Next Article in Journal
Germination of Phaseolus vulgaris L. Seeds after a Short Treatment with a Powerful RF Plasma
Next Article in Special Issue
MicroRNAs as Modulators of the Immune Response in T-Cell Acute Lymphoblastic Leukemia
Previous Article in Journal
Nonameric Peptide Orchestrates Signal Transduction in the Activating HLA-E/NKG2C/CD94 Immune Complex as Revealed by All-Atom Simulations
Previous Article in Special Issue
Preclinical Evaluation of Invariant Natural Killer T Cells Modified with CD38 or BCMA Chimeric Antigen Receptors for Multiple Myeloma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

NK Cells in Chronic Lymphocytic Leukemia and Their Therapeutic Implications

1
Centro di Ricerca Emato-Oncologica (CREO), Department of Medicine and Surgery, Institute of Hematology, University of Perugia, 06129 Perugia, Italy
2
Department of Medicine and Sciences of Aging, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
3
Department of Oncology and Hematology, Ospedale Civile “Santo Spirito”, ASL Pescara, 65124 Pescara, Italy
4
Department of Medicine and Surgery, University of Perugia, 06129 Perugia, Italy
*
Author to whom correspondence should be addressed.
Co-first authors.
Int. J. Mol. Sci. 2021, 22(13), 6665; https://doi.org/10.3390/ijms22136665
Submission received: 14 May 2021 / Revised: 16 June 2021 / Accepted: 18 June 2021 / Published: 22 June 2021

Abstract

:
Key features of chronic lymphocytic leukemia (CLL) are defects in the immune system and the ability of leukemic cells to evade immune defenses and induce immunosuppression, resulting in increased susceptibility to infections and disease progression. Several immune effectors are impaired in CLL, including T and natural killer (NK) cells. The role of T cells in defense against CLL and in CLL progression and immunotherapy has been extensively studied. Less is known about the role of NK cells in this leukemia, and data on NK cell alterations in CLL are contrasting. Besides studies showing that NK cells have intrinsic defects in CLL, there is a large body of evidence indicating that NK cell dysfunctions in CLL mainly depend on the escape mechanisms employed by leukemic cells. In keeping, it has been shown that NK cell functions, including antibody-dependent cellular cytotoxicity (ADCC), can be retained and/or restored after adequate stimulation. Therefore, due to their preserved ADCC function and the reversibility of CLL-related dysfunctions, NK cells are an attractive source for novel immunotherapeutic strategies in this disease, including chimeric antigen receptor (CAR) therapy. Recently, satisfying clinical responses have been obtained in CLL patients using cord blood-derived CAR-NK cells, opening new possibilities for further exploring NK cells in the immunotherapy of CLL. However, notwithstanding the promising results of this clinical trial, more evidence is needed to fully understand whether and in which CLL cases NK cell-based immunotherapy may represent a valid, alternative/additional therapeutic option for this leukemia. In this review, we provide an overview of the current knowledge about phenotypic and functional alterations of NK cells in CLL and the mechanisms by which CLL cells circumvent NK cell-mediated immunosurveillance. Additionally, we discuss the potential relevance of using NK cells in CLL immunotherapy.

1. Introduction

Chronic lymphocytic leukemia (CLL) is the most common adult leukemia in the Western world and is characterized by the accumulation of clonal CD5+/CD19+ B cells in peripheral blood, lymph nodes, spleen and bone marrow [1,2]. Leukemic cells in CLL display a highly biological heterogeneity due to genetic and epigenetic alterations and microenvironment stimuli [3]. This feature results in a highly variable clinical course, in terms of presentation, outcome and therapy responses, with some patients displaying an indolent disease who do not require therapy and other patients showing a rapidly progressive disease despite early treatment [4]. In recent years, the development of targeted therapies, such as the inhibitors of B cell receptor (BCR) signaling and of B cell lymphoma 2 (Bcl-2) protein, has changed the treatment landscape of CLL [5]. However, despite their remarkable antitumor activity, targeted agents have shown some limitations, including the development of drug resistance and the low efficacy in high-risk patients [6,7]. More recently, combined therapies including the Bruton’s tyrosine kinase (BTK) inhibitor ibrutinib and the Bcl-2 inhibitor venetoclax, with or without anti-CD20 monoclonal antibodies (mAbs), have shown promising results in high-risk and older patients with CLL, although an extended follow-up of the trials to assess long-term outcomes has not been done [8,9,10]. Therefore, additional treatments are necessary to obtain deeper responses and overcome drug resistance in CLL.
CLL is characterized by an acquired dysregulation of the immune system and the ability of leukemic cells to circumvent immune recognition and elimination which result in increased risk of infections, decreased antitumor surveillance and tumor progression [11,12]. Particularly, T cells have been shown to have several dysfunctions, including impaired cytotoxicity, proliferation and ability to form immune synapses [13,14].
Novel immunotherapeutic approaches, such as chimeric antigen receptor (CAR) transduced T cells and immune checkpoint blockade, have shown impressive activity in other lymphoid malignancies [15,16,17] but discouraging results in CLL [18,19,20], mainly due to defects in the effector T cells [13,14]. Therefore, it is necessary to study the therapeutic potential of other immune effector cells for more effective immunotherapeutic strategies.
Natural killer (NK) cells can evoke potent antitumor activity [21,22]. This function is largely mediated by combined signaling through a variety of activating and inhibitory receptors which recognize specific ligands expressed on tumor cells [23]. NK cells can also kill tumor cells by antibody-dependent cellular cytotoxicity (ADCC), mediated by the CD16 receptor (FcγRIIIa), which recognizes the fragment crystallizable (Fc) portion of IgG bound to the target cell [24]. A competent ADCC by NK cells is important for its significant role in the therapeutic efficacy of various specific mAbs, such as anti-CD20 mAbs used for treating different B cell malignancies, including CLL [25,26,27,28]. The important role of NK cells in defense against tumors and leukemia is well documented by the success achieved in the T-depleted, haploidentical hematopoietic stem cell transplantation (haplo-HSCT) setting to cure high-risk acute leukemia [29,30]. The benefit of this therapeutic approach is mainly due to the graft-versus-leukemia (GvL) effect of donor NK cells, arising from grafted stem cells and/or infused with the graft.
Despite their important role in antitumor immunity, the functions of NK cells in CLL are not yet well defined, and data on the expression of NK receptors and the functionality of NK cells in CLL patients are controversial. Defects in NK cell cytotoxicity in CLL were first described decades ago [31,32,33], although several studies have reported that NK cell functions, including ADCC, are unaffected in CLL [34,35] or restored after cytokine treatment [36,37,38,39,40,41]. Notably, if in CLL, NK cell functions are retained or restored by an adequate stimulation, NK cells might be exploited for novel immunotherapeutic strategies, such as those based on NK cells genetically modified with chimeric antigen receptors targeting tumor antigens (CAR-NK cells), or on engineered soluble molecules bridging activating receptors on NK cells to tumor antigens. Recently, a clinical trial evaluating cord blood-derived CAR-NK cells in a small number of patients with relapsed or refractory CLL has shown satisfactory responses, encouraging further studies on NK cells in the immunotherapy of CLL [42].
In this review, we analyze the alterations of NK cells in CLL and the mechanisms by which CLL cells evade NK immune surveillance. Additionally, we address recent advances on the immunotherapeutic potential of NK cells for CLL.

2. Overall View on NK Cells

2.1. Role of NK Cells in the Immune System

NK cells are components of the innate immune system with an important role in antitumor and antiviral defense [21,22,43] and belong to group 1 innate lymphoid cells (ILCs). Group 1 ILCs also include ILC1s, which, along with group 2 and group 3 ILCs, represent the innate counterpart of the different CD4+ T helper cell populations [44]. ILC1s include various subsets having different localizations and functional activities and some common characteristics with NK cells, such as the production of interferon γ (IFNγ) [45].
NK cell activities (degranulation, cytotoxicity and cytokine release) are finely regulated by the balance between activating and inhibitory germline-encoded receptors expressed on NK cell surface [23,46]. In normal conditions, NK cells are inactive due to the binding of inhibitory receptors with a spectrum of classical and nonclassical human leukocyte antigen (HLA)-class I molecules constitutively expressed on autologous “self” cells (missing self-hypothesis) [47,48,49]. Tumor-transformed, virus-infected or stressed cells downregulate or lack HLA-class I alleles, thus boosting the NK cell-mediated killing due to the engagement of NK activating receptors with ligands preferentially expressed on target cells and absent or weakly expressed on normal cells [50,51,52]. Once activated, NK cells mediate cytotoxicity by releasing cytotoxic granules containing perforins and granzymes and producing proapoptotic cytokines, such as IFNγ and tumor necrosis factor α (TNFα) [53,54]. NK cells can also kill targets by activating the signaling pathway of TNF family death receptors through the expression of Fas ligand and TNF-related apoptosis-inducing ligand (TRAIL) [55]. Additionally, NK cells can indirectly mediate antitumor responses by producing inflammatory cytokines that link the innate and adaptive immune responses [56]. Cytokines can also modulate NK cell activity by transmitting either activating (IL-2, IL-12, IL-18, IL-15, IL-21, IL-27 and type I IFN) or inhibitory signals, such as transforming growth factor β (TGFβ) and IL-10 [57,58,59,60].
Although NK cells have traditionally been considered components of the innate immune system due to the lack of receptor gene rearrangement, there is increasing evidence that they share many characteristics with adaptive lymphocytes. NK cells develop from the same common lymphoid progenitor that gives rise to T and B cells [61]. Similar to T and B cells, NK cells require common γ chain-dependent cytokines for their development and homeostasis [62,63], their responsiveness is tuned through an “education” or “licensing” process analogous to T cell development in the thymus [64,65,66] and, strikingly, they show memory-like features [67].
In humans, two main NK cell subsets were originally identified on the basis of the intensity of CD56 and CD16 surface expression. The two subsets are differently distributed in peripheral blood (PB) and tissues: CD56dim/CD16pos (CD56dim) are predominant in PB, while CD56bright/CD16neg (CD56bright) are more abundant in tissues. CD56bright NK cells are relatively immature and poorly cytolytic, secrete cytokines (primarily IFNγ and TNFα) and undergo intensive proliferation in response to IL-2 or IL-15 [68,69]. By contrast, CD56dim NK cells are terminally differentiated and display a strong cytolytic activity and a rapid cytokine secretion capability upon activation [68,70].
In both humans and mice, NK cells show some common features with ILC1, but also substantial differences [71]. One key characteristic distinguishing ILC1s from NK cells is localization. NK cells recirculate between tissues and blood, whereas ILC1s reside in tissues, including liver, small intestine, thymus, uterus and salivary glands [72]. This different behavior correlates with the expression of distinct homing and adhesion molecules [71]. NK cells and ILC1s have also distinct developmental requirements. In mice, it has been shown that NK cell development depends on the transcription factor eomesodermin (Eomes) [73,74], whereas ILC1s require various transcription factors, including T-bet, Hobit and Eomes, depending on the tissue localization [75,76]. Whether NK cells and ILC1s derive from a common progenitor or different progenitors remains unclear. Previous studies have proposed that NK cells branch off the ILC development at the stage of the early innate lymphoid progenitor, whereas ILC1s derive from the later common helper innate lymphoid progenitor, which gives rise to mature ILCs but has lost NK cell potential [77,78]. However, a common progenitor for NK cells and ILC1s has also been recently identified [79]. Another difference between NK cells and ILC1s is that these latter are supposed to be less cytotoxic than NK cells, based on the differential expression of granzymes and perforins [73]. However, given the high expression of TRAIL in ILC1s, cytotoxic mechanisms cannot be excluded in certain conditions.
All these observations suggest that it is very difficult to distinguish NK cells from ILC1s. This depends not only on the heterogeneity of cell populations but also on the plasticity of ILC1s, which may change their functional capability in a given microenvironment, particularly at tumor sites under the influence of cytokines [80]. Interestingly, it has been shown that TGF-β, present in the tumor microenvironment, can mediate NK cell conversion to ILC1s with proangiogenic and immune-tolerant features [81], resulting in transitional phenotypes and functions between NK cells and ILC1s, which further complicate their discrimination. As consequence, given that ILCs have been investigated only during the last 10 years, we cannot exclude that previous studies on NK cells may have overlooked the contributions of ILC1s. In the context of cancer, an inappropriate discrimination between these two cell types might also have led to erroneous conclusions regarding the specific impact of their targeting on tumors. Given the difficulties in distinguishing NK cells from ILC1s, novel approaches should be developed to better define NK/ILC1 identity in normal and pathological conditions. In this regard, a compelling study of Colonna’s group used single-cell RNA sequencing to elucidate gene signatures of mouse ILC1-NK cells from tissues, tumors and the circulation [82]. Strikingly, these authors identified unique transcription factors, phenotypic markers and metabolic features that distinguish tissue-resident NK cells and ILC1s from circulating NK cells, providing the guide for future spatial transcriptomic and immunohistochemical analyses [82].
NK cells have important functions in the immune system. Concerning the role of NK cells against pathogens, studies on healthy individuals and individuals with NK cell deficiencies have shown that NK cells are involved in the control of several infections, including Epstein–Barr virus (EBV) [83,84], herpes simplex [85], human immunodeficiency, influenza and hepatitis C viruses [43,86]. Additionally, there is evidence that NK cells can react to cytomegalovirus (CMV) infection and prevent CMV reactivation following allogeneic stem cell transplantation [87].
The involvement of NK cells in defense against cancer began to emerge when these cells were discovered, given their ability to kill tumor cell lines in vitro [88]. Thereafter, several studies have confirmed NK cell-mediated killing of other types of tumor cell lines in vitro and in experimental mouse models [89,90], where NK cells have been shown to be involved in rejection responses against induced and spontaneously developing tumors [91,92]. Over the years, it has become evident that NK cells are involved in tumor immunosurveillance [21]. Studies on animal models indicate that knockout of key NK cell activating receptors leads to a higher incidence of tumor formation compared to controls with wild-type expression of the receptors [93,94].
In humans, clinical follow-up studies have shown that individuals with low NK cell function early in life have an increased risk of cancer compared with matched controls [95]. Conversely, high density of tumor-infiltrating NK cells has been linked with a good prognosis in different carcinomas [96]. Additionally, clinical observations have indicated that a ligand repertoire on acute myeloid leukemia (AML) blast favoring NK cell activation is positively correlated with a better outcome of patients undergoing chemotherapy [97]. NK cells have also been shown to eliminate cancer stem cells, a subset of cells with self-renewal ability involved in the generation and evolution of tumors [98]. However, the critical role of NK cells in targeting human tumors emerges from the seminal studies of Velardi’s group on haplo-HSCT against AML [29,30], which have paved the way for intense research on NK cell-based cancer immunotherapy. In this context, as endogenous NK cells are defective in both solid tumors [99,100] and hematological malignancies [101,102], several efforts have been made to discover strategies for restoring and/or bolstering NK cell functions or for providing patients with functional NK cells [103].

2.2. NK cell Receptors and Ligands and Their Role in Regulating NK Cell Activity

NK cell functions are regulated by distinct receptors which upon interaction with specific ligands expressed on target cells integrate activating and inhibitory signals triggering or blocking NK cell cytotoxicity [23,46]. Major activating NK cell receptors are the natural cytotoxicity receptors (NCRs), type I molecules of the immunoglobulin-like (Ig) family, having a transmembrane domain associated with immunoreceptor tyrosine-based activation motif (ITAM)-bearing signaling proteins, namely FcεRIγ, CD3ζ and DAP12 [104]. NCRs include NKp30 and NKp46, which are constitutively expressed on NK cells, and NKp44 which is acquired upon activation [104]. Several NCR ligands have been identified. All NCRs can recognize heparan sulfate glycosaminoglycans (HSGs), significantly upregulated in the surface of tumor cells [105], and viral hemagglutinins (HAs) [106]. Important NKp30 ligands are the B-7 family member B7-H6, absent on healthy cells but highly expressed by a wide range of tumor cells [107], and HLA-B-associated transcript 3 (BAT3), also known as Bcl2-associated athanogene 6 (BAG6), which can be released by tumor cells inducing either NK cell activation or suppression [108,109]. NKp44 can recognize a splice variant of mixed-lineage leukemia 5 (21spe-MLL5), absent in the normal tissues but expressed in a variety of hematopoietic and nonhematopoietic tumors [110]. An additional NKp44 ligand is the proliferating cell nuclear antigen, a nuclear/cytoplasmic factor that can be expressed on the membrane of cancer cells and upon binding with NKp44 induces NK cell inhibition due to immunoreceptor tyrosine-based inhibitory motifs (ITIMs) located in the NKp44 cytoplasmic tail [111]. NKp44 also binds soluble ligands, including (i) platelet-derived growth factor (PDGF)-DD, which activates NK cells and NK cell-mediated release of proinflammatory cytokines and chemokines [112] and (ii) nidogen-1, a glycoprotein involved in the adhesion of cells with the extracellular matrix, which, once released by tumor cells, may prevent the NK cell-mediated attack, representing an immunosuppressive mechanism [113]. Other soluble NCR ligands have been identified, such as the complement factor P (CFP), also known as properdin, which is recognized by NKp46 [114]. In particular, it has been shown that CFP binds to a recombinant NKp46 Fc-fusion protein by inducing in NKp46 reporter cells an alternative signaling pathway that does not induce degranulation, but secretion of the chemokine XCL1 [114]. XCL1 has a direct antimicrobial activity [115] and has been shown to recruit dendritic cells capable of antigen cross-presentation for CD8+ T cell activation during bacterial and viral infections [116]. These findings suggest that microbes opsonized with CFP can stimulate NKp46-mediated antibacterial activity of NK cells.
Another major NK activating receptor is the NKG2D homodimer, a type II and C-type lectin-like molecule, which recognizes major histocompatibility complex (MHC) class I-related glycoproteins A and B (MIC-A and MIC-B) and six non-MHC-encoded UL16-binding proteins 1–6 (ULBP1-6), molecules restrictedly expressed in benign cells but upregulated in stressed and transformed cells [117]. However, tumor cells have developed several mechanisms to circumvent NKG2D-mediated recognition, including the release of soluble ligands [118,119,120], the negative regulation of NKG2D ligand expression at the post-transcriptional level [121] and the secretion of immunosuppressive cytokines that reduce NK cell surface expression of NKG2D [122]. NK cells also express costimulatory receptors that cooperate with NCRs and NKG2D by amplifying NK cell activation. Among them, DNAX accessory molecule 1 (DNAM-1) recognizes the poliovirus receptor (PVR) and nectin-2 expressed on various acute leukemias by inducing antitumor activity [123]. Other coreceptors include members of the signaling lymphocytic activation molecule family, such as NK-T-B-antigen (NTB-A), which displays homophilic interactions, and 2B4, which binds to CD48. High levels of CD48 and NTB-A have been found in EBV-infected B cells and lymphomas [124]. NK cells also express the coreceptor CD59 [125] and the adhesion molecule LFA-1, important for a polarized degranulation [126]. Additional activating receptors of NK cells are specific for HLA-class I molecules and include the CD94/NKG2C heterodimers, consisting of type II proteins of the C-type lectin family, and the killer cell Ig-like receptors (aKIRs) [127]. Ligands for only some of these receptors have been identified. CD94/NKG2C binds with low affinity to HLA-E, a nonclassical HLA-class I molecule characterized by limited polymorphism [128]. aKIRs are specific for epitopes shared by distinct groups of HLA-class I allotypes. Specifically, KIR2DS1 and KIR2DS2 recognize HLA-C C2 and HLA-C C1 allotypes, respectively; KIR2DS4 recognizes HLA-C bearing either C1 or C2 epitopes and only one HLA-A allotype; KIR3DS1 binds to HLA-B allotypes bearing Bw4 epitope [129,130]. Additionally, a potent activating receptor of NK cells is CD16, which mediates ADCC [24,27]. This receptor is of great clinical relevance for cancer immunotherapy and is widely exploited to enhance the antitumor NK cell activity using mAbs or engineered bispecific and trispecific constructs directed to tumor antigens [131,132].
In NK cells, inhibitory signals are mainly mediated by HLA-class I-binding receptors which regulate NK cell function and prevent NK cell-mediated damage to healthy tissues [47,65,66]. These receptors include inhibitor members of the KIR family (iKIRs) and the CD94/NKG2A heterodimer recognizing classical and nonclassical HLA-class I molecules, respectively. Both these types of receptors are involved in NK cell education, a functional maturation process that allows self-inhibited NK cells to become cytotoxic after interaction with cells lacking self HLA-class I expression but expressing ligands for activating NK receptors [48,49,65,66]. Strikingly, similarly to HLA molecules, iKIRs are encoded by a polygenic and polymorphic KIR gene family [133], which segregate independently of HLA-class I genes leading to diverse compound genotypes [134]. These characteristics influence the possible KIR/HLA interactions and lead to a great heterogeneity of NK cell phenotypes among different individuals [68,135,136]. Combinations of HLA-class I and KIR variants may also influence resistance to infections, susceptibility to autoimmune diseases and outcome after hematopoietic stem cell transplantation (HSCT) [134].
In humans, the main iKIRs include KIR2DL1, which binds to HLA-C C2 allotypes; KIR2DL2 and KIR2DL3, which recognize HLA-C C1 and two specific HLA-B allotypes; KIR3DL1, which is specific for HLA-B and some HLA-A allotypes sharing the Bw4 epitope; and KIR3DL2, which binds to some specific HLA-A allotypes [130]. KIR2DL4 is an atypical receptor that has both activating and inhibitory signaling domains and binds to the nonclassical HLA-class I molecule HLA-G [137].
CD94/NKG2A recognizes HLA-E with high affinity [128] and represents an important target for checkpoint inhibitor cancer immunotherapy [138]. An additional HLA-specific inhibitory receptor is the Ig-like transcript 2 (ILT2), also named CD85j, leukocyte Ig-like receptor B1 (LILRB1) or LIR-1, which interacts with classical (HLA-A, HLA-B, HLA-C) and nonclassical (HLA-G) HLA-class I molecules [139]. NK cells, similarly to T cells, also express other inhibitory checkpoints responsible for maintaining immune cell homeostasis [140]. One of them is the programmed death-1 (PD-1) receptor which binds to its ligands PD-L1 and PD-L2 [141,142]. These latter are expressed at low levels on healthy tissues but upregulated on various tumor types, including hematological malignancies, where a high expression is associated with poor prognosis [143]. Other inhibitory checkpoints of NK cells include (i) T cell Ig and ITIM domains (TIGIT) and CD96/Tactile, which bind to PVR and nectin-2 by competing with the costimulatory receptor DNAM-1 [144,145]; (ii) T cell Ig and mucin domain-containing protein 3 (TIM-3), whose main ligand is galectin-9 [146]; and (iii) lymphocyte-activation gene 3 (LAG-3), a receptor homologous to CD4 that recognizes HLA-class II molecules and whose effects on NK cell functions are not yet well defined [147]. Receptor–ligand interactions regulating NK cell activity are shown in Figure 1.

3. NK Cells in CLL

3.1. NK Cell Functions and Dysfunctions

Impaired cytolytic activity of NK cells in patients with CLL was first described in the early 1980s and was mainly attributed to intrinsic NK cell defects in cytotoxic machinery [31,32,33]. Later studies showed that autologous NK cells are unable to eliminate CLL cells not only due to their intrinsic defects but also due to immune escape mechanisms developed by leukemic cells [148,149,150,151,152]. By contrast, other authors reported that peripheral NK cells from CLL patients have the major phenotypic characteristics of competent NK cells and are functional in terms of degranulation, cytokine production and ADCC [34]. Additionally, there is evidence showing that CLL-derived NK cells retain intrinsic functionality, given that it can be restored by adequate activating signaling, including cytokines such as IL-2, IL-15, IL-21 and IL-27 [36,37,38,39,40,41,153], and anti-CD20 mAbs that induce ADCC [34,37,39,40,153,154]. Therefore, NK cells, given their preserved ADCC function and the reversibility of their defects, represent attractive effectors for immunotherapy in CLL [155]. In addition, there are studies showing that the expression of HLA-class I molecules on CLL cells is downregulated in 65–80% of patients [156,157], allowing CLL cells to escape from specific T lymphocyte surveillance and be targeted by NK cells.
Despite the impaired activity of NK cells in CLL, it has been shown that the number of NK cells is increased in the peripheral blood of CLL patients compared to healthy individuals and predicts good prognosis [158,159,160,161]. Conversely, other studies did not find any correlation between high NK cell numbers and CLL prognosis [162], whereas other authors showed that the expanded NK cells exhibit enhanced susceptibility to activation-induced cell death and express elevated levels of CD27, which is normally associated with an expansion of immature NK cells or a decline in mature NK cells [163]. One cause of the discrepancies in NK cell functionality in CLL may be CMV infection, which induces a reconfiguration of the phenotype and functions of the NK cell compartment, characterized by an adaptive expansion of mature NK cells expressing high levels of the activating receptor CD94/NKG2C [164,165]. Discrepancies in NK cell functions in CLL may also depend on different patient and healthy donor cohorts, varied criteria of patient selection, different experimental stimuli activating NK receptors or ADCC responses and the methods used. Altogether, these observations show that the role of NK cells against CLL has not been fully understood, underlining the need to better define NK cell functions and dysfunctions in this leukemia.

3.2. Alterations in NK Cell Receptor–Ligand System

Dysregulation and imbalance of activating and inhibitory NK cell receptors could be one of the main reasons for NK cell impairment in CLL. Therefore, particular attention has been focused on the alterations in the NK cell receptor–ligand system and their role in regulating the NK cell-mediated response against CLL cells. Several studies have shown that NK cells of CLL patients have a decreased expression of different activating receptors, such as NKp30, NKp46, NKG2D and DNAM-1, compared with healthy donors and that this altered phenotype is accompanied by an impaired cytotoxic activity, degranulation and killing of target cells [37,39,161,162,163,166]. Conversely, Costello et al. found that expression levels of NKp30, NKp40 and NKp46 are similar in NK cells of CLL patients and healthy donors, but analysis of different groups of CLL patients showed that lower levels of NKp30 and NKp44 are associated with poor prognostic factors [35]. Even the decrease in NKG2D expression on CLL-derived NK cells has been found more marked in patients with advanced and progressive disease, suggesting that CLL cells may play a role in downregulating NKG2D expression [162]. This observation is consistent with studies showing that coculture of CLL cells with NK cells from healthy donors decreases NKG2D expression, NK cell cytotoxicity and IFNγ production, indicating that CLL cells are able to hamper NK cell functions and create a hyporesponsive phenotype and further supporting that the reduced cytotoxicity of CLL-derived NK cells is not due to intrinsic defects in their effector programs [39]. Based on the evidence that CLL cells release high amounts of TGFβ, which has also been found in CLL patient serum [167], and that TGFβ reduces NKG2D expression on NK cells of healthy donors [166], it is plausible that TGFβ released by CLL cells represents a mechanism downregulating NKG2D expression on NK cells and consequently impairing NK cell activity. Additionally, in CLL, TGFβ as well as the immunosuppressive IL-10 can be also released by regulatory T cells and myeloid-derived suppressor cells, key players of immune dysfunctions in CLL [168,169]. Another mechanism that negatively impacts the activity of NK cells in CLL is the low expression on leukemic cells of the ligands for NK cell activating receptors, which is mainly due to the shedding of the ligands released as soluble molecules [148,152,170,171]. This event prevents tumor cell recognition by NK cells [37], representing an escape mechanism not only in CLL but also in several other malignancies [109,118]. The shedding of the ligands for the NK activating receptors in CLL and its role in impairing NK cell antileukemic activity is discussed in the next paragraph. Regarding the activating receptor CD94/NKG2C, whose expression has been found to be significantly increased in NK cells of normal individuals after CMV infection [164,165], a recent study has shown that CLL patients exhibit a reduced percentage of CD94/NKG2C+ NK cells compared with healthy donors, which is independent of CMV serostatus but is related to the exposure to leukemic cells, given its association with higher lymphocytosis [172]. These data are discordant with previous studies showing that in CLL patients, there is an expansion of CD94/NKG2C+ NK cells which is driven by CMV instead of the leukemic cells [39,173].
Concerning the NK cell inhibitory receptors, it has been shown that CD94/NKG2A hampers NK cell cytotoxicity against CLL cells through the binding to HLA-E molecules [174], which are highly expressed on the CLL cell surface [37,174,175]. In agreement, it has been shown in other malignancies that the binding of HLA-E to CD94/NKG2A induces signals that suppress cytokine secretion and direct cytotoxicity of effector cells against malignant cells, playing an important role in the tumor escape [138]. Studies to better define the inhibitory functions and the clinical relevance of HLA-E in CLL have revealed the presence of soluble HLA-E (sHLA-E) in the plasma of CLL patients [175]. In particular, these studies have shown that high levels of sHLA-E are associated with early disease progression and treatment requirement and impair the function of NK cells by skewing them towards an immunosuppressive phenotype. Additionally, sHLA-E correlates with the expression of the specific HLA-E*01:03 allele, which suggests that both HLA-E genotype and plasma sHLA-E levels are potential biomarkers for identifying CLL patients with a high risk of early disease progression and provides the first functional clues for HLA-E-mediated immune response modulation in CLL [175].
Studies on inhibitory KIRs in CLL-derived NK cells have reported that the expression levels of KIR2DL2/3 and KIR3DL1 are similar in NK cells from CLL patients and healthy individuals and remain stable during disease progression [34,37]. Conversely, a weak decrease in the expression of KIR2DL1, which recognizes group-2 HLA-Cw alleles, has been found in CLL patients with an unfavorable prognosis [34], and this association is probably due to the higher frequency of its ligand HLA-Cw*06 in CLL cohorts than in healthy controls [176,177]. These phenotypic features of NK cells do not completely explain NK cell dysfunction in CLL. McFarlane et al. reported that the impaired activity of NK cells in CLL is associated with a striking reduction in the frequency and viability of NK cells expressing KIR2DL1 and/or KIR3DL1, which progressively lose their functions over disease course [163]. These results suggest that mature KIR-expressing NK cells can respond to the high circulating CLL burden but undergo activation-induced apoptosis favoring the expansion of nonfunctional NK cells.
It has also been reported that NK cells from CLL patients, particularly those with advanced disease, overexpress the ILT2/CD85j inhibitory receptor [178,179], while CLL cells abnormally express its ligand HLA-G, which has been found to be associated with poor prognosis and to suppress NK cell-mediated cytotoxicity [149,179,180]. Indeed, blockade of either ILT2/CD85j or membrane-bound HLA-G with the corresponding neutralizing mAbs increases NK cell cytotoxicity against CLL cells [149,178]. Additionally, plasma samples from CLL patients have been reported to contain increased levels of soluble HLA-G and to be capable of dampening the viability and cytotoxic function of NK ceWe lls from healthy donors in vitro [181]. The role of membrane-bound and soluble HLA-G forms as a strategy of CLL cells to evade immune defenses is discussed in further detail in the next paragraph. As an additional mechanism that may affect NK antitumor activity in CLL, in line with data observed in conventional T cells, the immune checkpoint TIM-3 was found to be aberrantly expressed on the NK cell compartment of CLL patients and associated with poor prognostic factors [161]. In this context, recent studies have shown that CLL cells from patients with advanced clinical stage exhibit high mRNA levels of galectin-9, the ligand of TIM-3 [182]. In addition, the serum levels of galectin-9 have been found significantly increased in CLL patients compared with the control group and have been associated with poor cytogenetic and serum prognostic factors and treatment failure [183]. It has been shown in other malignancies that sustained TIM-3 expression on NK cells can lead to an exhausted/dysfunctional phenotype of NK cells that is rescued by TIM-3 blockade [184]. It is well known that interaction between PD-1 expressed on T cells of CLL patients and its ligand PD-L1 expressed on CLL cells strongly impairs T cell functions, inducing an exhausted T cell phenotype [185,186]. Despite the evidence that PD-L1 expression in other tumor cells results in functional NK cell impairment [187] and that CLL cells express high levels of PD-L1 [182,188], the involvement of the PD-1/PD-L1 axis in regulating NK cell functions in CLL patients remains to be defined.

3.3. Escape of CLL Cells from NK Cell Antitumor Activity

CLL cells employ multiple mechanisms to evade NK cell immune surveillance. One of them relies on the ability of tumor cells to release from their surface, through proteolytic shedding, the ligands for the activating receptors expressed on NK cells [120]. In other malignancies, it has been shown that soluble ligands counteract the immune surveillance by both NK and T cells by promoting the endocytosis and degradation of their receptors expressed on the surface of the effector cells, which thus are unable to recognize and eliminate tumor cells [119,189,190]. Additionally, soluble ligands impair the ability of NK cells to self-renew in the tumor host, thus perturbing NK cell homeostasis [191]. Given that NK cells play a key role in shaping adaptive immunity by providing IFNγ and priming dendritic cells, soluble ligands strongly impair tumor immunity. Soluble ligands detected in the plasma of cancer patients have also been identified as prognostic factors [117].
In the CLL context, Reiners et al. have shown that the soluble NKp30 ligand BAG6/BAT3 detected in the plasma of CLL patients suppresses NK cell cytotoxicity and even downregulates the expression of CD16 and CD56 on NK cells of healthy donors [148]. The same authors have also demonstrated that BAG6, when expressed on the surface of exosomes, is able to activate NK cell cytotoxicity, suggesting that exosomal BAG6 can represent a component of “induced self-activation” of NK cells and that a dysregulated balance of exosomal vs. soluble BAG6 expression may cause CLL evasion from NK cells [148]. Plasma of CLL patients also contains higher levels, compared with healthy donors, of other factors known to compromise NK cell function, such as macrophage migration inhibitory factor [192] and the soluble NKG2D ligands MIC-B and ULBP2 [148]. Additionally, plasma levels of soluble BAG6/BAT3, MIC-B and ULBP2 are further increased in advanced disease stages, suggesting a role as prognostic factors [148]. The prognostic significance of soluble MIC-A, MIC-B and ULBP2 in CLL has been confirmed in other studies which have shown that among these ligands, soluble ULBP2 is the most important prognostic marker to identify early-stage patients with risk of disease progression [170]. A comprehensive analysis of NKG2D ligand expression in CLL and other leukemias has shown that soluble NKG2D ligands in patient sera reduce NKG2D expression on NK cells, resulting in impaired NK antileukemic activity, which depends on the levels of surface-expressed NKG2D ligands [119].
HLA-G expression on the CLL cell surface can represent an additional mechanism by which these tumor cells escape the immune response [149]. HLA-G is a nonclassical HLA-class I molecule that is normally expressed in tissues where the immune system needs to be constantly suppressed, including fetal tissues, adult immune-privileged organs and cells of the hematopoietic lineage [193]. Upregulation of HLA-G in cancer contributes to serious immunosuppression, because besides inhibiting NK cell cytotoxicity, proliferation and transendothelial migration, it also inhibits the functions of cytolytic T cells, B cells and dendritic cells [194]. HLA-G also induces T cell apoptosis and differentiation of CD4+ and CD8+ T lymphocytes into regulatory T cells [195,196]. This wide range of effects is probably due to the widespread expression of its receptors. Indeed, in addition to KIR2DL4, HLA-G binds to multiple other inhibitory receptors, including ILT2/CD58j, ILT4 and CD160, which are expressed on several immune cells [194].
Membrane-bound HLA-G levels on CLL cells have been found elevated in CLL patients with progressive disease and short treatment-free survival, and a multivariate Cox regression analysis has revealed that the HLA-G status of CLL cells has an independent prognostic value similar to that of the established prognostic markers ZAP-70 and CD38 [180,197]. Additionally, in CLL patients with higher surface HLA-G expression, it has been shown that the sera contain higher levels of IL-10, suggesting that this cytokine may regulate HLA-G expression on CLL cells [180]. Conversely, other studies in a larger cohort of patients have observed a low expression of HLA-G on CLL cells of all samples and no significant correlation with clinical data or progression-free survival time, indicating that the prognostic role of HLA-G in CLL remains a controversial issue [198]. Increased plasma levels of soluble HLA-G have been reported in CLL patients compared to healthy donors, but no significant correlation has been found with known CLL prognosticators [199,200].
Compared to classical HLA, the HLA-G gene is conserved in the coding region but shows different polymorphisms in the 5′ upstream regulatory (URR) and the 3′ untranslated (UTR) regions [201]. A number of studies have indicated that HLA-G polymorphisms are associated with HLA-G expression, cancer susceptibility and cancer development [202]. Among these polymorphisms, a 14 base pair insertion/deletion (ins/del) (rs66554220) in the 3′ UTR has been shown to influence mRNA stability and protein production [203]. Interestingly, Rizzo et al. have shown that in CLL, there is a significant correlation between the del/del HLA-G genotype and increased plasma levels of soluble HLA-G, but not between this genotype and increased membrane HLA-G levels, probably because of the instability of membrane HLA-G forms which are rapidly released into the plasma [181]. The del/del HLA-G genotype is also associated with the expansion of circulating regulatory T cells, which in CLL positively correlate with the presence of clinical and biological features of aggressive disease [181,204]. Additionally, increased soluble HLA-G levels in del/del patients are associated with impaired NK cell cytotoxicity through its binding to KIR2DL4 ligand expressed by NK cells, as confirmed by in vitro incubation of normal NK cells with plasma samples from CLL patients with variable soluble HLA-G levels. [181].
In addition to evading and suppressing NK cell activity, CLL cells can also take advantage of the interactions with NK cells by receiving signals that promote tumor cell growth and survival. This observation is supported by studies showing that interaction between the glucocorticoid-induced TNFR-related protein (GITR) receptor and its ligand (GITRL) expressed at high levels in NK cells of CLL patients and CLL cells, respectively, induces in the latter the release of TNF, IL-6 and IL-8 [205], which are known to act as autocrine/paracrine growth and survival factors for CLL cells [206,207,208]. Similarly, even the interaction between the costimulatory molecule 4-1BB expressed on CLL-derived NK cells and the 4-1BBL, highly expressed on CLL cells, leads to the release of TNF by CLL cells [209]. Notably, both GITRL and 4-1BBL send signals that impair direct and antibody-induced NK cell cytotoxicity and IFNγ production [205,209], suggesting that both GITR/GITRL and 4-1BB/4-1BBL interactions may contribute to CLL pathophysiology and resistance to immunotherapy. Additionally, it has been reported that activated NK cells release soluble B cell activating factor (BAFF) which enhances the metabolic activity of CLL cells and reduces their susceptibility to direct NK cell cytotoxicity and ADCC induced by anti-CD20 therapeutic mAbs, effects which are prevented by the BAFF neutralizing mAb belimumab [210].
A hypothetical model of CLL escape from NK cell immune surveillance is shown in Figure 2.

4. Immunotherapeutic Approaches Involving NK Cells in CLL

NK cell-based therapeutic strategies in CLL and in other malignancies aim to potentiate and/or restore NK cell activity or to provide patients with functional NK cells able to kill tumor cells. These objectives may be achieved using different approaches, which are detailed below and summarized in Figure 3.

4.1. Enhancement of NK Cell-Mediated ADCC

Therapeutic approaches exploiting NK cell-mediated ADCC in CLL employ either tumor-specific mAbs or bispecific and trispecific killer engagers.

4.1.1. Monoclonal Antibodies

One of the NK cell-based therapeutic strategies in CLL relies on the ability of NK cells to kill cancer cells opsonized with mAbs via ADCC, a mechanism based on the engagement of CD16 receptor with the Fc fragment of IgG [24,27]. In CLL, therapeutic activation of NK cell-mediated ADCC is induced by various humanized mAbs that target different CLL surface antigens, including CD20, CD19 and CD37 (Figure 3A, left). mAbs targeting CD20 were the first immunotherapeutic approach in CLL, and the first approved anti-CD20 mAb was rituximab [28], which mainly acts by inducing ADCC of NK cells and complement-dependent cytotoxicity [211]. Although rituximab had limited success as a single agent [212], its efficacy was increased when it was used in combination with fludarabine and cyclophosphamide (FCR). FCR represents an option therapy for treatment-naïve and relapsed patients [213,214], but is less effective in patients with unmutated IGHV; mutated TP53; del (17p) and del (11q); and mutations in NOTCH1, SF3B1 and BIRC3 [215,216,217]. There are various explanations for the limited efficacy of anti-CD20 mAbs as monotherapy in CLL. For example, loss of CD20 antigen on CLL cells following rituximab treatment leads to expansion of antigen-loss variants resistant to NK cell-mediated ADCC [218]. The expression of particular polymorphisms of FcγRIIIa can represent an additional limitation that reduces the affinity of rituximab to FcγRIIIa on NK cells, resulting in poor clinical responses [219]. Furthermore, rituximab can induce monocyte-mediated immunosuppressive mechanisms, such as the release of ROS that inhibit NK cell-mediated ADCC, limiting the benefit of the therapy [220]. The limited efficacy of therapeutic mAbs as single agents might be also related to the impaired NK cell activity in the patients. This could be circumvented by the combination of the mAb with allogeneic NK cells. Studies have reported new protocols for activation/expansion of cord blood-derived NK cells, which, in combination with rituximab, mediate a high ADCC against primary CLL cells in vitro [221].
More recent anti-CD20 mAbs are ofatumumab, which targets a different epitope than rituximab, and obinutuzumab (GA101) and ublituximab (TG-11019), both having an engineered Fc fragment with increased affinity for CD16 [222]. Ofatumumab and obinutuzumab have shown efficacy in phase 3 clinical trials when used in combination with chemotherapy [223] or with inhibitors of BCR [224,225,226] or Bcl-2 [227]. Ublituximab has been shown to increase NK cell-mediated ADCC against CLL cells ex vivo compared to rituximab [154] and to have promising efficacy in phase 2 and/or phase 3 clinical trials either as a single agent or in combination with the BTK inhibitor ibrutinib and the next-generation PI3K inhibitor umbralisib in high-risk CLL [228,229,230].
An additional target for mAb-based therapeutic strategies in CLL is CD19. The anti-CD19 afucosylated mAb inebilizumab (MEDI-551) and the Fc-engineered (S239D/I332E) mAb tafasitamab (MOR208; XmAb5575) have been shown to enhance NK cell-mediated ADCC against B lymphoma and leukemia cell lines compared with unmodified anti-CD19 mAbs [231,232]. Inebilizumab and tafasitamab were also tested in phase 1 trials and showed tolerability and preliminary efficacy in previously treated and relapsed CLL [233,234].
Another target currently under investigation for CLL immunotherapy is CD37 [235]. Several CD37-targeting therapeutics have been clinically evaluated [236]. Among them, BI 836826 (MAb 37.1), an Fc-engineered mAb able to induce apoptosis and enhance NK cell-mediated ADCC, has been shown to potentiate the cytotoxicity of the PI3K inhibitor idelalisib in relapsed CLL cells ex vivo [237]. In a phase 1 study in relapsed/refractory CLL, acceptable tolerability and preliminary efficacy were observed [238]. An additional anti-CD37 therapeutic molecule that has been engineered to increase NK cell-mediated ADCC activity is otlertuzumab (TRU-016), a monospecific IgG fusion protein built using the ADAPTIR (modular protein technology) platform [239]. When used as a single agent, it has shown a modest activity and an acceptable safety profile in a phase 1 study enrolling treatment-naïve and pretreated CLL patients [240]. In a phase 2 study in patients with relapsed or refractory CLL, otlertuzumab in combination with bendamustine increased the response rate and prolonged the progression-free survival compared with bendamustine alone [241].

4.1.2. Bispecific and Trispecific Killer Cell Engagers

New potential therapeutic approaches able to boost NK cell activation at the tumor site by targeting CD16 involve the use of bispecific and trispecific killer engagers, BiKEs and TriKEs, respectively [132]. BiKE constructs comprise a single-chain variable fragment (scFv) domain specific for a tumor antigen and a second scFv specific for an activating receptor on effector cells, thus forming an immunological synapse and triggering cytotoxic responses [242]. TriKEs bind two different tumor antigens, allowing the recognition of cancer cells even when one antigen is lost, thus preventing tumor escape [218,243]. So far, in CLL, BiKEs and TriKEs engaging NK cells have been investigated only in a preclinical setting, but the obtained data show that these constructs have a high potential for CLL immunotherapy. The first evidence of the therapeutic potential of BiKEs for NK cell immunotherapy in CLL has been provided by Feys’ group, who generated different BiKE constructs for CD19 and CD16 able to induce in vitro ADCC against primary CLL cells [244,245]. Later, Gleason et al. showed the ability of a CD16/CD19 BiKE and a CD16/CD19/CD22 TriKE to directly activate NK cells through CD16 by increasing both NK cell cytotoxicity and production of IFNγ against CLL cells [246]. Although CD20 expression on CLL cells is higher than that of CD19, the CD16/CD19/CD22 construct has been more effective than rituximab in targeting CLL cells, suggesting that simultaneous targeting of CD22 and CD19 is advantageous [246]. The CD16xCD19 construct has also been modified to include the stimulatory IL-15 cytokine moiety. This new construct, the 161519 TriKE, induces potent healthy donor NK cell activation, proliferation and direct killing of primary CLL cells, holding great potential to cure refractory CLL [247] (Figure 3A, right).
Other authors have tested the possibility to activate anti-CLL NK cell cytotoxicity through the engagement of NKG2D by a new construct, namely ULBP2/aCD19/aCD19, a trispecific immunoligand containing ULBP2 as a natural ligand for NKG2D receptor on NK cells and two sets of a CD19-specific scFv (aCD19) to target CLL cells [248]. ULBP2/aCD19/aCD19 efficiently binds to all target moieties simultaneously by retargeting NK cells to kill tumor cells in an antigen-specific manner and mediates efficient NK cell-dependent killing of primary CLL cells both in allogenic and autologous settings. Additionally, ULBP2/aCD19/aCD19 has shown significant in vivo ability to activate and retarget NK cells to kill transplanted MEC1 cells in a xenograft mouse model [248].

4.2. Restoring NK Cell Functions by Targeting The Immune Checkpoints

A potential approach involving NK cells in CLL immunotherapy is the blocking of the immune checkpoints [249]. However, in CLL, most of the attention for unleashing antitumor responses with checkpoint inhibitors has been focused on T cells, with PD-1 as one of the most studied immune checkpoints [182,185,186,250,251,252,253]. Clinical trials evaluating PD-1-blocking mAbs in CLL have shown disappointing results, especially when they were used as single agents [20,253]. Satisfactory results in terms of response rate have been observed when PD-1-blocking mAbs were used, alone or in association with ibrutinib, in CLL patients, especially those with high levels of PD-L1 and PD-1 in the tumor microenvironment [20,253], undergoing Richter’s syndrome, a CLL transformation to aggressive lymphoma, mainly occurring as diffuse large B cell lymphoma [254].
Other immune checkpoints expressed on NK cells include inhibitory KIRs, CD94/NKG2A, ILT2/CD85j and LAG-3. The use of anti-KIRs or anti-CD94/NKG2A blocking mAbs recapitulates the condition of “missing-self” recognition, thus restoring NK cell-mediated antitumor responses [249]. The clinical relevance of KIR inhibition has been already shown in allogeneic haplo-mismatched stem cell transplantation in patients with AML [29,30]. Mismatches between KIRs on donor NK cells and recipient HLA-class I molecules enable NK cell activation, which is associated with improved relapse-free and overall survival, suggesting that in the absence of KIR interactions with HLA-class I molecules, alloreactive NK cells may eradicate residual leukemia [29,30].
In CLL, the fully human IgG4 mAb lirilumab (IPH2102), directed against a common epitope shared by KIR2DL1/2/3, has been evaluated. A first-in-human phase 1 study using this mAb has identified the doses able to fully saturate KIRs without deleterious clinical, hematological or immunological effects and has shown that a prolonged KIR blockade is safe and well tolerated in patients with CLL [255]. Additionally, the evidence that CLL cells overexpress HLA-E, the main ligand for CD94/NKG2A, has provided the rationale for using the humanized IgG4 anti-CD94/NKG2A mAb monalizumab (IPH2201) for CLL treatment [138] (Figure 3B). Preclinical studies have shown that monalizumab is able to restore direct cytotoxicity of CLL-derived NK cells against HLA-E-expressing targets, without impacting NK cell-mediated ADCC [174]. In vitro studies have shown that even mAbs blocking ILT2/CD85j, highly expressed in CLL-derived NK cells, are able in combination with the immunomodulatory drug lenalidomide to restore NK cell cytotoxicity, resulting in increased elimination of CLL cells [178]. Additionally, although the functional consequences of LAG-3 blockade in CLL have mainly been studied for T cells [256], it has recently been shown that in vitro treatment of CLL cells with the LAG-3-blocking mAb relatlimab (BMS-986016) restores NK cell proliferation and antitumor activity and in combination with lenalidomide significantly increases rituximab-mediated ADCC of NK cells and IL-2 production by T cells [257].

4.3. Allogeneic NK Cell Therapy

Allogeneic hematopoietic stem cell transplantation (allo-HSCT) has long been considered the only curative approach for high-risk patients with CLL, particularly those with relapsed/refractory disease or with TP53 aberration [258]. After BCR and Bcl-2 inhibitors became available and their efficacy in high-risk CLL patients was shown [259,260,261], considerations on the risk–benefit effects of an allo-HSCT in CLL have led to a dramatic decrease in the number of allo-HSCTs performed in both Europe and the United States [262,263,264].
It has been well demonstrated that in haplo-HSCT for high-risk acute myeloid and lymphoid leukemia, donor-derived alloreactive NK cells play a crucial role in the GvL effect due to KIR–HLA mismatches between donor and patient and the consequent lack of NK inhibition by KIR ligands [29,30,265,266]. Conversely, the GvL reaction in allo-HSCT for CLL seems to be mainly mediated by T cells, as indicated by clinical studies showing that increased relapses were observed using T cell-depleting strategies [263,267] and that unrelated donor KIR genotype neither improves GvL reactions nor reduces the incidence of relapse in CLL [268,269]. In keeping with these observations, in vitro studies have shown that allogeneic NK cells, after expansion/activation with an optimized protocol, are able to kill CLL cells independently of KIR–HLA mismatches [270]. However, although allogeneic NK cells do not mediate the GvL effect in a CLL-HSCT transplantation setting, they, unlike T cells, lack the potential to cause graft-versus-host disease (GvHD) [29,30,265,271,272], thus representing appealing and safe candidates for adoptive immunotherapy for CLL.

4.4. CAR-NK Cell Therapy

An emerging strategy of adoptive cell immunotherapy in CLL is based on the transfer of T or NK cells engineered with chimeric antigen receptors (CARs), which are able to recognize a specific antigen on tumor cells and to activate antitumor activity in engineered cells through signal transduction [273,274]. The first CAR-based therapy exploited in CLL consisted in the infusion of autologous, CD19-directed CAR-T cells [275]. Since then, a large body of clinical research has been performed for evaluating the safety and efficacy of this new therapy in patients with refractory/relapsed CLL [276,277,278,279,280]. Concerning the safety profile, CAR-T cells induce in CLL complications similar to those observed in other hematological malignancies, including cytokine release syndrome (CRS) and neurotoxicity [281]. Concerning the efficacy, although the initial trials of CAR-T cells in CLL showed promising results [276,277,278,279,280,282], more recent studies were discouraging because either the response rates or the remission rates were lower in CLL compared with other B cell malignancies [15,16,18,19]. The low efficacy of CAR-T cell therapy in CLL is mainly due to T cell exhaustion and increased T cell terminal differentiation, which hamper the expansion and the antitumor functions of autologous CAR-T cells [250,283,284]. Although this limitation could be overcome by using allogeneic CAR-T cells from healthy donors, this approach is problematic as these cells, even if HLA-matched, carry the risk of GvHD [285]. Recent studies have shown that anti-CLL activity of autologous CAR-T cells is improved when they are administered in combination with the BTK inhibitors ibrutinib or acalabrutinib [286,287] due to their ability to increase T cell number and function in CLL [288].
Given the low performance of CAR-T cells in CLL and their complex and expensive individual-patient-based manufacturing, there is a growing interest in NK cells as an alternative platform for CAR engineering. The possibility of exploiting anti-CD19 CAR-NK cells is appealing for several reasons. First, the risk of CRS or long-term adverse events could be reduced when using NK cells due to their different cytokine profile, short lifespan and low rate of expansion [289,290,291]. Second, CAR-NK cells recognize tumor cells not only through the CAR construct but also through their native receptors, adding ADCC-mediated mechanisms to the CAR-mediated cell killing and reducing the possibility of tumor escape in the case of downregulation of the CAR target antigen [292]. Additionally, NK cells for CAR therapy could be used in an allogeneic setting, because of their ability to contribute to GvL effect without causing GvHD [272]. Finally, allogeneic NK-CAR cells can be generated from multiple sources, including peripheral blood (PB), umbilical cord blood (CB), hematopoietic stem cells, induced pluripotent stem cells (bone marrow) and NK cell lines, thus providing an “off-the-shelf” product, unlike the personalized and patient-specific product that limits current CAR-T cell therapies [273]. There is evidence indicating that both adult PB and umbilical CB are good sources for generating anti-CD19 CAR-NK cells able to kill in vitro primary CLL cells [293]. Even NK-92 cells transfected with CD19-CAR or CD20-CAR, or redirected against different tumor antigens by adapter CAR technology using biotinylated antibodies as adapter molecules, have been shown to induce significant lysis of primary CLL cells and to contrast tumor antigen evasion mechanisms [294,295,296].
However, a major disadvantage of using NK cells for adoptive transfer is their low persistence in the absence of cytokine support [297]. Indeed, whereas this could be helpful in reducing long-term adverse effects and toxicity, it may significantly reduce their clinical efficacy. Interestingly, even the timing of the collection, in vitro expansion and adoptive transfer of autologous NK cells in cancer patients undergoing chemotherapy and PB stem cell transplantation has been shown to be of critical relevance in influencing the clinical efficacy of NK cells [298]. Thus, enhancing NK cell performance has been the subject of active research by many groups, who developed multiple strategies to genetically manipulate NK cells to express cytokines for autocrine proliferation [299,300,301]. In this context, Rezvani’s group generated CAR-CD19+ NK cells that not only persist and mediate an efficient killing of primary CLL cells in vitro but also incorporate safety measures to limit toxicity [301]. Specifically, CB-derived NK cells were transduced with a retroviral vector incorporating the genes for (i) CAR-CD19 to redirect their specificity, (ii) IL-15 to support their survival and proliferation and (iii) inducible caspase-9-based suicide gene (iC9), which can be pharmacologically activated to eliminate transduced cells in the event of unacceptable toxic effect (Figure 3C). Antitumor activity of these iC9/CAR.19/IL-15 CAR-NK cells was also demonstrated in a xenograft Raji lymphoma murine model. Based on these findings, the same authors undertook a phase 1 and 2 trial to assess the safety and efficacy of escalating doses of HLA-mismatched, CB-derived iC9/CAR.19/IL-15 CAR-NK cells for the treatment of relapsed or refractory CD19-positive B-lymphoid malignancies, including CLL [42]. Results showed that of the 11 treated patients, 8 rapidly responded (within 30 days), including 7 (4 with lymphoma and 3 with CLL) who achieved a complete remission without major toxic effects [42]. Altogether, the preclinical and early phase clinical results obtained using CAR-NK cells in CLL are encouraging and support their further development, especially if limitations of the current CAR-T cell therapy remain unsolved.

5. Conclusions

Here we reviewed current knowledge of the NK cell alterations in CLL, the different CLL evasion mechanisms from NK cell-mediated immune surveillance and the potential relevance of using NK cells in CLL immunotherapy. Despite contrasting studies about phenotype, functions and role of NK cells in immune defense against CLL, the most likely opinion is that impairment of NK cell activity is mainly due to escape mechanisms of CLL cells rather than to intrinsic defects in NK cells. This hypothesis is supported by several pieces of in vitro evidence showing that NK cell functions are retained and/or recovered after an appropriate stimulation, leaving hope that NK cells can be ideal candidates for CLL immunotherapy. Nonetheless, anti-CD20 mAbs, such as rituximab, have shown significant clinical responses only in combination with chemotherapeutic or targeted agents, but not as monotherapy, indicating that different mechanisms negatively affect NK cell-mediated ADCC in vivo. Therefore, a better understanding of the mechanisms of resistance and evasion of CLL cells from NK immune surveillance, as well as advances in engineering new therapeutic molecules able to restore NK cell activity, will help to improve the current NK cell-based therapies and to develop additional ones. In this context, different BiKE and TriKE constructs, which exploit the basic concepts of mAbs to retarget NK cells, have been generated. These constructs have shown satisfactory activity in the preclinical setting, holding great therapeutic potential for CLL treatment, but so far, none of them have entered clinical trials. The targeting of immune checkpoint inhibitors for reactivating the NK cell-mediated responses against CLL cells might be an appealing therapeutic strategy. However, as several immune checkpoints are expressed on both NK and T cells, it is difficult to establish how much of the clinical benefit of checkpoint blockade is attributed to NK cells, unless CLL cells have lost HLA-class I molecules. Additionally, the elements of this therapeutic approach, in particular the PD-1-blocking mAbs, failed to show satisfactory clinical response in CLL trials, regardless of whether they were used as single agents or in combination with ibrutinib, unless patients underwent Richter’s transformation. These results suggest that more investigation is needed to explore the potential of immune checkpoint blockade in CLL. However, what is clear, and also observed in other malignancies, is that blocking a single inhibitory receptor on either T or NK cells is unlikely to induce adequate immune responses and achieve a clinical benefit. Strikingly, significant clinical responses have recently been obtained in CLL patients using cord blood-derived CAR-NK cells, encouraging and supporting their further development, especially if limitations of the current CAR-T cell therapy in CLL remain unsolved. Nonetheless, several questions remain to be addressed for CAR-NK cell therapy in both CLL and other malignancies. These include the determination of the best source of NK cells for immunotherapy, the optimal vector system, the most biologically relevant signaling domain for CAR activation and the ideal ex vivo NK cell expansion strategy.
In conclusion, further investigation for optimizing NK cell immunotherapy in CLL is necessary. Considering that an antitumor response is mediated by different effector subpopulations, including NK cells, that cooperate and/or act in a coordinated fashion, it is likely that a multifaceted combination approach is what ultimately will be required to obtain the maximal benefits from the current and future NK cell-based immunotherapy in CLL and other malignancies.

Author Contributions

E.R. conceived and wrote the review. P.S., F.D.F., B.D.P., S.B., V.G., A.M., E.D., C.R., F.M.A., L.R., M.D.I. and E.R. wrote and referenced the review and prepared the figures. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the Associazione Italiana per la Ricerca sul Cancro (AIRC), IG 2018-ID 21352 to P.S.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kipps, T.J.; Stevenson, F.K.; Wu, C.J.; Croce, C.M.; Packham, G.; Wierda, W.G.; O’Brien, S.; Gribben, J.; Rai, K. Chronic lymphocytic leukaemia. Nat. Rev. Dis. Primers 2017, 3, 16096. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Fabbri, G.; Dalla-Favera, R. The molecular pathogenesis of chronic lymphocytic leukaemia. Nat. Rev. Cancer 2016, 16, 145–162. [Google Scholar] [CrossRef]
  3. Puente, X.S.; Jares, P.; Campo, E. Chronic lymphocytic leukemia and mantle cell lymphoma: Crossroads of genetic and microenvironment interactions. Blood 2018, 131, 2283–2296. [Google Scholar] [CrossRef] [PubMed]
  4. Hallek, M. Chronic lymphocytic leukemia: 2020 update on diagnosis, risk stratification and treatment. Am. J. Hematol. 2019, 94, 1266–1287. [Google Scholar] [CrossRef] [Green Version]
  5. Burger, J.A.; O’Brien, S. Evolution of CLL treatment—From chemoimmunotherapy to targeted and individualized therapy. Nat. Rev. Clin. Oncol. 2018, 15, 510–527. [Google Scholar] [CrossRef]
  6. Bond, D.A.; Woyach, J.A. Targeting BTK in CLL: Beyond Ibrutinib. Curr. Hematol. Malig. Rep. 2019, 14, 197–205. [Google Scholar] [CrossRef]
  7. Herling, C.D.; Abedpour, N.; Weiss, J.; Schmitt, A.; Jachimowicz, R.D.; Merkel, O.; Cartolano, M.; Oberbeck, S.; Mayer, P.; Berg, V.; et al. Clonal dynamics towards the development of venetoclax resistance in chronic lymphocytic leukemia. Nat. Commun. 2018, 9, 727. [Google Scholar] [CrossRef]
  8. Jain, N.; Keating, M.; Thompson, P.; Ferrajoli, A.; Burger, J.; Borthakur, G.; Takahashi, K.; Estrov, Z.; Fowler, N.; Kadia, T.; et al. Ibrutinib and Venetoclax for First-Line Treatment of CLL. N. Engl. J. Med. 2019, 380, 2095–2103. [Google Scholar] [CrossRef]
  9. Rogers, K.A.; Huang, Y.; Ruppert, A.S.; Abruzzo, L.V.; Andersen, B.L.; Awan, F.T.; Bhat, S.A.; Dean, A.; Lucas, M.; Banks, C.; et al. Phase II Study of Combination Obinutuzumab, Ibrutinib, and Venetoclax in Treatment-Naïve and Relapsed or Refractory Chronic Lymphocytic Leukemia. J. Clin. Oncol. 2020, 38, 3626–3637. [Google Scholar] [CrossRef]
  10. Hillmen, P.; Rawstron, A.C.; Brock, K.; Muñoz-Vicente, S.; Yates, F.J.; Bishop, R.; Boucher, R.; MacDonald, D.; Fegan, C.; McCaig, A.; et al. Ibrutinib Plus Venetoclax in Relapsed/Refractory Chronic Lymphocytic Leukemia: The CLARITY Study. J. Clin. Oncol. 2019, 37, 2722–2729. [Google Scholar] [CrossRef]
  11. Arruga, F.; Gyau, B.B.; Iannello, A.; Vitale, N.; Vaisitti, T.; Deaglio, S. Immune Response Dysfunction in Chronic Lymphocytic Leukemia: Dissecting Molecular Mechanisms and Microenvironmental Conditions. Int. J. Mol. Sci. 2020, 21, 1825. [Google Scholar] [CrossRef] [Green Version]
  12. Forconi, F.; Moss, P. Perturbation of the normal immune system in patients with CLL. Blood 2015, 126, 573–581. [Google Scholar] [CrossRef] [Green Version]
  13. Vlachonikola, E.; Stamatopoulos, K.; Chatzidimitriou, A. T Cells in Chronic Lymphocytic Leukemia: A Two-Edged Sword. Front. Immunol. 2021, 11, 612244. [Google Scholar] [CrossRef]
  14. Roessner, P.M.; Seiffert, M. T-cells in chronic lymphocytic leukemia: Guardians or drivers of disease? Leukemia 2020, 34, 2012–2024. [Google Scholar] [CrossRef]
  15. Park, J.H.; Rivière, I.; Gonen, M.; Wang, X.; Sénéchal, B.; Curran, K.J.; Sauter, C.; Wang, Y.; Santomasso, B.; Mead, E.; et al. Long-Term Follow-up of CD19 CAR Therapy in Acute Lymphoblastic Leukemia. N. Engl. J. Med. 2018, 378, 449–459. [Google Scholar] [CrossRef]
  16. Schuster, S.J.; Svoboda, J.; Chong, E.A.; Nasta, S.D.; Mato, A.R.; Anak, Ö.; Brogdon, J.L.; Pruteanu-Malinici, I.; Bhoj, V.; Landsburg, D.; et al. Chimeric Antigen Receptor T Cells in Refractory B-Cell Lymphomas. N. Engl. J. Med. 2017, 377, 2545–2554. [Google Scholar] [CrossRef]
  17. Xu-Monette, Z.Y.; Zhou, J.; Young, K.H. PD-1 expression and clinical PD-1 blockade in B-cell lymphomas. Blood 2018, 131, 68–83. [Google Scholar] [CrossRef] [Green Version]
  18. Lorentzen, C.L.; Straten, P.T. CD19-Chimeric Antigen Receptor T Cells for Treatment of Chronic Lymphocytic Leukaemia and Acute Lymphoblastic Leukaemia. Scand. J. Immunol. 2015, 82, 307–319. [Google Scholar] [CrossRef]
  19. Fraietta, J.A.; Lacey, S.F.; Orlando, E.J.; Pruteanu-Malinici, I.; Gohil, M.; Lundh, S.; Boesteanu, A.C.; Wang, Y.; O’Connor, R.S.; Hwang, W.T.; et al. Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia. Nat. Med. 2018, 24, 563–571. [Google Scholar] [CrossRef]
  20. Ding, W.; LaPlant, B.R.; Call, T.G.; Parikh, S.A.; Leis, J.F.; He, R.; Shanafelt, T.D.; Sinha, S.; Le-Rademacher, J.; Feldman, A.L.; et al. Pembrolizumab in patients with CLL and Richter transformation or with relapsed CLL. Blood 2017, 129, 3419–3427. [Google Scholar] [CrossRef]
  21. Malmberg, K.J.; Carlsten, M.; Björklund, A.; Sohlberg, E.; Bryceson, Y.T.; Ljunggren, H.G. Natural killer cell-mediated immunosurveillance of human cancer. Semin. Immunol. 2017, 31, 20–29. [Google Scholar] [CrossRef]
  22. Perera Molligoda Arachchige, A.S. Human NK cells: From development to effector functions. Innate Immun. 2021, 27, 212–229. [Google Scholar] [CrossRef]
  23. Sivori, S.; Vacca, P.; Del Zotto, G.; Munari, E.; Mingari, M.C.; Moretta, L. Human NK cells: Surface receptors, inhibitory checkpoints, and translational applications. Cell. Mol. Immunol. 2019, 16, 430–441. [Google Scholar] [CrossRef]
  24. Ochoa, M.C.; Minute, L.; Rodriguez, I.; Garasa, S.; Perez-Ruiz, E.; Inogés, S.; Melero, I.; Berraondo, P. Antibody-dependent cell cytotoxicity: Immunotherapy strategies enhancing effector NK cells. Immunol. Cell Biol. 2017, 95, 347–355. [Google Scholar] [CrossRef]
  25. Shanehbandi, D.; Majidi, J.; Kazemi, T.; Baradaran, B.; Aghebati-Maleki, L. CD20-based Immunotherapy of B-cell Derived Hematologic Malignancies. Curr. Cancer Drug Targets 2017, 17, 423–444. [Google Scholar] [CrossRef]
  26. Marshall, M.; Stopforth, R.J.; Cragg, M.S. Therapeutic Antibodies: What Have We Learnt from Targeting CD20 and Where Are We Going? Front. Immunol. 2017, 8, 1245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Battella, S.; Cox, M.C.; Santoni, A.; Palmieri, G. Natural killer (NK) cells and anti-tumor therapeutic mAb: Unexplored interactions. J. Leukoc. Biol. 2016, 99, 87–96. [Google Scholar] [CrossRef] [Green Version]
  28. Jaglowski, S.M.; Alinari, L.; Lapalombella, R.; Muthusamy, N.; Byrd, J.C. The clinical application of monoclonal antibodies in chronic lymphocytic leukemia. Blood 2010, 116, 3705–3714. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Ruggeri, L.; Capanni, M.; Urbani, E.; Perruccio, K.; Shlomchik, W.D.; Tosti, A.; Posati, S.; Rogaia, D.; Frassoni, F.; Aversa, F.; et al. Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants. Science 2002, 295, 2097–2100. [Google Scholar] [CrossRef] [Green Version]
  30. Ruggeri, L.; Aversa, F.; Martelli, M.F.; Velardi, A. Allogeneic hematopoietic transplantation and natural killer cell recognition of missing self. Immunol. Rev. 2006, 214, 202–218. [Google Scholar] [CrossRef]
  31. Ziegler, H.W.; Kay, N.E.; Zarling, J.M. Deficiency of natural killer cell activity in patients with chronic lymphocytic leukemia. Int. J. Cancer 1981, 27, 321–327. [Google Scholar] [CrossRef]
  32. Kay, N.E.; Zarling, J.M. Impaired natural killer activity in patients with chronic lymphocytic leukemia is associated with a deficiency of azurophilic cytoplasmic granules in putative NK cells. Blood 1984, 63, 305–309. [Google Scholar] [CrossRef] [Green Version]
  33. Katrinakis, G.; Kyriakou, D.; Papadaki, H.; Kalokyri, I.; Markidou, F.; Eliopoulos, G.D. Defective natural killer cell activity in B-cell chronic lymphocytic leukaemia is associated with impaired release of natural killer cytotoxic factor(s) but not of tumour necrosis factor-alpha. Acta Haematol. 1996, 96, 16–23. [Google Scholar] [CrossRef]
  34. Le Garff-Tavernier, M.; Decocq, J.; de Romeuf, C.; Parizot, C.; Dutertre, C.A.; Chapiro, E.; Davi, F.; Debré, P.; Prost, J.F.; Teillaud, J.L.; et al. Analysis of CD16+CD56dim NK cells from CLL patients: Evidence supporting a therapeutic strategy with optimized anti-CD20 monoclonal antibodies. Leukemia 2011, 25, 101–109. [Google Scholar] [CrossRef] [Green Version]
  35. Costello, R.T.; Knoblauch, B.; Sanchez, C.; Mercier, D.; Le Treut, T.; Sébahoun, G. Expression of natural killer cell activating receptors in patients with chronic lymphocytic leukaemia. Immunology 2012, 135, 151–157. [Google Scholar] [CrossRef]
  36. Kay, N.E.; Zarling, J. Restoration of impaired natural killer cell activity of B-chronic lymphocytic leukemia patients by recombinant interleukin-2. Am. J. Hematol. 1987, 24, 161–167. [Google Scholar] [CrossRef]
  37. Veuillen, C.; Aurran-Schleinitz, T.; Castellano, R.; Rey, J.; Mallet, F.; Orlanducci, F.; Pouyet, L.; Just-Landi, S.; Coso, D.; Ivanov, V.; et al. Primary B-CLL resistance to NK cell cytotoxicity can be overcome in vitro and in vivo by priming NK cells and monoclonal antibody therapy. J. Clin. Immunol. 2012, 32, 632–646. [Google Scholar] [CrossRef]
  38. Guven, H.; Gilljam, M.; Chambers, B.J.; Ljunggren, H.G.; Christensson, B.; Kimby, E.; Dilber, M.S. Expansion of natural killer (NK) and natural killer-like T (NKT)-cell populations derived from patients with B-chronic lymphocytic leukemia (B-CLL): A potential source for cellular immunotherapy. Leukemia 2003, 17, 1973–1980. [Google Scholar] [CrossRef]
  39. Hofland, T.; Endstra, S.; Gomes, C.; de Boer, R.; de Weerdt, I.; Bobkov, V.; Riedl, J.A.; Heukers, R.; Smit, M.J.; Eldering, E.; et al. Natural Killer Cell Hypo-responsiveness in Chronic Lymphocytic Leukemia can be Circumvented In Vitro by Adequate Activating Signaling. Hemasphere 2019, 3, e308. [Google Scholar] [CrossRef]
  40. Laprevotte, E.; Voisin, G.; Ysebaert, L.; Klein, C.; Daugrois, C.; Laurent, G.; Fournie, J.J.; Quillet-Mary, A. Recombinant human IL-15 trans-presentation by B leukemic cells from chronic lymphocytic leukemia induces autologous NK cell proliferation leading to improved anti-CD20 immunotherapy. J. Immunol. 2013, 191, 3634–3640. [Google Scholar] [CrossRef] [Green Version]
  41. Hemati, M.; Rasouli Nejad, Z.; Shokri, M.R.; Ghahremanfard, F.; Mir Mohammadkhani, M.; Kokhaei, P. IL-27 impact on NK cells activity: Implication for a robust anti-tumor response in chronic lymphocytic leukemia. Int. Immunopharmacol. 2020, 82, 106350. [Google Scholar] [CrossRef]
  42. 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]
  43. Lam, V.C.; Lanier, L.L. NK cells in host responses to viral infections. Curr. Opin. Immunol. 2017, 44, 43–51. [Google Scholar] [CrossRef] [Green Version]
  44. Artis, D.; Spits, H. The biology of innate lymphoid cells. Nature 2015, 517, 293–301. [Google Scholar] [CrossRef]
  45. Spits, H.; Bernink, J.H.; Lanier, L. NK cells and type 1 innate lymphoid cells: Partners in host disease. Nat. Immunol. 2016, 17, 758–764. [Google Scholar] [CrossRef]
  46. Lanier, L.L. Up on the tightrope: Natural killer cell activation and inhibition. Nat. Immunol. 2008, 9, 495–502. [Google Scholar] [CrossRef]
  47. Kumar, S. Natural killer cell cytotoxicity and its regulation by inhibitory receptors. Immunology 2018, 154, 383–393. [Google Scholar] [CrossRef]
  48. Anfossi, N.; André, P.; Guia, S.; Falk, C.S.; Roetynck, S.; Stewart, C.A.; Breso, V.; Frassati, C.; Reviron, D.; Middleton, D.; et al. Human NK cell education by inhibitory receptors for MHC class I. Immunity 2006, 25, 331–342. [Google Scholar] [CrossRef]
  49. Kärre, K. Natural killer cell recognition of missing self. Nat. Immunol. 2008, 9, 477–480. [Google Scholar] [CrossRef]
  50. Parikh, B.A.; Bern, M.D.; Piersma, S.J.; Yang, L.; Beckman, D.L.; Poursine-Laurent, J.; Plougastel-Douglas, B.; Yokoyama, W.M. Control of Viral Infection by Natural Killer Cell Inhibitory Receptors. Cell Rep. 2020, 32, 107969. [Google Scholar] [CrossRef] [PubMed]
  51. Xu, Y.; Wang, L.; Li, W.; Chen, B.; Liu, Y.; Wang, H.; Zhao, S.; Ye, L.; He, Y.; Zhou, C. Killer immunoglobulin-like receptors/human leukocyte antigen class-I, a crucial immune pathway in cancer. Ann. Transl. Med. 2020, 8, 244. [Google Scholar] [CrossRef] [PubMed]
  52. Quatrini, L.; Chiesa, M.D.; Sivori, S.; Mingari, M.C.; Pende, D.; Moretta, L. Human NK cells, their receptors and function. Eur. J. Immunol. 2021, in press. [Google Scholar] [CrossRef] [PubMed]
  53. Voskoboinik, I.; Smyth, M.J.; Trapani, J.A. Perforin-mediated target-cell death and immune homeostasis. Nat. Rev. Immunol. 2006, 6, 940–952. [Google Scholar] [CrossRef]
  54. 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 2010, 115, 2167–2176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Zhu, Y.; Huang, B.; Shi, J. Fas ligand and lytic granule differentially control cytotoxic dynamics of natural killer cell against cancer target. Oncotarget 2016, 7, 47163–47172. [Google Scholar] [CrossRef] [Green Version]
  56. Agaugué, S.; Marcenaro, E.; Ferranti, B.; Moretta, L.; Moretta, A. Human natural killer cells exposed to IL-2, IL-12, IL-18, or IL-4 differently modulate priming of naive T cells by monocyte-derived dendritic cells. Blood 2008, 112, 1776–1783. [Google Scholar] [CrossRef] [Green Version]
  57. Wu, Y.; Tian, Z.; Wei, H. Developmental and Functional Control of Natural Killer Cells by Cytokines. Front. Immunol. 2017, 8, 930. [Google Scholar] [CrossRef] [PubMed]
  58. 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]
  59. Floros, T.; Tarhini, A.A. Anticancer Cytokines: Biology and Clinical Effects of Interferon-α2, Interleukin (IL)-2, IL-15, IL-21, and IL-12. Semin. Oncol. 2015, 42, 539–548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Regis, S.; Dondero, A.; Caliendo, F.; Bottino, C.; Castriconi, R. NK Cell Function Regulation by TGF-β-Induced Epigenetic Mechanisms. Front. Immunol. 2020, 11, 311. [Google Scholar] [CrossRef] [Green Version]
  61. Scoville, S.D.; Freud, A.G.; Caligiuri, M.A. Cellular pathways in the development of human and murine innate lymphoid cells. Curr. Opin. Immunol. 2019, 56, 100–106. [Google Scholar] [CrossRef]
  62. Kennedy, M.K.; Glaccum, M.; Brown, S.N.; Butz, E.A.; Viney, J.L.; Embers, M.; Matsuki, N.; Charrier, K.; Sedger, L.; Willis, C.R.; et al. Reversible defects in natural killer and memory CD8 T cell lineages in interleukin 15-deficient mice. J. Exp. Med. 2000, 191, 771–780. [Google Scholar] [CrossRef] [Green Version]
  63. Lodolce, J.P.; Boone, D.L.; Chai, S.; Swain, R.E.; Dassopoulos, T.; Trettin, S.; Ma, A. IL-15 receptor maintains lymphoid homeostasis by supporting lymphocyte homing and proliferation. Immunity 1998, 9, 669–676. [Google Scholar] [CrossRef] [Green Version]
  64. 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]
  65. 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] [PubMed]
  66. Orr, M.T.; Lanier, L.L. Natural killer cell education and tolerance. Cell 2010, 142, 847–856. [Google Scholar] [CrossRef] [Green Version]
  67. Geary, C.D.; Sun, J.C. Memory responses of natural killer cells. Semin. Immunol. 2017, 31, 11–19. [Google Scholar] [CrossRef]
  68. Freud, A.G.; Mundy-Bosse, B.L.; Yu, J.; Caligiuri, M.A. The Broad Spectrum of Human Natural Killer Cell Diversity. Immunity 2017, 47, 820–833. [Google Scholar] [CrossRef] [Green Version]
  69. Stabile, H.; Fionda, C.; Gismondi, A.; Santoni, A. Role of Distinct Natural Killer Cell Subsets in Anticancer Response. Front. Immunol. 2017, 8, 293. [Google Scholar] [CrossRef] [Green Version]
  70. Cichicki, F.; Schlums, H.; Theorell, J.; Tesi, B.; Miller, J.S.; Ljunggren, H.G.; Bryceson, Y.T. Diversification and Functional Specialization of Human NK Cell Subsets. Curr. Top Microbiol. Immunol. 2016, 395, 63–94. [Google Scholar] [CrossRef] [PubMed]
  71. Colonna, M. Innate Lymphoid Cells: Diversity, Plasticity, and Unique Functions in Immunity. Immunity 2018, 48, 1104–1117. [Google Scholar] [CrossRef] [Green Version]
  72. Gasteiger, G.; Fan, X.; Dikiy, S.; Lee, S.Y.; Rudensky, A.Y. Tissue residency of innate lymphoid cells in lymphoid and nonlymphoid organs. Science 2015, 35, 981–985. [Google Scholar] [CrossRef] [Green Version]
  73. 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]
  74. Pikovskaya, O.; Chaix, J.; Rothman, N.J.; Collins, A.; Chen, Y.H.; Scipioni, A.M.; Vivier, E.; Reiner, S.L. Cutting edge: Eomesodermin is sufficient to direct type 1 innate lymphocyte development into the conventional NK lineage. J. Immunol. 2016, 196, 1449–1454. [Google Scholar] [CrossRef]
  75. Mackay, L.K.; Minnich, M.; Kragten, N.A.; Liao, Y.; Nota, B.; Seillet, C.; Zaid, A.; Man, K.; Preston, S.; Freestone, D.; et al. Hobit and Blimp1 instruct a universal transcriptional program of tissue residency in lymphocytes. Science 2016, 352, 459–463. [Google Scholar] [CrossRef] [Green Version]
  76. Filipovic, I.; Chiossone, L.; Vacca, P.; Hamilton, R.S.; Ingegnere, T.; Doisne, J.M.; Hawkes, D.A.; Mingari, M.C.; Sharkey, A.M.; Moretta, L.; et al. Molecular definition of group 1 innate lymphoid cells in the mouse uterus. Nat. Commun. 2018, 9, 4492. [Google Scholar] [CrossRef] [Green Version]
  77. Constantinides, M.G.; McDonald, B.D.; Verhoef, P.A.; Bendelac, A. A committed precursor to innate lymphoid cells. Nature 2014, 508, 397–401. [Google Scholar] [CrossRef] [Green Version]
  78. Klose, C.S.N.; Flach, M.; Mohle, 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] [PubMed] [Green Version]
  79. Xu, W.; Cherrier, D.E.; Chea, S.; Vosshenrich, C.; Serafini, N.; Petit, M.; Liu, P.; Golub, R.; Di Santo, J.P. An Id2RFP-Reporter Mouse Redefines Innate Lymphoid Cell Precursor Potentials. Immunity 2019, 50, 1054–1068.e3. [Google Scholar] [CrossRef] [Green Version]
  80. Bald, T.; Wagner, M.; Gao, Y.; Koyasu, S.; Smyth, M.J. Hide and seek: Plasticity of innate lymphoid cells in cancer. Semin. Immunol. 2019, 41, 101273. [Google Scholar] [CrossRef]
  81. Gao, Y.; Souza-Fonseca-Guimaraes, F.; Bald, T.; Foong, N.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]
  82. McFarland, A.P.; Yalin, A.; Wang, S.Y.; Cortez, V.S.; Landsberger, T.; Sudan, R.; Peng, V.; Miller, H.L.; Ricci, B.; David, E.; et al. Multi-tissue single-cell analysis deconstructs the complex programs of mouse natural killer and type 1 innate lymphoid cells in tissues and circulation. Immunity 2021, 54, 1320–1337.e4. [Google Scholar] [CrossRef] [PubMed]
  83. Orange, J.S. Natural killer cell deficiency. J. Allergy Clin. Immunol. 2013, 132, 515–525. [Google Scholar] [CrossRef] [Green Version]
  84. Chijioke, O.; Müller, A.; Feederle, R.; Barros, M.H.; Krieg, C.; Emmel, V.; Marcenaro, E.; Leung, C.S.; Antsiferova, O.; Landtwing, V.; et al. Human natural killer cells prevent infectious mononucleosis features by targeting lytic Epstein-Barr virus infection. Cell Rep. 2013, 5, 1489–1498. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Dai, H.S.; Caligiuri, M.A. Molecular Basis for the Recognition of Herpes Simplex Virus Type 1 Infection by Human Natural Killer Cells. Front. Immunol. 2018, 9, 183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Jost, S.; Altfeld, M. Control of human viral infections by natural killer cells. Annu. Rev. Immunol. 2013, 31, 163–194. [Google Scholar] [CrossRef] [PubMed]
  87. Foley, B.; Cooley, S.; Verneris, M.R.; Pitt, M.; Curtsinger, J.; Luo, X.; Lopez-Vergès, S.; Lanier, L.L.; Weisdorf, D.; Miller, J.S. Cytomegalovirus reactivation after allogeneic transplantation promotes a lasting increase in educated NKG2C+ natural killer cells with potent function. Blood 2012, 119, 2665–2674. [Google Scholar] [CrossRef] [PubMed]
  88. Kiessling, R.; Klein, E.; Pross, H.; Wigzell, H. “Natural” killer cells in the mouse. II. Cytotoxic cells with specificity for mouse Moloney leukemia cells. Characteristics of the killer cell. Eur. J. Immunol. 1975, 5, 117–121. [Google Scholar] [CrossRef]
  89. Kärre, K.; Ljunggren, H.G.; Piontek, G.; Kiessling, R. Selective rejection of H-2-deficient lymphoma variants suggests alternative immune defence strategy. Nature 1986, 319, 675–678. [Google Scholar] [CrossRef]
  90. Wu, J.; Lanier, L.L. Natural killer cells and cancer. Adv. Cancer Res. 2003, 90, 127–156. [Google Scholar] [CrossRef]
  91. Guillerey, C.; Smyth, M.J. NK Cells and Cancer Immunoediting. Curr. Top. Microbiol. Immunol. 2016, 395, 115–145. [Google Scholar] [CrossRef] [PubMed]
  92. Marcus, A.; Gowen, B.G.; Thompson, T.W.; Iannello, A.; Ardolino, M.; Deng, W.; Wang, L.; Shifrin, N.; Raulet, D.H. Recognition of tumors by the innate immune system and natural killer cells. Adv. Immunol. 2014, 122, 91–128. [Google Scholar] [CrossRef] [Green Version]
  93. Guerra, N.; Tan, Y.X.; Joncker, N.T.; Choy, A.; Gallardo, F.; Xiong, N.; Knoblaugh, S.; Cado, D.; Greenberg, N.M.; Raulet, D.H. NKG2D-deficient mice are defective in tumor surveillance in models of spontaneous malignancy. Immunity 2008, 28, 571–580. [Google Scholar] [CrossRef] [Green Version]
  94. Iguchi-Manaka, A.; Kai, H.; Yamashita, Y.; Shibata, K.; Tahara-Hanaoka, S.; Honda, S.; Yasui, T.; Kikutani, H.; Shibuya, K.; & Shibuya, A. Accelerated tumor growth in mice deficient in DNAM-1 receptor. J. Exp. Med. 2008, 205, 2959–2964. [Google Scholar] [CrossRef] [Green Version]
  95. Imai, K.; Matsuyama, S.; Miyake, S.; Suga, K.; Nakachi, K. Natural cytotoxic activity of peripheral-blood lymphocytes and cancer incidence: An 11-year follow-up study of a general population. Lancet 2000, 356, 1795–1799. [Google Scholar] [CrossRef]
  96. Zhang, S.; Liu, W.; Hu, B.; Wang, P.; Lv, X.; Chen, S.; Shao, Z. Prognostic Significance of Tumor-Infiltrating Natural Killer Cells in Solid Tumors: A Systematic Review and Meta-Analysis. Front. Immunol. 2020, 11, 1242. [Google Scholar] [CrossRef]
  97. Mastaglio, S.; Wong, E.; Perera, T.; Ripley, J.; Blombery, P.; Smyth, M.J.; Koldej, R.; Ritchie, D. Natural killer receptor ligand expression on acute myeloid leukemia impacts survival and relapse after chemotherapy. Blood Adv. 2018, 2, 335–346. [Google Scholar] [CrossRef] [Green Version]
  98. Luna, J.I.; Grossenbacher, S.K.; Murphy, W.J.; Canter, R.J. Targeting Cancer Stem Cells with Natural Killer Cell Immunotherapy. Expert Opin. Biol. Ther. 2017, 17, 313–324. [Google Scholar] [CrossRef] [Green Version]
  99. Charap, A.J.; Enokida, T.; Brody, R.; Sfakianos, J.; Miles, B.; Bhardwaj, N.; Horowitz, A. Landscape of natural killer cell activity in head and neck squamous cell carcinoma. J. Immunother. Cancer. 2020, 8, e001523. [Google Scholar] [CrossRef]
  100. Melaiu, O.; Lucarini, V.; Cifaldi, L.; Fruci, D. Influence of the Tumor Microenvironment on NK Cell Function in Solid Tumors. Front. Immunol. 2020, 10, 3038. [Google Scholar] [CrossRef]
  101. Fauriat, C.; Just-Landi, S.; Mallet, F.; Arnoulet, C.; Sainty, D.; Olive, D.; Costello, R.T. Deficient expression of NCR in NK cells from acute myeloid leukemia: Evolution during leukemia treatment and impact of leukemia cells in NCRdull phenotype induction. Blood 2007, 109, 323–330. [Google Scholar] [CrossRef]
  102. Carlsten, M.; Baumann, B.C.; Simonsson, M.; Jädersten, M.; Forsblom, A.M.; Hammarstedt, C.; Bryceson, Y.T.; Ljunggren, H.G.; Hellström-Lindberg, E.; Malmberg, K.J. Reduced DNAM-1 expression on bone marrow NK cells associated with impaired killing of CD34+ blasts in myelodysplastic syndrome. Leukemia 2010, 24, 1607–1616. [Google Scholar] [CrossRef]
  103. Liu, S.; Galat, V.; Galat, Y.; Lee, Y.; Wainwright, D.; Wu, J. NK cell-based cancer immunotherapy: From basic biology to clinical development. J. Hematol. Oncol. 2021, 14, 7. [Google Scholar] [CrossRef]
  104. Barrow, A.D.; Martin, C.J.; Colonna, M. The Natural Cytotoxicity Receptors in Health and Disease. Front. Immunol. 2019, 10, 909. [Google Scholar] [CrossRef] [Green Version]
  105. Hecht, M.L.; Rosental, B.; Horlacher, T.; Hershkovitz, O.; De Paz, J.L.; Noti, C.; Schauer, S.; Porgador, A.; Seeberger, P.H. Natural cytotoxicity receptors NKp30, NKp44 and NKp46 bind to different heparan sulfate/heparin sequences. J. Proteome Res. 2009, 8, 712–720. [Google Scholar] [CrossRef]
  106. Luczo, J.M.; Ronzulli, S.L.; Tompkins, S.M. Influenza A Virus Hemagglutinin and Other Pathogen Glycoprotein Interactions with NK Cell Natural Cytotoxicity Receptors NKp46, NKp44, and NKp30. Viruses 2021, 13, 156. [Google Scholar] [CrossRef]
  107. Brandt, C.S.; Baratin, M.; Yi, E.C.; Kennedy, J.; Gao, Z.; Fox, B.; Haldeman, B.; Ostrander, C.D.; Kaifu, T.; Chabannon, C.; et al. The B7 family member B7-H6 is a tumor cell ligand for the activating natural killer cell receptor NKp30 in humans. J. Exp. Med. 2009, 206, 1495–1503. [Google Scholar] [CrossRef] [Green Version]
  108. Daßler-Plenker, J.; Reiners, K.S.; van den Boorn, J.G.; Hansen, H.P.; Putschli, B.; Barnert, S.; Schuberth-Wagner, C.; Schubert, R.; Tüting, T.; Hallek, M.; et al. RIG-I activation induces the release of extracellular vesicles with antitumor activity. Oncoimmunology 2016, 5, e1219827. [Google Scholar] [CrossRef]
  109. Binici, J.; Hartmann, J.; Herrmann, J.; Schreiber, C.; Beyer, S.; Güler, G.; Vogel, V.; Tumulka, F.; Abele, R.; Mäntele, W.; et al. A soluble fragment of the tumor antigen BCL2-associated athanogene 6 (BAG-6) is essential and sufficient for inhibition of NKp30 receptor-dependent cytotoxicity of natural killer cells. J. Biol. Chem. 2013, 288, 34295–34303. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Baychelier, F.; Sennepin, A.; Ermonval, M.; Dorgham, K.; Debré, P.; Vieillard, V. Identification of a cellular ligand for the natural cytotoxicity receptor NKp44. Blood 2013, 122, 2935–2942. [Google Scholar] [CrossRef] [Green Version]
  111. Rosental, B.; Brusilovsky, M.; Hadad, U.; Oz, D.; Appel, M.Y.; Afergan, F.; Yossef, R.; Rosenberg, L.A.; Aharoni, A.; Cerwenka, A.; et al. Proliferating cell nuclear antigen is a novel inhibitory ligand for the natural cytotoxicity receptor NKp44. J. Immunol. 2011, 187, 5693–5702. [Google Scholar] [CrossRef]
  112. Barrow, A.D.; Edeling, M.A.; Trifonov, V.; Luo, J.; Goyal, P.; Bohl, B.; Bando, J.K.; Kim, A.H.; Walker, J.; Andahazy, M.; et al. Natural Killer Cells Control Tumor Growth by Sensing a Growth Factor. Cell 2018, 172, 534–548. [Google Scholar] [CrossRef] [Green Version]
  113. Gaggero, S.; Bruschi, M.; Petretto, A.; Parodi, M.; Del Zotto, G.; Lavarello, C.; Prato, C.; Santucci, L.; Barbuto, A.; Bottino, C.; et al. Nidogen-1 is a novel extracellular ligand for the NKp44 activating receptor. Oncoimmunology 2018, 7, e1470730. [Google Scholar] [CrossRef]
  114. Narni-Mancinelli, E.; Gauthier, L.; Baratin, M.; Guia, S.; Fenis, A.; Deghmane, A.E.; Rossi, B.; Fourquet, P.; Escalière, B.; Kerdiles, Y.M.; et al. Complement factor P is a ligand for the natural killer cell-activating receptor NKp46. Sci. Immunol. 2017, 2, eaam9628. [Google Scholar] [CrossRef] [Green Version]
  115. Nevins, A.M.; Subramanian, A.; Tapia, J.L.; Delgado, D.P.; Tyler, R.C.; Jensen, D.R.; Ouellette, A.J.; Volkman, B.F. A requirement for metamorphic interconversion in the antimicrobial activity of chemokine XCL1. Biochemistry 2016, 55, 3784–3793. [Google Scholar] [CrossRef]
  116. Alexandre, Y.O.; Ghilas, S.; Sanchez, C.; Le Bon, A.; Crozat, K.; Dalod, M. XCR1+ dendritic cells promote memory CD8+ T cell recall upon secondary infections with Listeria monocytogenes or certain viruses. J. Exp. Med. 2016, 213, 75–92. [Google Scholar] [CrossRef] [Green Version]
  117. Dhar, P.; Wu, J.D. NKG2D and its ligands in cancer. Curr. Opin. Immunol. 2018, 51, 55–61. [Google Scholar] [CrossRef]
  118. Xing, S.; Ferrari de Andrade, L. NKG2D and MICA/B shedding: A ‘tag game’ between NK cells and malignant cells. Clin. Transl. Immunol. 2020, 9, e1230. [Google Scholar] [CrossRef]
  119. Hilpert, J.; Grosse-Hovest, L.; Grünebach, F.; Buechele, C.; Nuebling, T.; Raum, T.; Steinle, A.; Salih, H.R. Comprehensive analysis of NKG2D ligand expression and release in leukemia: Implications for NKG2D-mediated NK cell responses. J. Immunol. 2012, 189, 1360–1371. [Google Scholar] [CrossRef] [Green Version]
  120. Zingoni, A.; Vulpis, E.; Loconte, L.; Santoni, A. NKG2D Ligand Shedding in Response to Stress: Role of ADAM10. Front. Immunol. 2020, 11, 447. [Google Scholar] [CrossRef]
  121. Berhani, O.; Nachmani, D.; Yamin, R.; Schmiedel, D.; Bar-On, Y.; Mandelboim, O. Vigilin Regulates the Expression of the Stress-Induced Ligand MICB by Interacting with Its 5′ Untranslated Region. J. Immunol. 2017, 198, 3662–3670. [Google Scholar] [CrossRef] [Green Version]
  122. Lazarova, M.; Steinle, A. Impairment of NKG2D-Mediated Tumor Immunity by TGF-β. Front. Immunol. 2019, 10, 2689. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Pende, D.; Spaggiari, G.M.; Marcenaro, S.; Martini, S.; Rivera, P.; Capobianco, A.; Falco, M.; Lanino, E.; Pierri, I.; Zambello, R.; et al. Analysis of the receptor-ligand interactions in the natural killer-mediated lysis of freshly isolated myeloid or lymphoblastic leukemias: Evidence for the involvement of the Poliovirus receptor (CD155) and Nectin-2 (CD112). Blood 2005, 105, 2066–2073. [Google Scholar] [CrossRef]
  124. McArdel, S.L.; Terhorst, C.; Sharpe, A.H. Roles of CD48 in regulating immunity and tolerance. Clin. Immunol. 2016, 164, 10–20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Marcenaro, E.; Augugliaro, R.; Falco, M.; Castriconi, R.; Parolini, S.; Sivori, S.; Romeo, E.; Millo, R.; Moretta, L.; Bottino, C.; et al. CD59 is physically and functionally associated with natural cytotoxicity receptors and activates human NK cell-mediated cytotoxicity. Eur. J. Immunol. 2003, 33, 3367–3376. [Google Scholar] [CrossRef]
  126. Bryceson, Y.T.; March, M.E.; Barber, D.F.; Ljunggren, H.G.; Long, E.O. Cytolytic granule polarization and degranulation controlled by different receptors in resting NK cells. J. Exp. Med. 2005, 202, 1001–1012. [Google Scholar] [CrossRef]
  127. Stewart, C.A.; Laugier-Anfossi, F.; Vély, F.; Saulquin, X.; Riedmuller, J.; Tisserant, A.; Gauthier, L.; Romagné, F.; Ferracci, G.; Arosa, F.A.; et al. Recognition of peptide-MHC class I complexes by activating killer immunoglobulin-like receptors. Proc. Natl. Acad. Sci. USA 2005, 102, 13224–13229. [Google Scholar] [CrossRef] [Green Version]
  128. Braud, V.M.; Allan, D.S.; O’Callaghan, C.A.; Söderström, K.; D’Andrea, A.; Ogg, G.S.; Lazetic, S.; Young, N.T.; Bell, J.I.; Phillips, J.H.; et al. HLA-E binds to natural killer cell receptors CD94/NKG2A, B and C. Nature 1998, 391, 795–799. [Google Scholar] [CrossRef]
  129. Blunt, M.D.; Khakoo, S.I. Activating killer cell immunoglobulin-like receptors: Detection, function and therapeutic use. Int. J. Immunogenet. 2020, 47, 1–12. [Google Scholar] [CrossRef] [Green Version]
  130. Pende, D.; Falco, M.; Vitale, M.; Cantoni, C.; Vitale, C.; Munari, E.; Bertaina, A.; Moretta, F.; Del Zotto, G.; Pietra, G.; et al. Killer Ig-Like Receptors (KIRs): Their Role in NK Cell Modulation and Developments Leading to Their Clinical Exploitation. Front. Immunol. 2019, 10, 1179. [Google Scholar] [CrossRef] [Green Version]
  131. Gauthier, M.; Laroye, C.; Bensoussan, D.; Boura, C.; Decot, V. Natural Killer cells and monoclonal antibodies: Two partners for successful antibody dependent cytotoxicity against tumor cells. Crit. Rev. Oncol. Hematol. 2021, 160, 103261. [Google Scholar] [CrossRef]
  132. 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] [Green Version]
  133. Martin, A.M.; Kulski, J.K.; Gaudieri, S.; Witt, C.S.; Freitas, E.M.; Trowsdale, J.; Christiansen, F.T. Comparative genomic analysis, diversity and evolution of two KIR haplotypes A and B. Gene 2004, 335, 121–131. [Google Scholar] [CrossRef] [PubMed]
  134. Parham, P. MHC class I molecules and KIRs in human history, health and survival. Nat. Rev. Immunol. 2005, 5, 201–214. [Google Scholar] [CrossRef]
  135. Horowitz, A.; Djaoud, Z.; Nemat-Gorgani, N.; Blokhuis, J.; Hilton, H.G.; Béziat, V.; Malmberg, K.J.; Norman, P.J.; Guethlein, L.A.; Parham, P. Class I HLA haplotypes form two schools that educate NK cells in different ways. Sci. Immunol. 2016, 1, eaag1672. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Manser, A.R.; Weinhold, S.; Uhrberg, M. Human KIR repertoires: Shaped by genetic diversity and evolution. Immunol. Rev. 2015, 267, 178–196. [Google Scholar] [CrossRef] [PubMed]
  137. Rajagopalan, S.; Long, E.O. KIR2DL4 (CD158d): An activation receptor for HLA-G. Front. Immunol. 2012, 3, 258. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. André, P.; Denis, C.; Soulas, C.; Bourbon-Caillet, C.; Lopez, J.; Arnoux, T.; Bléry, M.; Bonnafous, C.; Gauthier, L.; Morel, A.; et al. Anti-NKG2A mAb Is a Checkpoint Inhibitor that Promotes Anti-tumor Immunity by Unleashing Both T and NK Cells. Cell 2018, 175, 1731–1743. [Google Scholar] [CrossRef] [Green Version]
  139. Colonna, M.; Samaridis, J.; Cella, M.; Angman, L.; Allen, R.L.; O’Callaghan, C.A.; Dunbar, R.; Ogg, G.S.; Cerundolo, V.; Rolink, A. Human myelomonocytic cells express an inhibitory receptor for classical and nonclassical MHC class I molecules. J. Immunol. 1998, 160, 3096–3100. [Google Scholar]
  140. Chiossone, L.; Vienne, M.; Kerdiles, Y.M.; Vivier, E. Natural killer cell immunotherapies against cancer: Checkpoint inhibitors and more. Semin. Immunol. 2017, 31, 55–63. [Google Scholar] [CrossRef]
  141. Pesce, S.; Greppi, M.; Tabellini, G.; Rampinelli, F.; Parolini, S.; Olive, D.; Moretta, L.; Moretta, A.; Marcenaro, E. Identification of a subset of human natural killer cells expressing high levels of programmed death 1: A phenotypic and functional characterization. J. Allergy Clin. Immunol. 2017, 139, 335–346. [Google Scholar] [CrossRef] [Green Version]
  142. Pesce, S.; Greppi, M.; Grossi, F.; Del Zotto, G.; Moretta, L.; Sivori, S.; Genova, C.; Marcenaro, E. PD/1-PD-Ls Checkpoint: Insight on the Potential Role of NK Cells. Front. Immunol. 2019, 10, 1242. [Google Scholar] [CrossRef]
  143. Guan, J.; Wang, R.; Hasan, S.; Tao, L.; Wazir, M.; Jain, A.G.; Zhu, X.; Perkins, S.; Mohamed, S.; Chang, C.C.; et al. Prognostic Significance of the Dynamic Change of Programmed Death-ligand 1 Expression in Patients with Multiple Myeloma. Cureus 2019, 11, e4401. [Google Scholar] [CrossRef] [Green Version]
  144. Dougall, W.C.; Kurtulus, S.; Smyth, M.J.; Anderson, A.C. TIGIT and CD96: New checkpoint receptor targets for cancer immunotherapy. Immunol. Rev. 2017, 276, 112–120. [Google Scholar] [CrossRef] [PubMed]
  145. Zhou, X.M.; Li, W.Q.; Wu, Y.H.; Han, L.; Cao, X.G.; Yang, X.M.; Wang, H.F.; Zhao, W.S.; Zhai, W.J.; Qi, Y.M.; et al. Intrinsic Expression of Immune Checkpoint Molecule TIGIT Could Help Tumor Growth in vivo by Suppressing the Function of NK and CD8(+) T Cells. Front. Immunol. 2018, 9, 2821. [Google Scholar] [CrossRef] [Green Version]
  146. Kandel, S.; Adhikary, P.; Li, G.; Cheng, K. The TIM3/Gal9 signaling pathway: An emerging target for cancer immunotherapy. Cancer Lett. 2021, 510, 67–78. [Google Scholar] [CrossRef] [PubMed]
  147. Ruffo, E.; Wu, R.C.; Bruno, T.C.; Workman, C.J.; Vignali, D.A.A. Lymphocyte-activation gene 3 (LAG3): The next immune checkpoint receptor. Semin. Immunol. 2019, 42, 101305. [Google Scholar] [CrossRef]
  148. Reiners, K.S.; Topolar, D.; Henke, A.; Simhadri, V.R.; Kessler, J.; Sauer, M.; Bessler, M.; Hansen, H.P.; Tawadros, S.; Herling, M.; et al. Soluble ligands for NK cell receptors promote evasion of chronic lymphocytic leukemia cells from NK cell anti-tumor activity. Blood 2013, 121, 3658–3665. [Google Scholar] [CrossRef] [PubMed]
  149. Maki, G.; Hayes, G.M.; Naji, A.; Tyler, T.; Carosella, E.D.; Rouas-Freiss, N.; Gregory, S.A. NK resistance of tumor cells from multiple myeloma and chronic lymphocytic leukemia patients: Implication of HLA-G. Leukemia 2008, 22, 998–1006. [Google Scholar] [CrossRef] [Green Version]
  150. Jewell, A.P.; Worman, C.P.; Giles, F.J.; Goldstone, A.H.; Lydyard, P.M. Resistance of chronic lymphocytic leukaemia cells to interferon-alpha generated lymphokine activated killer cells. Leuk. Lymphoma 1992, 7, 473–480. [Google Scholar] [CrossRef]
  151. Spitz, D.L.; Zucker-Franklin, D.; Nabi, Z.F. Unmasking of cryptic natural killer (NK) cell recognition sites on chronic lymphocytic leukemia lymphocytes. Am. J. Hematol. 1988, 28, 155–161. [Google Scholar] [CrossRef]
  152. Huergo-Zapico, L.; Gonzalez-Rodriguez, A.P.; Contesti, J.; Gonzalez, E.; López-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. Immunother. 2012, 61, 1201–1210. [Google Scholar] [CrossRef]
  153. Eskelund, C.W.; Nederby, L.; Thysen, A.H.; Skovbo, A.; Roug, A.S.; Hokland, M.E. Interleukin-21 and rituximab enhance NK cell functionality in patients with B-cell chronic lymphocytic leukaemia. Leuk. Res. 2011, 35, 914–920. [Google Scholar] [CrossRef]
  154. Le Garff-Tavernier, M.; Herbi, L.; de Romeuf, C.; Nguyen-Khac, F.; Davi, F.; Grelier, A.; Boudjoghra, M.; Maloum, K.; Choquet, S.; Urbain, R.; et al. Antibody-dependent cellular cytotoxicity of the optimized anti-CD20 monoclonal antibody ublituximab on chronic lymphocytic leukemia cells with the 17p deletion. Leukemia 2014, 28, 230–233. [Google Scholar] [CrossRef]
  155. Hofland, T.; Eldering, E.; Kater, A.P.; Tonino, S.H. Engaging Cytotoxic T and NK Cells for Immunotherapy in Chronic Lymphocytic Leukemia. Int. J. Mol. Sci. 2019, 20, 4315. [Google Scholar] [CrossRef] [Green Version]
  156. Demanet, C.; Mulder, A.; Deneys, V.; Worsham, M.J.; Maes, P.; Claas, F.H.; Ferrone, S. Down-regulation of HLA-A and HLA-Bw6, but not HLA-Bw4, allospecificities in leukemic cells: An escape mechanism from CTL and NK attack? Blood 2004, 103, 3122–3130. [Google Scholar] [CrossRef] [Green Version]
  157. Verheyden, S.; Ferrone, S.; Mulder, A.; Claas, F.H.; Schots, R.; De Moerloose, B.; Benoit, Y.; Demanet, C. Role of the inhibitory KIR ligand HLA-Bw4 and HLA-C expression levels in the recognition of leukemic cells by Natural Killer cells. Cancer Immunol. Immunother. 2009, 58, 855–865. [Google Scholar] [CrossRef] [Green Version]
  158. Wang, W.T.; Zhu, H.Y.; Wu, Y.J.; Xia, Y.; Wu, J.Z.; Wu, W.; Liang, J.H.; Wang, L.; Fan, L.; Li, J.Y.; et al. Elevated absolute NK cell counts in peripheral blood predict good prognosis in chronic lymphocytic leukemia. J. Cancer Res. Clin. Oncol. 2018, 144, 449–457. [Google Scholar] [CrossRef]
  159. Gonzalez-Rodriguez, A.P.; Contesti, J.; Huergo-Zapico, L.; Lopez-Soto, A.; Fernández-Guizán, A.; Acebes-Huerta, A.; Gonzalez-Huerta, A.J.; Gonzalez, E.; Fernandez-Alvarez, C.; Gonzalez, S. Prognostic significance of CD8 and CD4 T cells in chronic lymphocytic leukemia. Leuk. Lymphoma 2010, 51, 1829–1836. [Google Scholar] [CrossRef]
  160. Palmer, S.; Hanson, C.A.; Zent, C.S.; Porrata, L.F.; Laplant, B.; Geyer, S.M.; Markovic, S.N.; Call, T.G.; Bowen, D.A.; Jelinek, D.F.; et al. Prognostic importance of T and NK-cells in a consecutive series of newly diagnosed patients with chronic lymphocytic leukaemia. Br. J. Haematol. 2008, 141, 607–614. [Google Scholar] [CrossRef]
  161. Hadadi, L.; Hafezi, M.; Amirzargar, A.A.; Sharifian, R.A.; Abediankenari, S.; Asgarian-Omran, H. Dysregulated Expression of Tim-3 and NKp30 Receptors on NK Cells of Patients with Chronic Lymphocytic Leukemia. Oncol. Res. Treat. 2019, 42, 202–208. [Google Scholar] [CrossRef]
  162. Huergo-Zapico, L.; Acebes-Huerta, A.; Gonzalez-Rodriguez, A.P.; Contesti, J.; Gonzalez-García, E.; Payer, A.R.; Villa-Alvarez, M.; Fernández-Guizán, A.; López-Soto, A.; Gonzalez, S. Expansion of NK cells and reduction of NKG2D expression in chronic lymphocytic leukemia. Correlation with progressive disease. PLoS ONE 2014, 9, e108326. [Google Scholar] [CrossRef]
  163. MacFarlane, A.W., 4th; Jillab, M.; Smith, M.R.; Alpaugh, R.K.; Cole, M.E.; Litwin, S.; Millenson, M.M.; Al-Saleem, T.; Cohen, A.D.; Campbell, K.S. NK cell dysfunction in chronic lymphocytic leukemia is associated with loss of the mature cells expressing inhibitory killer cell Ig-like receptors. Oncoimmunology 2017, 6, e1330235. [Google Scholar] [CrossRef]
  164. Gumá, M.; Angulo, A.; Vilches, C.; Gómez-Lozano, N.; Malats, N.; López-Botet, M. Imprint of human cytomegalovirus infection on the NK cell receptor repertoire. Blood 2004, 104, 3664–3671. [Google Scholar] [CrossRef] [Green Version]
  165. López-Botet, M.; Muntasell, A.; Vilches, C. The CD94/NKG2C+ NK-cell subset on the edge of innate and adaptive immunity to human cytomegalovirus infection. Semin. Immunol. 2014, 26, 145–151. [Google Scholar] [CrossRef]
  166. Parry, H.M.; Stevens, T.; Oldreive, C.; Zadran, B.; McSkeane, T.; Rudzki, Z.; Paneesha, S.; Chadwick, C.; Stankovic, T.; Pratt, G.; et al. NK cell function is markedly impaired in patients with chronic lymphocytic leukaemia but is preserved in patients with small lymphocytic lymphoma. Oncotarget 2016, 7, 68513–68526. [Google Scholar] [CrossRef] [Green Version]
  167. Lotz, M.; Ranheim, E.; Kipps, T.J. Transforming growth factor beta as endogenous growth inhibitor of chronic lymphocytic leukemia B cells. J. Exp. Med. 1994, 179, 999–1004. [Google Scholar] [CrossRef]
  168. Lad, D.; Hoeppli, R.; Huang, Q.; Garcia, R.; Xu, L.; Toze, C.; Broady, R.; Levings, M. Regulatory T-cells drive immune dysfunction in CLL. Leuk. Lymphoma 2018, 59, 486–489. [Google Scholar] [CrossRef]
  169. Kowalska, W.; Bojarska-Junak, A. Monocytic MDSC as a source of immunosuppressive cytokines in chronic lymphocytic leukemia (CLL) microenvironment. Folia Histochem. Cytobiol. 2020, 58, 25–36. [Google Scholar] [CrossRef] [Green Version]
  170. Nückel, H.; Switala, M.; Sellmann, L.; Horn, P.A.; Dürig, J.; Dührsen, U.; Küppers, R.; Grosse-Wilde, H.; Rebmann, V. The prognostic significance of soluble NKG2D ligands in B-cell chronic lymphocytic leukemia. Leukemia 2010, 24, 1152–1159. [Google Scholar] [CrossRef]
  171. Sánchez-Martínez, D.; Lanuza, P.M.; Gómez, N.; Muntasell, A.; Cisneros, E.; Moraru, M.; Azaceta, G.; Anel, A.; Martínez-Lostao, L.; Villalba, M.; et al. Activated Allogeneic NK Cells Preferentially Kill Poor Prognosis B-Cell Chronic Lymphocytic Leukemia Cells. Front. Immunol. 2016, 7, 454. [Google Scholar] [CrossRef]
  172. Puiggros, A.; Blanco, G.; Muntasell, A.; Rodríguez-Rivera, M.; Nonell, L.; Altadill, M.; Puigdecanet, E.; Arnal, M.; Calvo, X.; Gimeno, E.; et al. Reduced expansion of CD94/NKG2C+ NK cells in chronic lymphocytic leukemia and CLL-like monoclonal B-cell lymphocytosis is not related to increased human cytomegalovirus seronegativity or NKG2C deletions. Int. J. Lab. Hematol. 2021, in press. [Google Scholar] [CrossRef]
  173. Petersen, L.; Roug, A.S.; Skovbo, A.; Thysen, A.H.; Eskelund, C.W.; Hokland, M.E. The CD94/NKG2C-expressing NK cell subset is augmented in chronic lymphocytic leukemia patients with positive human cytomegalovirus serostatus. Viral Immunol. 2009, 22, 333–337. [Google Scholar] [CrossRef]
  174. 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] [Green Version]
  175. Wagner, B.; da Silva Nardi, F.; Schramm, S.; Kraemer, T.; Celik, A.A.; Dürig, J.; Horn, P.A.; Dührsen, U.; Nückel, H.; Rebmann, V. HLA-E allelic genotype correlates with HLA-E plasma levels and predicts early progression in chronic lymphocytic leukemia. Cancer 2017, 123, 814–823. [Google Scholar] [CrossRef]
  176. Linet, M.S.; Bias, W.B.; Dorgan, J.F.; McCaffrey, L.D.; Humphrey, R.L. HLA antigens in chronic lymphocytic leukemia. Tissue Antigens 1988, 31, 71–78. [Google Scholar] [CrossRef]
  177. Mueller, L.P.; Machulla, H.K. Increased frequency of homozygosity for HLA class II loci in female patients with chronic lymphocytic leukemia. Leuk. Lymphoma 2002, 43, 1013–1019. [Google Scholar] [CrossRef]
  178. Villa-Álvarez, M.; Lorenzo-Herrero, S.; Gonzalez-Rodriguez, A.P.; López-Soto, A.; Payer, A.R.; Gonzalez-Garcia, E.; Huergo-Zapico, L.; Gonzalez, S. Ig-like transcript 2 (ILT2) suppresses T cell function in chronic lymphocytic leukemia. Oncoimmunology 2017, 6, e1353856. [Google Scholar] [CrossRef]
  179. Własiuk, P.; Tomczak, W.; Zając, M.; Dmoszyńska, A.; Giannopoulos, K. Total expression of HLA-G and TLR-9 in chronic lymphocytic leukemia patients. Hum. Immunol. 2013, 74, 1592–1597. [Google Scholar] [CrossRef]
  180. Nückel, H.; Rebmann, V.; Dürig, J.; Dührsen, U.; Grosse-Wilde, H. HLA-G expression is associated with an unfavorable outcome and immunodeficiency in chronic lymphocytic leukemia. Blood 2005, 105, 1694–1698. [Google Scholar] [CrossRef] [Green Version]
  181. Rizzo, R.; Audrito, V.; Vacca, P.; Rossi, D.; Brusa, D.; Stignani, M.; Bortolotti, D.; D’Arena, G.; Coscia, M.; Laurenti, L.; et al. HLA-G is a component of the chronic lymphocytic leukemia escape repertoire to generate immune suppression: Impact of the HLA-G 14 base pair (rs66554220) polymorphism. Haematologica 2014, 99, 888–896. [Google Scholar] [CrossRef]
  182. Taghiloo, S.; Allahmoradi, E.; Ebadi, R.; Tehrani, M.; Hosseini-Khah, Z.; Janbabaei, G.; Shekarriz, R.; Asgarian-Omran, H. Upregulation of Galectin-9 and PD-L1 Immune Checkpoints Molecules in Patients with Chronic Lymphocytic Leukemia. Asian Pac. J. Cancer Prev. 2017, 18, 2269–2274. [Google Scholar] [CrossRef]
  183. Wdowiak, K.; Gallego-Colon, E.; Francuz, T.; Czajka-Francuz, P.; Ruiz-Agamez, N.; Kubeczko, M.; Grochoła, I.; Wybraniec, M.T.; Chudek, J.; Wojnar, J. Increased serum levels of Galectin-9 in patients with chronic lymphocytic leukemia. Oncol. Lett. 2019, 17, 1019–1029. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Gallois, A.; Silva, I.; Osman, I.; Bhardwaj, N. Reversal of natural killer cell exhaustion by TIM-3 blockade. Oncoimmunology 2015, 3, e946365. [Google Scholar] [CrossRef] [Green Version]
  185. Brusa, D.; Serra, S.; Coscia, M.; Rossi, D.; D’Arena, G.; Laurenti, L.; Jaksic, O.; Fedele, G.; Inghirami, G.; Gaidano, G.; et al. The PD-1/PD-L1 axis contributes to T-cell dysfunction in chronic lymphocytic leukemia. Haematologica 2013, 98, 953–963. [Google Scholar] [CrossRef]
  186. Palma, M.; Gentilcore, G.; Heimersson, K.; Mozaffari, F.; Näsman-Glaser, B.; Young, E.; Rosenquist, R.; Hansson, L.; Österborg, A.; Mellstedt, H. T cells in chronic lymphocytic leukemia display dysregulated expression of immune checkpoints and activation markers. Haematologica 2017, 102, 562–572. [Google Scholar] [CrossRef] [Green Version]
  187. Beldi-Ferchiou, A.; Lambert, M.; Dogniaux, S.; Vély, F.; Vivier, E.; Olive, D.; Dupuy, S.; Levasseur, F.; Zucman, D.; Lebbé, C.; et al. PD-1 mediates functional exhaustion of activated NK cells in patients with Kaposi sarcoma. Oncotarget 2016, 7, 72961–72977. [Google Scholar] [CrossRef] [Green Version]
  188. Grzywnowicz, M.; Karczmarczyk, A.; Skorka, K.; Zajac, M.; Zaleska, J.; Chocholska, S.; Tomczak, W.; Giannopoulos, K. Expression of Programmed Death 1 Ligand in Different Compartments of Chronic Lymphocytic Leukemia. Acta Haematol. 2015, 134, 255–262. [Google Scholar] [CrossRef]
  189. 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]
  190. Zhang, J.; Basher, F.; Wu, J.D. NKG2D Ligands in Tumor Immunity: Two Sides of a Coin. Front. Immunol. 2015, 6, 97. [Google Scholar] [CrossRef] [Green Version]
  191. Liu, G.; Lu, S.; Wang, X.; Page, S.T.; Higano, C.S.; Plymate, S.R.; Greenberg, N.M.; Sun, S.; Li, Z.; Wu, J.D. Perturbation of NK cell peripheral homeostasis accelerates prostate carcinoma metastasis. J. Clin. Investig. 2013, 123, 4410–4422. [Google Scholar] [CrossRef]
  192. Reinart, N.; Nguyen, P.H.; Boucas, J.; Rosen, N.; Kvasnicka, H.M.; Heukamp, L.; Rudolph, C.; Ristovska, V.; Velmans, T.; Mueller, C.; et al. Delayed development of chronic lymphocytic leukemia in the absence of macrophage migration inhibitory factor. Blood 2013, 121, 812–821. [Google Scholar] [CrossRef] [Green Version]
  193. Carosella, E.D. The tolerogenic molecule HLA-G. Immunol. Lett. 2011, 138, 22–24. [Google Scholar] [CrossRef]
  194. Liu, L.; Wang, L.; Zhao, L.; He, C.; Wang, G. The Role of HLA-G in Tumor Escape: Manipulating the Phenotype and Function of Immune Cells. Front. Oncol. 2020, 10, 597468. [Google Scholar] [CrossRef]
  195. LeMaoult, J.; Krawice-Radanne, I.; Dausset, J.; Carosella, E.D. HLA-G1-expressing antigen-presenting cells induce immunosuppressive CD4+ T cells. Proc. Natl. Acad. Sci. USA 2004, 101, 7064–7069. [Google Scholar] [CrossRef] [Green Version]
  196. Lin, A.; Yan, W.H. Human Leukocyte Antigen-G (HLA-G) Expression in Cancers: Roles in Immune Evasion, Metastasis and Target for Therapy. Mol. Med. 2015, 21, 782–791. [Google Scholar] [CrossRef]
  197. Erikci, A.A.; Karagoz, B.; Ozyurt, M.; Ozturk, A.; Kilic, S.; Bilgi, O. HLA-G expression in B chronic lymphocytic leukemia: A new prognostic marker? Hematology 2009, 14, 101–105. [Google Scholar] [CrossRef]
  198. Perez-Chacon, G.; Rosado, S.; Rebolleda, N.; Losada-Fernandez, I.; Vargas, J.A.; Morado, M.; Jorda, J.; Perez-Aciego, P. Prognostic irrelevance of HLA-G in B-cell chronic lymphocytic leukemia. Int. J. Lab. Hematol. 2009, 31, 327–337. [Google Scholar] [CrossRef]
  199. Sebti, Y.; Le Friec, G.; Pangault, C.; Gros, F.; Drénou, B.; Guilloux, V.; Bernard, M.; Lamy, T.; Fauchet, R.; Amiot, L. Soluble HLA-G molecules are increased in lymphoproliferative disorders. Hum. Immunol. 2003, 64, 1093–1101. [Google Scholar] [CrossRef]
  200. Giannopoulos, K.; Schmitt, M.; Kowal, M.; Własiuk, P.; Bojarska-Junak, A.; Roliński, J.; Dmoszyńska, A. The significance of soluble HLA-G plasma levels as well as messenger HLA-G for B-cell chronic lymphocytic leukemia (B-CLL). Leuk. Res. 2008, 32, 1815–1819. [Google Scholar] [CrossRef] [PubMed]
  201. Castelli, E.C.; Mendes-Junior, C.T.; Veiga-Castelli, L.C.; Roger, M.; Moreau, P.; Donadi, E.A. A comprehensive study of polymorphic sites along the HLA-G gene: Implication for gene regulation and evolution. Mol. Biol. Evol. 2011, 28, 3069–3086. [Google Scholar] [CrossRef] [Green Version]
  202. Zhang, Y.; Yu, S.; Han, Y.; Wang, Y.; Sun, Y. Human leukocyte antigen-G expression and polymorphisms promote cancer development and guide cancer diagnosis/treatment. Oncol. Lett. 2018, 15, 699–709. [Google Scholar] [CrossRef]
  203. Rousseau, P.; Le Discorde, M.; Mouillot, G.; Marcou, C.; Carosella, E.D.; Moreau, P. The 14 bp deletion-insertion polymorphism in the 3′ UT region of the HLA-G gene influences HLA-G mRNA stability. Hum. Immunol. 2003, 64, 1005–1010. [Google Scholar] [CrossRef]
  204. Weiss, L.; Melchardt, T.; Egle, A.; Grabmer, C.; Greil, R.; Tinhofer, I. Regulatory T cells predict the time to initial treatment in early stage chronic lymphocytic leukemia. Cancer 2011, 117, 2163–2169. [Google Scholar] [CrossRef] [PubMed]
  205. Buechele, C.; Baessler, T.; Wirths, S.; Schmohl, J.U.; Schmiedel, B.J.; Salih, H.R. Glucocorticoid-induced TNFR-related protein (GITR) ligand modulates cytokine release and NK cell reactivity in chronic lymphocytic leukemia (CLL). Leukemia 2012, 26, 991–1000. [Google Scholar] [CrossRef]
  206. Ferrajoli, A.; Keating, M.J.; Manshouri, T.; Giles, F.J.; Dey, A.; Estrov, Z.; Koller, C.A.; Kurzrock, R.; Thomas, D.A.; Faderl, S.; et al. The clinical significance of tumor necrosis factor-alpha plasma level in patients having chronic lymphocytic leukemia. Blood 2002, 100, 1215–1219. [Google Scholar] [CrossRef]
  207. Lai, R.; O’Brien, S.; Maushouri, T.; Rogers, A.; Kantarjian, H.; Keating, M.; Albitar, M. Prognostic value of plasma interleukin-6 levels in patients with chronic lymphocytic leukemia. Cancer 2002, 95, 1071–1075. [Google Scholar] [CrossRef]
  208. Binsky, I.; Haran, M.; Starlets, D.; Gore, Y.; Lantner, F.; Harpaz, N.; Leng, L.; Goldenberg, D.M.; Shvidel, L.; Berrebi, A.; et al. IL-8 secreted in a macrophage migration-inhibitory factor- and CD74-dependent manner regulates B cell chronic lymphocytic leukemia survival. Proc. Natl. Acad. Sci. USA 2007, 104, 13408–13413. [Google Scholar] [CrossRef] [Green Version]
  209. Buechele, C.; Baessler, T.; Schmiedel, B.J.; Schumacher, C.E.; Grosse-Hovest, L.; Rittig, K.; Salih, H.R. 4-1BB ligand modulates direct and Rituximab-induced NK-cell reactivity in chronic lymphocytic leukemia. Eur. J. Immunol. 2012, 42, 737–748. [Google Scholar] [CrossRef]
  210. Wild, J.; Schmiedel, B.J.; Maurer, A.; Raab, S.; Prokop, L.; Stevanović, S.; Dörfel, D.; Schneider, P.; Salih, H.R. Neutralization of (NK-cell-derived) B-cell activating factor by Belimumab restores sensitivity of chronic lymphoid leukemia cells to direct and Rituximab-induced NK lysis. Leukemia 2015, 29, 1676–1683. [Google Scholar] [CrossRef] [Green Version]
  211. Boross, P.; Leusen, J.H.; Boross, P.; Leusen, J.H. Mechanisms of action of CD20 antibodies. Am. J. Cancer Res. 2012, 2, 676–690. [Google Scholar]
  212. Jaglowski, S.M.; Byrd, J.C. Rituximab in chronic lymphocytic leukemia. Semin. Hematol. 2010, 47, 156–169. [Google Scholar] [CrossRef]
  213. Fischer, K.; Bahlo, J.; Fink, A.M.; Goede, V.; Herling, C.D.; Cramer, P.; Langerbeins, P.; von Tresckow, J.; Engelke, A.; Maurer, C.; et al. Long-term remissions after FCR chemoimmunotherapy in previously untreated patients with CLL: Updated results of the CLL8 trial. Blood 2016, 127, 208–215. [Google Scholar] [CrossRef] [Green Version]
  214. Eichhorst, B.; Fink, A.M.; Bahlo, J.; Busch, R.; Kovacs, G.; Maurer, C.; Lange, E.; Köppler, H.; Kiehl, M.; Sökler, M.; et al. First-line chemoimmunotherapy with bendamustine and rituximab versus fludarabine, cyclophosphamide, and rituximab in patients with advanced chronic lymphocytic leukaemia (CLL10): An international, open-label, randomised, phase 3, non-inferiority trial. Lancet Oncol. 2016, 17, 928–942. [Google Scholar] [CrossRef]
  215. Stilgenbauer, S.; Schnaiter, A.; Paschka, P.; Zenz, T.; Rossi, M.; Döhner, K.; Bühler, A.; Böttcher, S.; Ritgen, M.; Kneba, M.; et al. Gene mutations and treatment outcome in chronic lymphocytic leukemia: Results from the CLL8 trial. Blood 2014, 123, 3247–3254. [Google Scholar] [CrossRef] [Green Version]
  216. Sportoletti, P.; Baldoni, S.; Del Papa, B.; Aureli, P.; Dorillo, E.; Ruggeri, L.; Plebani, S.; Amico, V.; Di Tommaso, A.; Rosati, E.; et al. A revised NOTCH1 mutation frequency still impacts survival while the allele burden predicts early progression in chronic lymphocytic leukemia. Leukemia 2014, 28, 436–439. [Google Scholar] [CrossRef]
  217. De Falco, F.; Sabatini, R.; Del Papa, B.; Falzetti, F.; Di Ianni, M.; Sportoletti, P.; Baldoni, S.; Screpanti, I.; Marconi, P.; Rosati, E. Notch signaling sustains the expression of Mcl-1 and the activity of eIF4E to promote cell survival in CLL. Oncotarget 2015, 6, 16559–16572. [Google Scholar] [CrossRef] [Green Version]
  218. Vyas, M.; Müller, R.; Pogge von Strandmann, E. Antigen Loss Variants: Catching Hold of Escaping Foes. Front. Immunol. 2017, 8, 175. [Google Scholar] [CrossRef] [Green Version]
  219. Cartron, G.; Dacheux, L.; Salles, G.; Solal-Celigny, P.; Bardos, P.; Colombat, P.; Watier, H. Therapeutic activity of humanized anti-CD20 monoclonal antibody and polymorphism in IgG Fc receptor FcgammaRIIIa gene. Blood 2002, 99, 754–758. [Google Scholar] [CrossRef] [Green Version]
  220. Werlenius, O.; Aurelius, J.; Hallner, A.; Akhiani, A.A.; Simpanen, M.; Martner, A.; Andersson, P.O.; Hellstrand, K.; Thorén, F.B. Reactive oxygen species induced by therapeutic CD20 antibodies inhibit natural killer cell-mediated antibody-dependent cellular cytotoxicity against primary CLL cells. Oncotarget 2016, 7, 32046–32053. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  221. Sanchez-Martinez, D.; Allende-Vega, N.; Orecchioni, S.; Talarico, G.; Cornillon, A.; Vo, D.N.; Rene, C.; Lu, Z.Y.; Krzywinska, E.; Anel, A.; et al. Expansion of allogeneic NK cells with efficient antibody-dependent cell cytotoxicity against multiple tumors. Theranostics 2018, 8, 3856–3869. [Google Scholar] [CrossRef] [PubMed]
  222. van der Horst, H.J.; Nijhof, I.S.; Mutis, T.; Chamuleau, M. Fc-Engineered Antibodies with Enhanced Fc-Effector Function for the Treatment of B-Cell Malignancies. Cancers 2020, 12, 3041. [Google Scholar] [CrossRef] [PubMed]
  223. Offner, F.; Robak, T.; Janssens, A.; Govind Babu, K.; Kloczko, J.; Grosicki, S.; Mayer, J.; Panagiotidis, P.; Schuh, A.; Pettitt, A.; et al. A five-year follow-up of untreated patients with chronic lymphocytic leukaemia treated with ofatumumab and chlorambucil: Final analysis of the Complement 1 phase 3 trial. Br. J. Haematol. 2020, 190, 736–740. [Google Scholar] [CrossRef]
  224. Jones, J.A.; Robak, T.; Brown, J.R.; Awan, F.T.; Badoux, X.; Coutre, S.; Loscertales, J.; Taylor, K.; Vandenberghe, E.; Wach, M.; et al. Efficacy and safety of idelalisib in combination with ofatumumab for previously treated chronic lymphocytic leukaemia: An open-label, randomised phase 3 trial. Lancet Haematol. 2017, 4, e114–e126. [Google Scholar] [CrossRef]
  225. Moreno, C.; Greil, R.; Demirkan, F.; Tedeschi, A.; Anz, B.; Larratt, L.; Simkovic, M.; Samoilova, O.; Novak, J.; Ben-Yehuda, D.; et al. Ibrutinib plus obinutuzumab versus chlorambucil plus obinutuzumab in first-line treatment of chronic lymphocytic leukaemia (iLLUMINATE): A multicentre, randomised, open-label, phase 3 trial [published correction appears in Lancet Oncol. 2019 Jan;20(1):e10]. Lancet Oncol. 2019, 20, 43–56. [Google Scholar] [CrossRef]
  226. Sharman, J.P.; Egyed, M.; Jurczak, W.; Skarbnik, A.; Pagel, J.M.; Flinn, I.W.; Kamdar, M.; Munir, T.; Walewska, R.; Corbett, G.; et al. Acalabrutinib with or without obinutuzumab versus chlorambucil and obinutuzumab for treatment-naive chronic lymphocytic leukaemia (ELEVATE TN): A randomised, controlled, phase 3 trial. Lancet 2020, 395, 1278–1291. [Google Scholar] [CrossRef]
  227. Fischer, K.; Al-Sawaf, O.; Bahlo, J.; Fink, A.M.; Tandon, M.; Dixon, M.; Robrecht, S.; Warburton, S.; Humphrey, K.; Samoylova, O.; et al. Venetoclax and Obinutuzumab in Patients with CLL and Coexisting Conditions. N. Engl. J. Med. 2019, 380, 2225–2236. [Google Scholar] [CrossRef]
  228. Sawas, A.; Farber, C.M.; Schreeder, M.T.; Khalil, M.Y.; Mahadevan, D.; Deng, C.; Amengual, J.E.; Nikolinakos, P.G.; Kolesar, J.M.; Kuhn, J.G.; et al. A phase 1/2 trial of ublituximab, a novel anti-CD20 monoclonal antibody, in patients with B-cell non-Hodgkin lymphoma or chronic lymphocytic leukaemia previously exposed to rituximab. Br. J. Haematol. 2017, 177, 243–253. [Google Scholar] [CrossRef]
  229. Sharman, J.P.; Farber, C.M.; Mahadevan, D.; Schreeder, M.T.; Brooks, H.D.; Kolibaba, K.S.; Fanning, S.; Klein, L.; Greenwald, D.R.; Sportelli, P.; et al. Ublituximab (TG-1101), a novel glycoengineered anti-CD20 antibody, in combination with ibrutinib is safe and highly active in patients with relapsed and/or refractory chronic lymphocytic leukaemia: Results of a phase 2 trial. Br. J. Haematol. 2017, 176, 412–420. [Google Scholar] [CrossRef]
  230. Sharman, J.P.; Brander, D.M.; Mato, A.R.; Ghosh, N.; Schuster, S.J.; Kambhampati, S.; Burke, J.M.; Lansigan, F.; Schreeder, M.T.; Lunin, S.D.; et al. Ublituximab plus ibrutinib versus ibrutinib alone for patients with relapsed or refractory high-risk chronic lymphocytic leukaemia (GENUINE): A phase 3, multicentre, open-label, randomised trial. Lancet Haematol. 2021, 8, e254–e266. [Google Scholar] [CrossRef]
  231. Cardarelli, P.M.; Rao-Naik, C.; Chen, S.; Huang, H.; Pham, A.; Moldovan-Loomis, M.C.; Pan, C.; Preston, B.; Passmore, D.; Liu, J.; et al. A nonfucosylated human antibody to CD19 with potent B-cell depletive activity for therapy of B-cell malignancies. Cancer Immunol. Immunother. 2010, 59, 257–265. [Google Scholar] [CrossRef]
  232. Horton, H.M.; Bernett, M.J.; Pong, E.; Peipp, M.; Karki, S.; Chu, S.Y.; Richards, J.O.; Vostiar, I.; Joyce, P.F.; Repp, R.; et al. Potent in vitro and in vivo activity of an Fc-engineered anti-CD19 monoclonal antibody against lymphoma and leukemia. Cancer Res. 2008, 68, 8049–8057. [Google Scholar] [CrossRef] [Green Version]
  233. Ohmachi, K.; Ogura, M.; Suehiro, Y.; Ando, K.; Uchida, T.; Choi, I.; Ogawa, Y.; Kobayashi, M.; Fukino, K.; Yokoi, Y.; et al. A multicenter phase I study of inebilizumab, a humanized anti-CD19 monoclonal antibody, in Japanese patients with relapsed or refractory B-cell lymphoma and multiple myeloma. Int. J. Hematol. 2019, 109, 657–664. [Google Scholar] [CrossRef]
  234. Jurczak, W.; Zinzani, P.L.; Gaidano, G.; Goy, A.; Provencio, M.; Nagy, Z.; Robak, T.; Maddocks, K.; Buske, C.; Ambarkhane, S.; et al. Phase IIa study of the CD19 antibody MOR208 in patients with relapsed or refractory B-cell non-Hodgkin’s lymphoma. Ann. Oncol. 2018, 29, 1266–1272. [Google Scholar] [CrossRef]
  235. Payandeh, Z.; Noori, E.; Khalesi, B.; Mard-Soltani, M.; Abdolalizadeh, J.; Khalili, S. Anti-CD37 targeted immunotherapy of B-Cell malignancies. Biotechnol. Lett. 2018, 40, 1459–1466. [Google Scholar] [CrossRef]
  236. Witkowska, M.; Smolewski, P.; Robak, T. Investigational therapies targeting CD37 for the treatment of B-cell lymphoid malignancies. Expert. Opin. Investig. Drugs 2018, 27, 171–177. [Google Scholar] [CrossRef]
  237. Betrian, S.; Ysebaert, L.; Heider, K.H.; Delord, J.P.; Fournié, J.J.; Quillet-Mary, A. Idelalisib improves CD37 antibody BI 836826 cytotoxicity against chemo-resistant /relapse-initiating CLL cells: A rationale for combination treatment. Blood Cancer J. 2016, 6, e496. [Google Scholar] [CrossRef]
  238. Stilgenbauer, S.; Aurran Schleinitz, T.; Eichhorst, B.; Lang, F.; Offner, F.; Rossi, J.F.; Schroyens, W.; Van Den Neste, E.; Ysebaert, L.; von Wangenheim, U.; et al. Phase 1 first-in-human trial of the anti-CD37 antibody BI 836826 in relapsed/refractory chronic lymphocytic leukemia. Leukemia 2019, 33, 2531–2535. [Google Scholar] [CrossRef]
  239. Robak, T.; Robak, P.; Smolewski, P. TRU-016, a humanized anti-CD37 IgG fusion protein for the potential treatment of B-cell malignancies. Curr. Opin. Investig. Drugs 2009, 10, 1383–1390. [Google Scholar]
  240. Byrd, J.C.; Pagel, J.M.; Awan, F.T.; Forero, A.; Flinn, I.W.; Deauna-Limayo, D.P.; Spurgeon, S.E.; Andritsos, L.A.; Gopal, A.K.; Leonard, J.P.; et al. A phase 1 study evaluating the safety and tolerability of otlertuzumab, an anti-CD37 mono-specific ADAPTIR therapeutic protein in chronic lymphocytic leukemia. Blood 2014, 123, 1302–1308. [Google Scholar] [CrossRef] [Green Version]
  241. Robak, T.; Hellmann, A.; Kloczko, J.; Loscertales, J.; Lech-Maranda, E.; Pagel, J.M.; Mato, A.; Byrd, J.C.; Awan, F.T.; Hebart, H.; et al. Randomized phase 2 study of otlertuzumab and bendamustine versus bendamustine in patients with relapsed chronic lymphocytic leukaemia. Br. J. Haematol. 2017, 176, 618–628. [Google Scholar] [CrossRef]
  242. Labrijn, A.F.; Janmaat, M.L.; Reichert, J.M.; Parren, P. Bispecific antibodies: A mechanistic review of the pipeline. Nat. Rev. Drug Discov. 2019, 18, 585–608. [Google Scholar] [CrossRef]
  243. Goebeler, M.E.; Bargou, R.C. T cell-engaging therapies—BiTEs and beyond. Nat. Rev. Clin. Oncol. 2020, 17, 418–434. [Google Scholar] [CrossRef]
  244. Bruenke, J.; Fischer, B.; Barbin, K.; Schreiter, K.; Wachter, Y.; Mahr, K.; Titgemeyer, F.; Niederweis, M.; Peipp, M.; Zunino, S.J.; et al. A recombinant bispecific single-chain Fv antibody against HLA class II and FcgammaRIII (CD16) triggers effective lysis of lymphoma cells. Br. J. Haematol. 2004, 125, 167–179. [Google Scholar] [CrossRef] [PubMed]
  245. Kellner, C.; Bruenke, J.; Horner, H.; Schubert, J.; Schwenkert, M.; Mentz, K.; Barbin, K.; Stein, C.; Peipp, M.; Stockmeyer, B.; et al. Heterodimeric bispecific antibody-derivatives against CD19 and CD16 induce effective antibody-dependent cellular cytotoxicity against B-lymphoid tumor cells. Cancer Lett. 2011, 303, 128–139. [Google Scholar] [CrossRef]
  246. Gleason, M.K.; Verneris, M.R.; Todhunter, D.A.; Zhang, B.; McCullar, V.; Zhou, S.X.; Panoskaltsis-Mortari, A.; Weiner, L.M.; Vallera, D.A.; Miller, J.S. Bispecific and trispecific killer cell engagers directly activate human NK cells through CD16 signaling and induce cytotoxicity and cytokine production. Mol. Cancer Ther. 2012, 11, 2674–2684. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  247. Felices, M.; Kodal, B.; Hinderlie, P.; Kaminski, M.F.; Cooley, S.; Weisdorf, D.J.; Vallera, D.A.; Miller, J.S.; Bachanova, V. Novel CD19-targeted TriKE restores NK cell function and proliferative capacity in CLL. Blood Adv. 2019, 3, 897–907. [Google Scholar] [CrossRef] [PubMed]
  248. Vyas, M.; Schneider, A.C.; Shatnyeva, O.; Reiners, K.S.; Tawadros, S.; Kloess, S.; Köhl, U.; Hallek, M.; Hansen, H.P.; Pogge von Strandmann, E. Mono- and dual-targeting triplebodies activate natural killer cells and have anti-tumor activity in vitro and in vivo against chronic lymphocytic leukemia. Oncoimmunology 2016, 5, e1211220. [Google Scholar] [CrossRef] [Green Version]
  249. Muntasell, A.; Ochoa, M.C.; Cordeiro, L.; Berraondo, P.; López-Díaz de Cerio, A.; Cabo, M.; López-Botet, M.; Melero, I. Targeting NK-cell checkpoints for cancer immunotherapy. Curr. Opin. Immunol. 2017, 45, 73–81. [Google Scholar] [CrossRef] [PubMed]
  250. Riches, J.C.; Davies, J.K.; McClanahan, F.; Fatah, R.; Iqbal, S.; Agrawal, S.; Ramsay, A.G.; Gribben, J.G. T cells from CLL patients exhibit features of T-cell exhaustion but retain capacity for cytokine production. Blood 2013, 121, 1612–1621. [Google Scholar] [CrossRef]
  251. Ramsay, A.G.; Clear, A.J.; Fatah, R.; Gribben, J.G. Multiple inhibitory ligands induce impaired T-cell immunologic synapse function in chronic lymphocytic leukemia that can be blocked with lenalidomide: Establishing a reversible immune evasion mechanism in human cancer. Blood 2012, 120, 1412–1421. [Google Scholar] [CrossRef] [PubMed]
  252. McClanahan, F.; Hanna, B.; Miller, S.; Clear, A.J.; Lichter, P.; Gribben, J.G.; Seiffert, M. PD-L1 checkpoint blockade prevents immune dysfunction and leukemia development in a mouse model of chronic lymphocytic leukemia. Blood 2015, 126, 203–211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  253. Younes, A.; Brody, J.; Carpio, C.; Lopez-Guillermo, A.; Ben-Yehuda, D.; Ferhanoglu, B.; Nagler, A.; Ozcan, M.; Avivi, I.; Bosch, F.; et al. Safety and activity of ibrutinib in combination with nivolumab in patients with relapsed non-Hodgkin lymphoma or chronic lymphocytic leukaemia: A phase 1/2a study. Lancet Haematol. 2019, 6, e67–e78. [Google Scholar] [CrossRef]
  254. Condoluci, A.; Rossi, D. Richter Syndrome. Curr. Oncol. Rep. 2021, 23, 26. [Google Scholar] [CrossRef]
  255. Vey, N.; Karlin, L.; Sadot-Lebouvier, S.; Broussais, F.; Berton-Rigaud, D.; Rey, J.; Charbonnier, A.; Marie, D.; André, P.; Paturel, C.; et al. A phase 1 study of lirilumab (antibody against killer immunoglobulin-like receptor antibody KIR2D.; IPH2102) in patients with solid tumors and hematologic malignancies. Oncotarget 2018, 9, 17675–17688. [Google Scholar] [CrossRef] [Green Version]
  256. Shapiro, M.; Herishanu, Y.; Katz, B.Z.; Dezorella, N.; Sun, C.; Kay, S.; Polliack, A.; Avivi, I.; Wiestner, A.; Perry, C. Lymphocyte activation gene 3: A novel therapeutic target in chronic lymphocytic leukemia. Haematologica 2017, 102, 874–882. [Google Scholar] [CrossRef] [Green Version]
  257. Sordo-Bahamonde, C.; Lorenzo-Herrero, S.; González-Rodríguez, A.P.; Payer, Á.R.; González-García, E.; López-Soto, A.; Gonzalez, S. LAG-3 Blockade with Relatlimab (BMS-986016) Restores Anti-Leukemic Responses in Chronic Lymphocytic Leukemia. Cancers 2021, 13, 2112. [Google Scholar] [CrossRef]
  258. Dreger, P.; Corradini, P.; Kimby, E.; Michallet, M.; Milligan, D.; Schetelig, J.; Wiktor-Jedrzejczak, W.; Niederwieser, D.; Hallek, M.; Montserrat, E. Indications for allogeneic stem cell transplantation in chronic lymphocytic leukemia. Chronic Leukemia Working Party of the EBMT. Leukemia 2007, 21, 12–17. [Google Scholar] [CrossRef] [Green Version]
  259. Burger, J.A.; Barr, P.M.; Robak, T.; Owen, C.; Ghia, P.; Tedeschi, A.; Bairey, O.; Hillmen, P.; Coutre, S.E.; Devereux, S.; et al. Long-term efficacy and safety of first-line ibrutinib treatment for patients with CLL/SLL: 5 years of follow-up from the phase 3 RESONATE-2 study. Leukemia 2020, 34, 787–798. [Google Scholar] [CrossRef] [Green Version]
  260. Flinn, I.W.; Hillmen, P.; Montillo, M.; Nagy, Z.; Illés, Á.; Etienne, G.; Delgado, J.; Kuss, B.J.; Tam, C.S.; Gasztonyi, Z.; et al. The phase 3 DUO trial: Duvelisib vs ofatumumab in relapsed and refractory CLL/SLL. Blood 2018, 132, 2446–2455. [Google Scholar] [CrossRef] [Green Version]
  261. Stilgenbauer, S.; Eichhorst, B.; Schetelig, J.; Hillmen, P.; Seymour, J.F.; Coutre, S.; Jurczak, W.; Mulligan, S.P.; Schuh, A.; Assouline, S.; et al. Venetoclax for patients with chronic lymphocytic leukemia with 17p deletion: Results from the full population of a phase II pivotal trial. J. Clin. Oncol. 2018, 36, 1973–1980. [Google Scholar] [CrossRef]
  262. Gribben, J.G. How and when I do allogeneic transplant in CLL. Blood 2018, 132, 31–39. [Google Scholar] [CrossRef] [Green Version]
  263. Dreger, P.; Schetelig, J.; Andersen, N.; Corradini, P.; van Gelder, M.; Gribben, J.; Kimby, E.; Michallet, M.; Moreno, C.; Stilgenbauer, S.; et al. Managing high-risk CLL during transition to a new treatment era: Stem cell transplantation or novel agents? European Research Initiative on CLL (ERIC) and the European Society for Blood and Marrow Transplantation (EBMT). Blood 2014, 124, 3841–3849. [Google Scholar] [CrossRef] [PubMed]
  264. D’Souza, A.; Fretham, C.; Lee, S.J.; Arora, M.; Brunner, J.; Chhabra, S.; Devine, S.; Eapen, M.; Hamadani, M.; Hari, P.; et al. Current Use of and Trends in Hematopoietic Cell Transplantation in the United States. Biol. Blood Marrow Transplant. 2020, 26, E177–E182. [Google Scholar] [CrossRef]
  265. Locatelli, F.; Pende, D.; Falco, M.; Della Chiesa, M.; Moretta, A.; Moretta, L. NK Cells Mediate a Crucial Graft-versus-Leukemia Effect in Haploidentical-HSCT to Cure High-Risk Acute Leukemia. Trends Immunol. 2018, 39, 577–590. [Google Scholar] [CrossRef] [PubMed]
  266. Hamilton, B.K.; Copelan, E.A. Concise review: The role of hematopoietic stem cell transplantation in the treatment of acute myeloid leukemia. Stem Cells 2012, 30, 1581–1586. [Google Scholar] [CrossRef]
  267. Gribben, J.G.; Zahrieh, D.; Stephans, K.; Bartlett-Pandite, L.; Alyea, E.P.; Fisher, D.C.; Freedman, A.S.; Mauch, P.; Schlossman, R.; Sequist, L.V.; et al. Autologous and allogeneic stem cell transplantations for poor-risk chronic lymphocytic leukemia. Blood 2005, 106, 389–396. [Google Scholar] [CrossRef] [Green Version]
  268. Bachanova, V.; Weisdorf, D.J.; Wang, T.; Marsh, S.G.E.; Cereb, N.; Haagenson, M.D.; Spellman, S.R.; Lee, S.J.; Guethlein, L.A.; Parham, P.; et al. Donor Killer Cell Immunoglobulin-Like Receptor Genotype Does Not Improve Graft-versus-Leukemia Responses in Chronic Lymphocytic Leukemia after Unrelated Donor Transplant: A Center for International Blood and Marrow Transplant Research Analysis. Biol. Blood Marrow Transplant. 2019, 25, 949–954. [Google Scholar] [CrossRef]
  269. van Gorkom, G.; van Gelder, M.; Eikema, D.J.; Blok, H.J.; van Lint, M.T.; Koc, Y.; Ciceri, F.; Beelen, D.; Chevallier, P.; Selleslag, D.; et al. CLL subcommittee; Chronic Malignancies Working Party of the EBMT. Outcomes of haploidentical stem cell transplantation for chronic lymphocytic leukemia: A retrospective study on behalf of the chronic malignancies working party of the EBMT. Bone Marrow Transplant. 2018, 53, 255–263. [Google Scholar] [CrossRef] [Green Version]
  270. Calvo, T.; Reina-Ortiz, C.; Giraldos, D.; Gascón, M.; Woods, D.; Asenjo, J.; Marco-Brualla, J.; Azaceta, G.; Izquierdo, I.; Palomera, L.; et al. Expanded and activated allogeneic NK cells are cytotoxic against B-chronic lymphocytic leukemia (B-CLL) cells with sporadic cases of resistance. Sci. Rep. 2020, 10, 19398. [Google Scholar] [CrossRef]
  271. Di Ianni, M.; Del Papa, B.; Zei, T.; Iacucci Ostini, R.; Cecchini, D.; Cantelmi, M.G.; Baldoni, S.; Sportoletti, P.; Cavalli, L.; Carotti, A.; et al. T regulatory cell separation for clinical application. Transfus. Apher. Sci. 2012, 47, 213–216. [Google Scholar] [CrossRef]
  272. Olson, J.A.; Leveson-Gower, D.B.; Gill, S.; Baker, J.; Beilhack, A.; Negrin, R.S. NK cells mediate reduction of GVHD by inhibiting activated, alloreactive T cells while retaining GVT effects. Blood 2010, 115, 4293–4301. [Google Scholar] [CrossRef] [Green Version]
  273. Fesnak, A.D.; June, C.H.; Levine, B.L. Engineered T cells: The promise and challenges of cancer immunotherapy. Nat. Rev. Cancer 2016, 16, 566–581. [Google Scholar] [CrossRef]
  274. Rafei, H.; Daher, M.; Rezvani, K. Chimeric antigen receptor (CAR) natural killer (NK)-cell therapy: Leveraging the power of innate immunity. Br. J. Haematol. 2021, 193, 216–230. [Google Scholar] [CrossRef] [PubMed]
  275. Porter, D.L.; Levine, B.L.; Kalos, M.; Bagg, A.; June, C.H. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N. Engl. J. Med. 2011, 365, 725–733. [Google Scholar] [CrossRef] [Green Version]
  276. Brentjens, R.J.; Rivière, I.; Park, J.H.; Davila, M.L.; Wang, X.; Stefanski, J.; Taylor, C.; Yeh, R.; Bartido, S.; Borquez-Ojeda, O.; et al. Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood 2011, 118, 4817–4828. [Google Scholar] [CrossRef] [PubMed]
  277. Kalos, M.; Levine, B.L.; Porter, D.L.; Katz, S.; Grupp, S.A.; Bagg, A.; June, C.H. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci. Transl. Med. 2011, 95, 95ra73. [Google Scholar] [CrossRef] [Green Version]
  278. Porter, D.L.; Hwang, W.T.; Frey, N.V.; Lacey, S.F.; Shaw, P.A.; Loren, A.W.; Bagg, A.; Marcucci, K.T.; Shen, A.; Gonzalez, V.; et al. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci. Transl. Med. 2015, 7, 303ra139. [Google Scholar] [CrossRef] [Green Version]
  279. Turtle, C.J.; Hay, K.A.; Hanafi, L.A.; Li, D.; Cherian, S.; Chen, X.; Wood, B.; Lozanski, A.; Byrd, J.C.; Heimfeld, S.; et al. Durable molecular remissions in chronic lymphocytic leukemia treated with CD19-specific chimeric antigen receptor-modified T cells after failure of ibrutinib. J. Clin. Oncol. 2017, 35, 3010–3020. [Google Scholar] [CrossRef]
  280. Geyer, M.B.; Rivière, I.; Sénéchal, B.; Wang, X.; Wang, Y.; Purdon, T.J.; Hsu, M.; Devlin, S.M.; Halton, E.; Lamanna, N.; et al. Autologous CD19-targeted CAR T cells in patients with residual CLL following initial purine analog-based therapy. Mol. Ther. J. Am. Soc. Gene Ther. 2018, 26, 1896–1905. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  281. Skorka, K.; Ostapinska, K.; Malesa, A.; Giannopoulos, K. The Application of CAR-T Cells in Haematological Malignancies. Arch. Immunol. Ther. Exp. 2020, 68, 34. [Google Scholar] [CrossRef] [PubMed]
  282. Lemal, R.; Tournilhac, O. State-of-the-art for CAR T-cell therapy for chronic lymphocytic leukemia in 2019. J. Immunother. Cancer 2019, 7, 202. [Google Scholar] [CrossRef] [Green Version]
  283. Hoffmann, J.M.; Schubert, M.L.; Wang, L.; Huckelhoven, A.; Sellner, L.; Stock, S.; Schmitt, A.; Kleist, C.; Gern, U.; Loskog, A.; et al. Differences in expansion potential of naive chimeric antigen receptor T cells from healthy donors and untreated chronic lymphocytic leukemia patients. Front. Immunol. 2018, 8, 1956. [Google Scholar] [CrossRef] [Green Version]
  284. Magalhaes, I.; Kalland, I.; Kochenderfer, J.N.; Osterborg, A.; Uhlin, M.; Mattsson, J. CD19 Chimeric Antigen Receptor T Cells From Patients With Chronic Lymphocytic Leukemia Display an Elevated IFN-gamma Production Profile. J. Immunother. 2018, 41, 73–83. [Google Scholar] [CrossRef]
  285. Goulmy, E. Human minor histocompatibility antigens: New concepts for marrow transplantation and adoptive immunotherapy. Immunol. Rev. 1997, 157, 125–140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  286. Gauthier, J.; Hirayama, A.V.; Purushe, J.; Hay, K.A.; Lymp, J.; Li, D.H.; Yeung, C.C.S.; Sheih, A.; Pender, B.S.; Hawkins, R.M.; et al. Feasibility and efficacy of CD19-targeted CAR T cells with concurrent ibrutinib for CLL after ibrutinib failure. Blood 2020, 135, 1650–1660. [Google Scholar] [CrossRef] [PubMed]
  287. Qin, J.S.; Johnstone, T.G.; Baturevych, A.; Hause, R.J.; Ragan, S.P.; Clouser, C.R.; Jones, J.C.; Ponce, R.; Krejsa, C.M.; Salmon, R.A.; et al. Antitumor Potency of an Anti-CD19 Chimeric Antigen Receptor T-Cell Therapy, Lisocabtagene Maraleucel in Combination with Ibrutinib or Acalabrutinib. J. Immunother. 2020, 43, 107–120. [Google Scholar] [CrossRef]
  288. Long, M.; Beckwith, K.; Do, P.; Mundy, B.L.; Gordon, A.; Lehman, A.M.; Maddocks, K.J.; Cheney, C.; Jones, J.A.; Flynn, J.M.; et al. Ibrutinib treatment improves T cell number and function in CLL patients. J. Clin. Investig. 2017, 127, 3052–3064. [Google Scholar] [CrossRef] [PubMed]
  289. Wang, L.; Dou, M.; Ma, Q.; Yao, R.; Liu, J. Chimeric antigen receptor (CAR)-modified NK cells against cancer: Opportunities and challenges. Int. Immunopharmacol. 2019, 74, 105695. [Google Scholar] [CrossRef]
  290. Rotolo, R.; Leuci, V.; Donini, C.; Cykowska, A.; Gammaitoni, L.; Medico, G.; Valabrega, G.; Aglietta, M.; Sangiolo, D. CAR-Based Strategies beyond T Lymphocytes: Integrative Opportunities for Cancer Adoptive Immunotherapy. Int. J. Mol. Sci. 2019, 20, 2839. [Google Scholar] [CrossRef] [Green Version]
  291. Rezvani, K. Adoptive cell therapy using engineered natural killer cells. Bone Marrow Transplant. 2019, 54, 785–788. [Google Scholar] [CrossRef]
  292. Sotillo, E.; Barrett, D.M.; Black, K.L.; Bagashev, A.; Oldridge, D.; Wu, G.; Sussman, R.; Lanauze, C.; Ruella, M.; Gazzara, M.R.; et al. Convergence of acquired mutations and alternative splicing of CD19 enables resistance to CART-19 immunotherapy. Cancer Discov. 2015, 5, 1282–1295. [Google Scholar] [CrossRef] [Green Version]
  293. Herrera, L.; Santos, S.; Vesga, M.A.; Anguita, J.; Martin-Ruiz, I.; Carrascosa, T.; Juan, M.; Eguizabal, C. Adult peripheral blood and umbilical cord blood NK cells are good sources for effective CAR therapy against CD19 positive leukemic cells. Sci. Rep. 2019, 9, 18729. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  294. 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] [PubMed] [Green Version]
  295. Muller, T.; Uherek, C.; Maki, G.; Chow, K.U.; Schimpf, A.; Klingemann, H.G.; Tonn, T.; Wels, W.S. Expression of a CD20-specific chimeric antigen receptor enhances cytotoxic activity of NK cells and overcomes NK-resistance of lymphoma and leukemia cells. Cancer Immunol. Immunother. 2008, 57, 411–423. [Google Scholar] [CrossRef] [PubMed]
  296. Grote, S.; Mittelstaet, J.; Baden, C.; Chun-Ho Chan, K.; Seitz, C.; Schlegel, P.; Kaiser, A.; Handgretinger, R.; Schleicher, S. Adapter chimeric antigen receptor (AdCAR)-engineered NK-92 cells: An off-the-shelf cellular therapeutic for universal tumor targeting. Oncoimmunology 2020, 9, 1825177. [Google Scholar] [CrossRef]
  297. Fujisaki, H.; Kakuda, H.; Shimasaki, N.; Imai, C.; Ma, J.; Lockey, T.; Eldridge, P.; Leung, W.H.; Campana, D. Expansion of highly cytotoxic human natural killer cells for cancer cell therapy. Cancer Res. 2009, 69, 4010–4017. [Google Scholar] [CrossRef] [Green Version]
  298. Clausen, J.; Petzer, A.L.; Vergeiner, B.; Enk, M.; Stauder, R.; Gastl, G.; Gunsilius, E. Optimal timing for the collection and in vitro expansion of cytotoxic CD56(+) lymphocytes from patients undergoing autologous peripheral blood stem cell transplantation. J. Hematother. Stem Cell Res. 2001, 10, 513–521. [Google Scholar] [CrossRef]
  299. Imamura, M.; Shook, D.; Kamiya, T.; Shimasaki, N.; Chai, S.M.; Coustan-Smith, E.; Imai, C.; Campana, D. Autonomous growth and increased cytotoxicity of natural killer cells expressing membrane-bound interleukin-15. Blood 2014, 124, 1081–1088. [Google Scholar] [CrossRef] [Green Version]
  300. Nagashima, S.; Mailliard, R.; Kashii, Y.; Reichert, T.E.; Herberman, R.B.; Robbins, P.; Whiteside, T.L. Stable transduction of the interleukin-2 gene into human natural killer cell lines and their phenotypic and functional characterization in vitro and in vivo. Blood 1998, 91, 3850–3861. [Google Scholar] [CrossRef] [Green Version]
  301. Liu, E.; Tong, Y.; Dotti, G.; Shaim, H.; Savoldo, B.; Mukherjee, M.; Orange, J.; Wan, X.; Lu, X.; Reynolds, A. Cord blood NK cells engineered to express IL-15 and a CD19-targeted CAR show long-term persistence and potent antitumor activity. Leukemia 2018, 32, 520–531. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Receptor–ligand interactions regulating natural killer (NK) cell functions. Schematic representation of NK cell activity regulation by signals triggered by cell surface receptors. Activating and inhibitory receptors with their corresponding ligands, as well as adhesion molecules and receptors for stimulating cytokines, are shown.
Figure 1. Receptor–ligand interactions regulating natural killer (NK) cell functions. Schematic representation of NK cell activity regulation by signals triggered by cell surface receptors. Activating and inhibitory receptors with their corresponding ligands, as well as adhesion molecules and receptors for stimulating cytokines, are shown.
Ijms 22 06665 g001
Figure 2. A hypothetical model of chronic lymphocytic leukemia (CLL) escape from natural killer (NK) cell immune surveillance. The escape of CLL cells from the NK cell response relies on multiple mechanisms: (i) reduced expression of activating receptors on NK cells or their ligands on CLL cells; (ii) release by CLL cells of soluble ligands for NK cell activating receptors; (iii) increased expression of inhibitory receptors on NK cell surface and of their ligands on CLL cells; (iv) NK cell-induced signals that increase CLL cell growth/survival and metabolic activity and impair CLL cell susceptibility to NK cell-mediated cytotoxicity. The red and blue arrows indicate increased and decreased cell surface expression, respectively, of NK cell receptors or their ligands on CLL cells. Dotted line indicates GITR–GITRL interaction. Dotted arrows indicate intracellular signaling.
Figure 2. A hypothetical model of chronic lymphocytic leukemia (CLL) escape from natural killer (NK) cell immune surveillance. The escape of CLL cells from the NK cell response relies on multiple mechanisms: (i) reduced expression of activating receptors on NK cells or their ligands on CLL cells; (ii) release by CLL cells of soluble ligands for NK cell activating receptors; (iii) increased expression of inhibitory receptors on NK cell surface and of their ligands on CLL cells; (iv) NK cell-induced signals that increase CLL cell growth/survival and metabolic activity and impair CLL cell susceptibility to NK cell-mediated cytotoxicity. The red and blue arrows indicate increased and decreased cell surface expression, respectively, of NK cell receptors or their ligands on CLL cells. Dotted line indicates GITR–GITRL interaction. Dotted arrows indicate intracellular signaling.
Ijms 22 06665 g002
Figure 3. Natural killer (NK) cells in chronic lymphocytic leukemia (CLL) immunotherapy. Different strategies have been developed to harness NK cell activity against CLL cells: (A) Enhancement of NK cell-mediated antibody-dependent cellular cytotoxicity (ADCC) using tumor-specific monoclonal antibodies (mAbs) (left) or bispecific and trispecific killer cell engagers (BiKE and TriKE) (right). (B) Restoration of NK cell functions using mAbs targeting immune checkpoints. (C) Chimeric antigen receptor (CAR)-NK cell therapy.
Figure 3. Natural killer (NK) cells in chronic lymphocytic leukemia (CLL) immunotherapy. Different strategies have been developed to harness NK cell activity against CLL cells: (A) Enhancement of NK cell-mediated antibody-dependent cellular cytotoxicity (ADCC) using tumor-specific monoclonal antibodies (mAbs) (left) or bispecific and trispecific killer cell engagers (BiKE and TriKE) (right). (B) Restoration of NK cell functions using mAbs targeting immune checkpoints. (C) Chimeric antigen receptor (CAR)-NK cell therapy.
Ijms 22 06665 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Sportoletti, P.; De Falco, F.; Del Papa, B.; Baldoni, S.; Guarente, V.; Marra, A.; Dorillo, E.; Rompietti, C.; Adamo, F.M.; Ruggeri, L.; et al. NK Cells in Chronic Lymphocytic Leukemia and Their Therapeutic Implications. Int. J. Mol. Sci. 2021, 22, 6665. https://doi.org/10.3390/ijms22136665

AMA Style

Sportoletti P, De Falco F, Del Papa B, Baldoni S, Guarente V, Marra A, Dorillo E, Rompietti C, Adamo FM, Ruggeri L, et al. NK Cells in Chronic Lymphocytic Leukemia and Their Therapeutic Implications. International Journal of Molecular Sciences. 2021; 22(13):6665. https://doi.org/10.3390/ijms22136665

Chicago/Turabian Style

Sportoletti, Paolo, Filomena De Falco, Beatrice Del Papa, Stefano Baldoni, Valerio Guarente, Andrea Marra, Erica Dorillo, Chiara Rompietti, Francesco Maria Adamo, Loredana Ruggeri, and et al. 2021. "NK Cells in Chronic Lymphocytic Leukemia and Their Therapeutic Implications" International Journal of Molecular Sciences 22, no. 13: 6665. https://doi.org/10.3390/ijms22136665

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