitory Receptors Specific for Self-MHC Molecules. Blood 2005, 105, 4416–4423. [Google Scholar] [CrossRef] [Green Version]
  93. Kim, S.; Poursine-Laurent, J.; Truscott, S.M.; Lybarger, L.; Song, Y.J.; Yang, L.; French, A.R.; Sunwoo, J.B.; Lemieux, S.; Hansen, T.H.; et al. Licensing of Natural Killer Cells by Host Major Histocompatibility Complex Class I Molecules. Nature 2005, 436, 709–713. [Google Scholar] [CrossRef]
  94. Judge, S.J.; Murphy, W.J.; Canter, R.J. Characterizing the Dysfunctional NK Cell: Assessing the Clinical Relevance of Exhaustion, Anergy, and Senescence. Front. Cell. Infect. Microbiol. 2020, 10, 49. [Google Scholar] [CrossRef] [Green Version]
  95. Sojka, D.K.; Plougastel-Douglas, B.; Yang, L.; Pak-Wittel, M.A.; Artyomov, M.N.; Ivanova, Y.; Zhong, C.; Chase, J.M.; Rothman, P.B.; Yu, J.; et al. Tissue-Resident Natural Killer (NK) Cells Are Cell Lineages Distinct from Thymic and Conventional Splenic NK Cells. eLife 2014, 2014, e01659. [Google Scholar] [CrossRef] [PubMed]
  96. Heinrich, B.; Gertz, E.M.; Schäffer, A.A.; Craig, A.; Ruf, B.; Subramanyam, V.; McVey, J.C.; Diggs, L.P.; Heinrich, S.; Rosato, U. The Tumour Microenvironment Shapes Innate Lymphoid Cells in Patients with Hepatocellular Carcinoma. Gut 2022, 71, 1161–1175. [Google Scholar] [CrossRef] [PubMed]
  97. Verrier, T.; Satoh-Takayama, N.; Serafini, N.; Marie, S.; di Santo, J.P.; Vosshenrich, C.A.J. Phenotypic and Functional Plasticity of Murine Intestinal NKp46+ Group 3 Innate Lymphoid Cells. J. Immunol. 2016, 196, 4731–4738. [Google Scholar] [CrossRef] [Green Version]
  98. Cella, M.; Gamini, R.; Sécca, C.; Collins, P.L.; Zhao, S.; Peng, V.; Robinette, M.L.; Schettini, J.; Zaitsev, K.; Gordon, W. Subsets of ILC3− ILC1-like Cells Generate a Diversity Spectrum of Innate Lymphoid Cells in Human Mucosal Tissues. Nat. Immunol. 2019, 20, 980–991. [Google Scholar] [CrossRef] [PubMed]
  99. Bernink, J.H.; Krabbendam, L.; Germar, K.; de Jong, E.; Gronke, K.; Kofoed-Nielsen, M.; Munneke, J.M.; Hazenberg, M.D.; Villaudy, J.; Buskens, C.J. Interleukin-12 and-23 Control Plasticity of CD127+ Group 1 and Group 3 Innate Lymphoid Cells in the Intestinal Lamina Propria. Immunity 2015, 43, 146–160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Klose, C.S.N.; Flach, M.; Möhle, L.; Rogell, L.; Hoyler, T.; Ebert, K.; Fabiunke, C.; Pfeifer, D.; Sexl, V.; Fonseca-Pereira, D.; et al. Differentiation of Type 1 ILCs from a Common Progenitor to All Helper-like Innate Lymphoid Cell Lineages. Cell 2014, 157, 340–356. [Google Scholar] [CrossRef] [Green Version]
  101. Daussy, C.; Faure, F.; Mayol, K.; Viel, S.; Gasteiger, G.; Charrier, E.; Bienvenu, J.; Henry, T.; Debien, E.; Hasan, U.A.; et al. T-Bet and Eomes Instruct the Development of Two Distinct Natural Killer Cell Lineages in the Liver and in the Bone Marrow. J. Exp. Med. 2014, 211, 563–577. [Google Scholar] [CrossRef]
  102. Gao, F.; Chambon, P.; Tellides, G.; Kong, W.; Zhang, X.; Li, W. Disruption of TGF-β Signaling in Smooth Muscle Cell Prevents Flow-Induced Vascular Remodeling. Biochem. Biophys. Res. Commun. 2014, 454, 245–250. [Google Scholar] [CrossRef]
  103. Cortez, V.S.; Ulland, T.K.; Cervantes-Barragan, L.; Bando, J.K.; Robinette, M.L.; Wang, Q.; White, A.J.; Gilfillan, S.; Cella, M.; Colonna, M. SMAD4 Impedes the Conversion of NK Cells into ILC1-like Cells by Curtailing Non-Canonical TGF-β Signaling. Nat. Immunol. 2017, 18, 995–1003. [Google Scholar] [CrossRef] [Green Version]
  104. Hawke, L.G.; Mitchell, B.Z.; Ormiston, M.L. TGF-β and IL-15 Synergize through MAPK Pathways to Drive the Conversion of Human NK Cells to an Innate Lymphoid Cell 1–like Phenotype. J. Immunol. 2020, 204, 3171–3181. [Google Scholar] [CrossRef]
  105. Lee, J.-C.; Lee, K.-M.; Kim, D.-W.; Heo, D.S. Elevated TGF-Β1 Secretion and Down-Modulation of NKG2D Underlies Impaired NK Cytotoxicity in Cancer Patients. J. Immunol. 2004, 172, 7335–7340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Crane, C.A.; Han, S.J.; Barry, J.J.; Ahn, B.J.; Lanier, L.L.; Parsa, A.T. TGF-β Downregulates the Activating Receptor NKG2D on NK Cells and CD8+ T Cells in Glioma Patients. Neuro Oncol. 2010, 12, 7–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. 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-1 1. J. Immunol. 2009, 182, 240–249. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Jun, E.; Song, A.Y.; Choi, J.W.; Lee, H.H.; Kim, M.Y.; Ko, D.H.; Kang, H.J.; Kim, S.W.; Bryceson, Y.; Kim, S.C.; et al. Progressive Impairment of NK Cell Cytotoxic Degranulation Is Associated with TGF-Β1 Deregulation and Disease Progression in Pancreatic Cancer. Front. Immunol. 2019, 10, 1354. [Google Scholar] [CrossRef] [PubMed]
  109. Castriconi, R.; Cantoni, C.; Chiesa, M.D.; Vitale, M.; Marcenaro, E.; Conte, R.; Biassoni, R.; Bottino, C.; Moretta, L.; Moretta, A. Transforming Growth Factor 1 Inhibits Expression of NKp30 and NKG2D Receptors: Consequences for the NK-Mediated Killing of Dendritic Cells. Natl. Inst. Health 2003, 100, 4120–4125. [Google Scholar] [CrossRef] [Green Version]
  110. Viel, S.; Marçais, A.; Souza-Fonseca Guimaraes, F.; Loftus, R.; Rabilloud, J.; Grau, M.; Degouve, S.; Djebali, S.; Sanlaville, A.; Charrier, E.; et al. TGF-b Inhibits the Activation and Functions of NK Cells by Repressing the MTOR Pathway. Sci. Signal. 2016, 9, ra19. [Google Scholar] [CrossRef]
  111. Xu, L.; Ma, Q.; Zhu, J.; Li, J.; Xue, B.; Gao, J.; Sun, C.; Zang, Y.; Zhou, Y.; Yang, D. Combined Inhibition of JAK1, 2/Stat3-PD-L1 Signaling Pathway Suppresses the Immune Escape of Castration-resistant Prostate Cancer to NK Cells in Hypoxia. Mol. Med. Rep. 2018, 17, 8111–8120. [Google Scholar]
  112. Ashkar, A.A.; Di Santo, J.P.; Croy, B.A. Interferon Contributes to Initiation of Uterine Vascular Modification, Decidual Integrity, and Uterine Natural Killer Cell Maturation during Normal Murine Pregnancy. J. Exp. Med. 2000, 192, 259–269. [Google Scholar] [CrossRef] [Green Version]
  113. Montaldo, E.; Vacca, P.; Chiossone, L.; Croxatto, D.; Loiacono, F.; Martini, S.; Ferrero, S.; Walzer, T.; Moretta, L.; Mingari, M.C. Unique Eomes+ NK Cell Subsets Are Present in Uterus and Decidua during Early Pregnancy. Front. Immunol. 2016, 6, 646. [Google Scholar] [CrossRef] [Green Version]
  114. Parr, E.L.; Parr, M.B.; Zheng, L.M.; Young, J.D.E. Mouse Granulated Metrial Gland Cells Originate by Local Activation of Uterine Natural Killer Lymphocytes. Biol. Reprod. 1991, 44, 834–841. [Google Scholar] [CrossRef]
  115. Yadi, H.; Burke, S.; Madeja, Z.; Hemberger, M.; Moffett, A.; Colucci, F. Unique Receptor Repertoire in Mouse Uterine NK Cells. J. Immunol. 2008, 181, 6140–6147. [Google Scholar] [CrossRef] [Green Version]
  116. Mallidi, T.V.; Craig, L.E.; Schloemann, S.R.; Riley, J.K. Murine Endometrial and Decidual NK1.1+ Natural Killer Cells Display a B220+CD11c+ Cell Surface Phenotype. Biol. Reprod. 2009, 81, 310–318. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Moffett, A.; Loke, C. Immunology of Placentation in Eutherian Mammals. Nat. Rev. Immunol. 2006, 6, 584–594. [Google Scholar] [CrossRef] [PubMed]
  118. Hatta, K.; Macleod, R.J.; Gerber, S.A.; Croy, B.A. Emerging Themes in Uterine Natural Killer Cell Heterogeneity and Function. Am. J. Reprod. Immunol. 2012, 68, 282–289. [Google Scholar] [CrossRef] [PubMed]
  119. Fraser, R.; Whitley, G.S.J.; Thilaganathan, B.; Cartwright, J.E. Decidual Natural Killer Cells Regulate Vessel Stability: Implications for Impaired Spiral Artery Remodelling. J. Reprod. Immunol. 2015, 110, 54–60. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Vacca, P.; Cantoni, C.; Vitale, M.; Prato, C.; Canegallo, F.; Fenoglio, D.; Ragni, N.; Moretta, L.; Mingari, M.C. Crosstalk between Decidual NK and CD14+ Myelomonocytic Cells Results in Induction of Tregs and Immunosuppression. Proc. Natl. Acad. Sci. USA 2010, 107, 11918–11923. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. 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]
  122. Keskin, D.B.; Allan, D.S.J.; Rybalov, B.; Andzelm, M.M.; Stern, J.N.H.; Kopcow, H.D.; Koopman, L.A.; Strominger, J.L. TGF Promotes Conversion of CD16 Peripheral Blood NK Cells into CD16 NK Cells with Similarities to Decidual NK Cells. Proc. Natl. Acad. Sci. USA 2007, 104, 3378–3383. [Google Scholar] [CrossRef] [Green Version]
  123. Cerdeira, A.S.; Rajakumar, A.; Royle, C.M.; Lo, A.; Husain, Z.; Thadhani, R.I.; Sukhatme, V.P.; Karumanchi, S.A.; Kopcow, H.D. Conversion of Peripheral Blood NK Cells to a Decidual NK-like Phenotype by a Cocktail of Defined Factors. J. Immunol. 2013, 190, 3939–3948. [Google Scholar] [CrossRef] [Green Version]
  124. Hawke, L.G.; Whitford, M.K.M.; Ormiston, M.L. The Production of Pro-Angiogenic VEGF-A Isoforms by Hypoxic Human NK Cells Is Independent of Their TGF-β-Mediated Conversion to an ILC1-Like Phenotype. Front. Immunol. 2020, 11, 1903. [Google Scholar] [CrossRef]
  125. Bruno, A.; Focaccetti, C.; Pagani, A.; Imperatori, A.S.; Spagnoletti, M.; Rotolo, N.; Cantelmo, A.R.; Franzi, F.; Capella, C.; Ferlazzo, G.; et al. The Proangiogenic Phenotype of Natural Killer Cells in Patients with Non-Small Cell Lung Cancer. Neoplasia 2013, 15, 133–142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Gotthardt, D.; Trifinopoulos, J.; Sexl, V.; Putz, E.M. JAK/STAT Cytokine Signaling at the Crossroad of NK Cell Development and Maturation. Front. Immunol. 2019, 10, 2590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Tanaka, H.; Shinto, O.; Yashiro, M.; Yamazoe, S.; Iwauchi, T.; Muguruma, K.; Kubo, N.; Ohira, M.; Hirakawa, K. Transforming Growth Factor β Signaling Inhibitor, SB-431542, Induces Maturation of Dendritic Cells and Enhances Anti-Tumor Activity. Oncol. Rep. 2010, 24, 1637–1643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Takeuchi, K.; Abe, M.; Hiasa, M.; Oda, A.; Amou, H.; Kido, S.; Harada, T.; Tanaka, O.; Miki, H.; Nakamura, S. TGF-β Inhibition Restores Terminal Osteoblast Differentiation to Suppress Myeloma Growth. PLoS ONE 2010, 5, e9870. [Google Scholar] [CrossRef]
  129. Lu, Y.; Jiang, F.; Zheng, X.; Katakowski, M.; Buller, B.; To, S.-S.T.; Chopp, M. TGF-Β1 Promotes Motility and Invasiveness of Glioma Cells through Activation of ADAM17. Oncol. Rep. 2011, 25, 1329–1335. [Google Scholar] [PubMed] [Green Version]
  130. Biswas, S.; Guix, M.; Rinehart, C.; Dugger, T.C.; Chytil, A.; Moses, H.L.; Freeman, M.L.; Arteaga, C.L. Inhibition of TGF-β with Neutralizing Antibodies Prevents Radiation-Induced Acceleration of Metastatic Cancer Progression. J. Clin. Investig. 2007, 117, 1305–1313. [Google Scholar] [CrossRef] [Green Version]
  131. Chen, X.; Yang, Y.; Zhou, Q.; Weiss, J.M.; Howard, O.Z.; McPherson, J.M.; Wakefield, L.M.; Oppenheim, J.J. Effective Chemoimmunotherapy with Anti-TGFβ Antibody and Cyclophosphamide in a Mouse Model of Breast Cancer. PLoS ONE 2014, 9, e85398. [Google Scholar] [CrossRef]
  132. Nam, J.-S.; Terabe, M.; Mamura, M.; Kang, M.-J.; Chae, H.; Stuelten, C.; Kohn, E.; Tang, B.; Sabzevari, H.; Anver, M.R. An Anti–Transforming Growth Factor β Antibody Suppresses Metastasis via Cooperative Effects on Multiple Cell Compartments. Cancer Res. 2008, 68, 3835–3843. [Google Scholar] [CrossRef] [Green Version]
  133. Otegbeye, F.; Ojo, E.; Moreton, S.; Mackowski, N.; Lee, D.A.; De Lima, M.; Wald, D.N. Inhibiting TGF-Beta Signaling Preserves the Function of Highly Activated, in Vitro Expanded Natural Killer Cells in AML and Colon Cancer Models. PLoS ONE 2018, 13, e0191358. [Google Scholar] [CrossRef] [Green Version]
  134. Anderton, M.J.; Mellor, H.R.; Bell, A.; Sadler, C.; Pass, M.; Powell, S.; Steele, S.J.; Roberts, R.R.A.; Heier, A. Induction of Heart Valve Lesions by Small-Molecule ALK5 Inhibitors. Toxicol. Pathol. 2011, 39, 916–924. [Google Scholar] [CrossRef] [Green Version]
  135. Li, M.O.; Wan, Y.Y.; Sanjabi, S.; Robertson, A.K.L.; Flavell, R.A. Transforming Growth Factor-β Regulation of Immune Responses. Annu. Rev. Immunol. 2006, 24, 99–146. [Google Scholar] [CrossRef] [PubMed]
  136. Burga, R.A.; Yvon, E.; Chorvinsky, E.; Fernandes, R.; Cruz, C.R.; Bollard, C.M. Engineering the TGFb Receptor to Enhance the Therapeutic Potential of Natural Killer Cells as an Immunotherapy for Neuroblastoma. Clin. Cancer Res. 2019, 25, 4400–4412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Wang, Z.; Guo, L.; Song, Y.; Zhang, Y.; Lin, D.; Hu, B.; Mei, Y.; Sandikin, D.; Liu, H. Augmented Anti-Tumor Activity of NK-92 Cells Expressing Chimeric Receptors of TGF-ΒR II and NKG2D. Cancer Immunol. Immunother. 2017, 66, 537–548. [Google Scholar] [CrossRef] [PubMed]
  138. Yang, B.; Liu, H.; Shi, W.; Wang, Z.; Sun, S.; Zhang, G.; Hu, Y.; Liu, T.; Jiao, S. Blocking Transforming Growth Factor-β Signaling Pathway Augments Antitumor Effect of Adoptive NK-92 Cell Therapy. Int. Immunopharmacol. 2013, 17, 198–204. [Google Scholar] [CrossRef]
  139. Lim, S.A.; Kim, J.; Jeon, S.; Shin, M.H.; Kwon, J.; Kim, T.-J.; Im, K.; Han, Y.; Kwon, W.; Kim, S.-W.; et al. Defective Localization with Impaired Tumor Cytotoxicity Contributes to the Immune Escape of NK Cells in Pancreatic Cancer Patients. Front. Immunol. 2019, 10, 496. [Google Scholar] [CrossRef] [Green Version]
  140. Klokker, M.; Kharazmi, A.; Galbo, H.; Bygbjerg, I.; Pedersen, B.K. Influence of in Vivo Hypobaric Hypoxia on Function of Lymphocytes, Neutrocytes, Natural Killer Cells, and Cytokines. J. Appl. Physiol. 1993, 74, 1100–1106. [Google Scholar] [CrossRef]
  141. Loeffler, D.A.; Heppner, G.H.; Juneau, P.L. Natural Killer-cell Activity under Conditions Reflective of Tumor Micro-environment. Int. J. Cancer 1991, 48, 895–899. [Google Scholar] [CrossRef]
  142. Balsamo, M.; Manzini, C.; Pietra, G.; Raggi, F.; Blengio, F.; Mingari, M.C.; Varesio, L.; Moretta, L.; Bosco, M.C.; Vitale, M. Hypoxia Downregulates the Expression of Activating Receptors Involved in NK-Cell-Mediated Target Cell Killing without Affecting ADCC. Eur. J. Immunol. 2013, 43, 2756–2764. [Google Scholar] [CrossRef]
  143. Shohet, R.V.; Garcia, J.A. Keeping the Engine Primed: HIF Factors as Key Regulators of Cardiac Metabolism and Angiogenesis during Ischemia. J. Mol. Med. 2007, 85, 1309–1315. [Google Scholar] [CrossRef] [Green Version]
  144. Halligan, D.N.; Murphy, S.J.E.; Taylor, C.T. The Hypoxia-Inducible Factor (HIF) Couples Immunity with Metabolism. In Seminars in Immunology; Elsevier: Amsterdam, The Netherlands, 2016; Volume 28, pp. 469–477. [Google Scholar]
  145. Parodi, M.; Raggi, F.; Cangelosi, D.; Manzini, C.; Balsamo, M.; Blengio, F.; Eva, A.; Varesio, L.; Pietra, G.; Moretta, L.; et al. Hypoxia Modifies the Transcriptome of Human NK Cells, Modulates Their Immunoregulatory Profile, and Influences NK Cell Subset Migration. Front. Immunol. 2018, 9, 2358. [Google Scholar] [CrossRef] [Green Version]
  146. Velásquez, S.Y.; Killian, D.; Schulte, J.; Sticht, C.; Thiel, M.; Lindner, H.A. Short Term Hypoxia Synergizes with Interleukin 15 Priming in Driving Glycolytic Gene Transcription and Supports Human Natural Killer Cell Activities. J. Biol. Chem. 2016, 291, 12960–12977. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Lu, Y.; Hu, J.; Sun, W.; Duan, X.; Chen, X. Hypoxia-Mediated Immune Evasion of Pancreatic Carcinoma Cells. Mol. Med. Rep. 2015, 11, 3666–3672. [Google Scholar] [CrossRef] [Green Version]
  148. Ou, Z.L.; Luo, Z.; Wei, W.; Liang, S.; Gao, T.L.; Lu, Y. Bin Hypoxia-Induced Shedding of MICA and HIF1A-Mediated Immune Escape of Pancreatic Cancer Cells from NK Cells: Role of Circ_0000977/MiR-153 Axis. RNA Biol. 2019, 16, 1592–1603. [Google Scholar] [CrossRef] [PubMed]
  149. Yamada, N.; Yamanegi, K.; Ohyama, H.; Hata, M.; Nakasho, K.; Futani, H.; Okamura, H.; Terada, N. Hypoxia Downregulates the Expression of Cell Surface MICA without Increasing Soluble MICA in Osteosarcoma Cells in a HIF-1α-Dependent Manner. Int. J. Oncol. 2012, 41, 2005–2012. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Barsoum, I.B.; Hamilton, T.K.; Li, X.; Cotechini, T.; Miles, E.A.; Siemens, D.R.; Graham, C.H. Hypoxia Induces Escape from Innate Immunity in Cancer Cells via Increased Expression of ADAM10: Role of Nitric Oxide. Cancer Res. 2011, 71, 7433–7441. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Gotthardt, D.; Putz, E.M.; Grundschober, E.; Prchal-Murphy, M.; Straka, E.; Kudweis, P.; Heller, G.; Bago-Horvath, Z.; Witalisz-Siepracka, A.; Cumaraswamy, A.A.; et al. STAT5 Is a Key Regulator in NK Cells and Acts as a Molecular Switch from Tumor Surveillance to Tumor Promotion. Cancer Discov. 2016, 6, 414–429. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Krzywinska, E.; Kantari-Mimoun, C.; Kerdiles, Y.; Sobecki, M.; Isagawa, T.; Gotthardt, D.; Castells, M.; Haubold, J.; Millien, C.; Viel, T.; et al. Loss of HIF-1α in Natural Killer Cells Inhibits Tumour Growth by Stimulating Non-Productive Angiogenesis. Nat. Commun. 2017, 8, 1597. [Google Scholar] [CrossRef] [Green Version]
  153. Ni, J.; Wang, X.; Stojanovic, A.; Zhang, Q.; Wincher, M.; Bühler, L.; Arnold, A.; Correia, M.P.; Winkler, M.; Koch, P.S.; et al. Single-Cell RNA Sequencing of Tumor-Infiltrating NK Cells Reveals That Inhibition of Transcription Factor HIF-1α Unleashes NK Cell Activity. Immunity 2020, 52, 1075–1087.e8. [Google Scholar] [CrossRef]
  154. Sun, X.; Kanwar, J.R.; Leung, E.; Lehnert, K.; Wang, D.; Krissansen, G.W. Gene Transfer of Antisense Hypoxia Inducible Factor-1 Enhances the Therapeutic Efficacy of Cancer Immunotherapy. Gene Ther. 2001, 8, 638–645. [Google Scholar] [CrossRef] [Green Version]
  155. Solocinski, K.; Padget, M.R.; Fabian, K.P.; Wolfson, B.; Cecchi, F.; Hembrough, T.; Benz, S.C.; Rabizadeh, S.; Soon-Shiong, P.; Schlom, J.; et al. Overcoming Hypoxia-Induced Functional Suppression of NK Cells. J. Immunother. Cancer 2020, 8, e000246. [Google Scholar] [CrossRef]
  156. Frumento, G.; Rotondo, R.; Tonetti, M.; Damonte, G.; Benatti, U.; Ferrara, G.B. Tryptophan-Derived Catabolites Are Responsible for Inhibition of T and Natural Killer Cell Proliferation Induced by Indoleamine 2,3-Dioxygenase. J. Exp. Med. 2002, 196, 459–468. [Google Scholar] [CrossRef] [Green Version]
  157. Schroecksnadel, K.; Kaser, S.; Ledochowski, M.; Neurauter, G.; Mur, E.; Herold, M.; Fuchs, D. Increased Degradation of Tryptophan in Blood of Patients with Rheumatoid Arthritis. J. Rheumatol. 2003, 30, 1935–1939. [Google Scholar]
  158. Brown, R.R.; Ozaki, Y.; Datta, S.P.; Borden, E.C.; Sondel, P.M.; Malone, D.G. Implications of Interferon-Induced Tryptophan Catabolism in Cancer, Autoimmune Diseases and Aids BT—Kynurenine and Serotonin Pathways: Progress in Tryptophan Research; Schwarcz, R., Young, S.N., Brown, R.R., Eds.; Springer: New York, NY, USA; Boston, MA, USA, 1991; pp. 425–435. ISBN 978-1-4684-5952-4. [Google Scholar]
  159. Yufit, T.; Vining, V.; Brown, R.R.; Varga, J.; Wang, L. Inhibition of Type I Collagen MRNA Expression Independent of Tryptophan Depletion in Interferon-γ-Treated Human Dermal Fibroblasts. J. Investig. Dermatol. 1995, 105, 388–393. [Google Scholar] [CrossRef] [Green Version]
  160. Mellor, A.L.; Munn, D.H. Ido Expression by Dendritic Cells: Tolerance and Tryptophan Catabolism. Nat. Rev. Immunol. 2004, 4, 762–774. [Google Scholar] [CrossRef] [PubMed]
  161. Uyttenhove, C.; Pilotte, L.; Théate, I.; Stroobant, V.; Colau, D.; Parmentier, N.; Boon, T.; Van den Eynde, B.J. Evidence for a Tumoral Immune Resistance Mechanism Based on Tryptophan Degradation by Indoleamine 2,3-Dioxygenase. Nat. Med. 2003, 9, 1269–1274. [Google Scholar] [CrossRef] [PubMed]
  162. Chiesa, M.D.; 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] [PubMed]
  163. Terness, P.; Bauer, T.M.; Röse, L.; Dufter, C.; Watzlik, A.; Simon, H.; Opelz, G. Inhibition of Allogeneic T Cell Proliferation by Indoleamine 2,3-Dioxygenase–Expressing Dendritic Cells: Mediation of Suppression by Tryptophan Metabolites. J. Exp. Med. 2002, 196, 447–457. [Google Scholar] [CrossRef] [Green Version]
  164. Song, H.; Park, H.; Kim, Y.-S.; Kim, K.D.; Lee, H.-K.; Cho, D.-H.; Yang, J.-W.; Hur, D.Y. L-Kynurenine-Induced Apoptosis in Human NK Cells Is Mediated by Reactive Oxygen Species. Int. Immunopharmacol. 2011, 11, 932–938. [Google Scholar] [CrossRef] [PubMed]
  165. Wang, D.; Saga, Y.; Mizukami, H.; Sato, N.; Nonaka, H.; Fujiwara, H.; Takei, Y.; Machida, S.; Takikawa, O.; Ozawa, K.; et al. Indoleamine-2,3-Dioxygenase, an Immunosuppressive Enzyme That Inhibits Natural Killer Cell Function, as a Useful Target for Ovarian Cancer Therapy. Int. J. Oncol. 2012, 40, 929–934. [Google Scholar] [CrossRef] [Green Version]
  166. Zhang, J.; Han, X.; Hu, X.; Jin, F.; Gao, Z.; Yin, L.; Qin, J.; Yin, F.; Li, C.; Wang, Y. IDO1 Impairs NK Cell Cytotoxicity by Decreasing NKG2D/NKG2DLs via Promoting MiR-18a. Mol. Immunol. 2018, 103, 144–155. [Google Scholar] [CrossRef]
  167. Komiya, T.; Huang, C.H. Updates in the Clinical Development of Epacadostat and Other Indoleamine 2,3-Dioxygenase 1 Inhibitors (IDO1) for Human Cancers. Front. Oncol. 2018, 8, 423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Muller, A.J.; DuHadaway, J.B.; Donover, P.S.; Sutanto-Ward, E.; Prendergast, G.C. Inhibition of Indoleamine 2,3-Dioxygenase, an Immunoregulatory Target of the Cancer Suppression Gene Bin1, Potentiates Cancer Chemotherapy. Nat. Med. 2005, 11, 312–319. [Google Scholar] [CrossRef] [PubMed]
  169. Yang, H.J.; Yen, M.C.; Lin, C.C.; Lin, C.M.; Chen, Y.L.; Weng, T.Y.; Huang, T.T.; Wu, C.L.; Lai, M.D. A Combination of the Metabolic Enzyme Inhibitor APO866 and the Immune Adjuvant L-1-Methyl Tryptophan Induces Additive Antitumor Activity. Exp. Biol. Med. 2010, 235, 869–876. [Google Scholar] [CrossRef]
  170. Günther, J.; Däbritz, J.; Wirthgen, E. Limitations and Off-Target Effects of Tryptophan-Related IDO Inhibitors in Cancer Treatment. Front. Immunol. 2019, 10, 1801. [Google Scholar] [CrossRef] [PubMed]
  171. Hou, D.-Y.; Muller, A.J.; Sharma, M.D.; DuHadaway, J.; Banerjee, T.; Johnson, M.; Mellor, A.L.; Prendergast, G.C.; Munn, D.H. Inhibition of Indoleamine 2, 3-Dioxygenase in Dendritic Cells by Stereoisomers of 1-Methyl-Tryptophan Correlates with Antitumor Responses. Cancer Res. 2007, 67, 792–801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Qian, F.; Villella, J.; Wallace, P.K.; Mhawech-Fauceglia, P.; Tario, J.D.; Andrews, C.; Matsuzaki, J.; Valmori, D.; Ayyoub, M.; Frederick, P.J.; et al. Efficacy of Levo-1-Methyl Tryptophan and Dextro-1-Methyl Tryptophan in Reversing Indoleamine-2,3-Dioxygenase-Mediated Arrest of T-Cell Proliferation in Human Epithelial Ovarian Cancer. Cancer Res. 2009, 69, 5498–5504. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Perera Molligoda Arachchige, A.S. NK Cell-Based Therapies for HIV Infection: Investigating Current Advances and Future Possibilities. J. Leukoc. Biol. 2022, 111, 921–931. [Google Scholar] [CrossRef]
  174. Zhen, A.; Kamata, M.; Rezek, V.; Rick, J.; Levin, B.; Kasparian, S.; Chen, I.S.Y.; Yang, O.O.; Zack, J.A.; Kitchen, S.G. HIV-Specific Immunity Derived from Chimeric Antigen Receptor-Engineered Stem Cells. Mol. Ther. 2015, 23, 1358–1367. [Google Scholar] [CrossRef] [Green Version]
  175. Yang, H.L.; Zhou, W.J.; Chang, K.K.; Mei, J.; Huang, L.Q.; Wang, M.Y.; Meng, Y.; Ha, S.Y.; Li, D.J.; Li, M.Q. The Crosstalk between Endometrial Stromal Cells and Macrophages Impairs Cytotoxicity of NK Cells in Endometriosis by Secreting IL-10 and TGF-β. Reproduction 2017, 154, 815–825. [Google Scholar] [CrossRef] [Green Version]
  176. Liu, X.T.; Sun, H.T.; Zhang, Z.F.; Shi, R.X.; Liu, L.B.; Yu, J.J.; Zhou, W.J.; Gu, C.J.; Yang, S.L.; Liu, Y.K.; et al. Indoleamine 2,3-Dioxygenase Suppresses the Cytotoxicity of NK Cells in Response to Ectopic Endometrial Stromal Cells in Endometriosis. Reproduction 2018, 156, 397–404. [Google Scholar] [CrossRef] [Green Version]
  177. Ormiston, M.L.; Chang, C.; Long, L.L.; Soon, E.; Jones, D.; Machado, R.; Treacy, C.; Toshner, M.R.; Campbell, K.; Riding, A.; et al. Impaired Natural Killer Cell Phenotype and Function in Idiopathic and Heritable Pulmonary Arterial Hypertension. Circulation 2012, 126, 1099–1109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Rätsep, X.T.M.; Moore, S.D.; Jafri, S.; Mitchell, M.; Brady, H.J.M.; Mandelboim, O.; Southwood, M.; Morrell, N.W.; Colucci, F.; Ormiston, M.L.; et al. Spontaneous Pulmonary Hypertension in Genetic Mouse Models of Natural Killer Cell Deficiency. Am. J. Physiol.—Lung Cell. Mol. Physiol. 2018, 315, 977–990. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Current NK cell therapies. Schematic showing the current use of NK cell therapies, starting with source, expansion, and activation, then demonstrating the recently developed alternative approaches. Figure created with BioRender.
Figure 1. Current NK cell therapies. Schematic showing the current use of NK cell therapies, starting with source, expansion, and activation, then demonstrating the recently developed alternative approaches. Figure created with BioRender.
Cancers 15 01743 g001
Figure 2. NK cell suppression. Schematic showing NK cell suppression after exposure to (A) TGFβ, (B) hypoxia, or (C) IDO. Figure created with BioRender (https://www.biorender.com/ [accessed on 10 February 2013]).
Figure 2. NK cell suppression. Schematic showing NK cell suppression after exposure to (A) TGFβ, (B) hypoxia, or (C) IDO. Figure created with BioRender (https://www.biorender.com/ [accessed on 10 February 2013]).
Cancers 15 01743 g002
Figure 3. Current interventions targeting NK cell suppressive factors. NK cell suppression via (A) TGFβ, (B) hypoxia, or (C) IDO. Figure created with BioRender (https://www.biorender.com/ [accessed on 11 February 2013]).
Figure 3. Current interventions targeting NK cell suppressive factors. NK cell suppression via (A) TGFβ, (B) hypoxia, or (C) IDO. Figure created with BioRender (https://www.biorender.com/ [accessed on 11 February 2013]).
Cancers 15 01743 g003
Table 1. Advantages and Limitations to Current NK Cell Therapies.
Table 1. Advantages and Limitations to Current NK Cell Therapies.
AdvantagesLimitations
Source
Peripheral Blood NK CellsReliable source of CD34 progenitor cells [64]NK cells make up only ~10% of all lymphocytes in peripheral blood
High expression of CD16+Extensive purification is required to reduce contamination [21]
Clinical studies have shown success with these cells after extensive enrichment and purification [69]Isolating large amounts of PB NK cells is difficult [70,71,72]
Cryopreservation has been shown to reduce cytotoxicity [71,73]
Umbilical Cord NK CellsGreater abundance than PB NK cells (15–30% of total lymphocytes) [23]UCB NK cells are immature
Fewer contaminating T cells in UCB than PB, reducing the risk of graft-versus-host disease [64]
Associated with good toleranceMay have reduced cytotoxic function [22]
Minimal graft-vs-host-disease or toxicity [24]
Induced Pluripotent NK CellEasily genetically modifiedLimited clinical success to date
High availabilityComplex differentiation steps
Ability to generate multiple doses from a single healthy donor [31,71]Safety concerns regarding toxicity
Commercial NK Cell LinesEasy to obtainMust undergo irradiation to prevent malignant expansion, which could limit persistence.
Highly cytotoxic
Easily expandable [32]
NK92 cells are the only cell line that has shown success in pre-clinical studies [31] Efficiency of cells after expansion is variable (4–95%) [74]
Expansion
Feeder CellsEffective expansion of large numbers of NK cells [75].Difficult to maintain cytotoxic function after expansion [49].
Feeder-Free ExpansionLarge amounts of highly active NK cells have been produced Cytotoxic function after expansion has not been well reported
Activation
IL-2Ability to restore NK cell cytotoxicity after exposure to various stressors [54].Systemic IL-2 leads to significant toxicity
Other Activating CytokinesLess toxic than IL-2Thought to provide only minimal clinical benefit
Many combination therapies are required to provide a therapeutic benefit
Genetic ManipulationAbility to target specific pathways of interestRelatively newer area of study
Ability to avoid toxic effects associated with global therapies
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Coyle, K.M.; Hawke, L.G.; Ormiston, M.L. Addressing Natural Killer Cell Dysfunction and Plasticity in Cell-Based Cancer Therapeutics. Cancers 2023, 15, 1743. https://doi.org/10.3390/cancers15061743

AMA Style

Coyle KM, Hawke LG, Ormiston ML. Addressing Natural Killer Cell Dysfunction and Plasticity in Cell-Based Cancer Therapeutics. Cancers. 2023; 15(6):1743. https://doi.org/10.3390/cancers15061743

Chicago/Turabian Style

Coyle, Kassandra M., Lindsey G. Hawke, and Mark L. Ormiston. 2023. "Addressing Natural Killer Cell Dysfunction and Plasticity in Cell-Based Cancer Therapeutics" Cancers 15, no. 6: 1743. https://doi.org/10.3390/cancers15061743

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