Skip to main content

The role of exhausted natural killer cells in the immunopathogenesis and treatment of leukemia

Abstract

The immune responses to cancer cells involve both innate and acquired immune cells. In the meantime, the most attention has been drawn to the adaptive immune cells, especially T cells, while, it is now well known that the innate immune cells, especially natural killer (NK) cells, play a vital role in defending against malignancies. While the immune cells are trying to eliminate malignant cells, cancer cells try to prevent the function of these cells and suppress immune responses. The suppression of NK cells in various cancers can lead to the induction of an exhausted phenotype in NK cells, which will impair their function. Recent studies have shown that the occurrence of this phenotype in various types of leukemic malignancies can affect the prognosis of the disease, and targeting these cells may be considered a new immunotherapy method in the treatment of leukemia. Therefore, a detailed study of exhausted NK cells in leukemic diseases can help both to understand the mechanisms of leukemia progression and to design new treatment methods by creating a deeper understanding of these cells. Here, we will comprehensively review the immunobiology of exhausted NK cells and their role in various leukemic malignancies.

Video Abstract

Introduction

Regarding the increased prevalence of hematopoietic malignancies and the existence of difficulties in treatment, it is essential to study the etiology and immunopathogenesis of blood cancers, especially leukemias. Despite all the progress achieved, chemotherapy is the main therapeutic strategy for almost all hematopoietic malignancies which magnifies the importance of identifying novel and efficacious therapeutic targets [1, 2].

Both innate and adaptive immune responses are critical in the defense against cancer cells. Although it is generally supposed that adaptive immune cells, particularly T cells, are an essential part of the immune response against cancer cells, natural killer (NK) cells also play a critical role in the defense against malignant cells. They are the most critical innate immune lymphocytes in defense against infections and cancers. While impaired cytotoxic function of NK cells is correlated with cancer progression, the upregulation of activating receptors on NK cells is correlated with better disease prognosis [1, 2]. Similarly, the accumulation of functional NK cells in the tumor microenvironment has also been associated with low grades of cancer [3, 4].

Cancer cells can induce exhaustion in NK cells by changing the phenotype and function of NK cells and suppressing their anti-tumor function. The term "exhaustion" was initially used for T lymphocytes, wherein these cells undergo phenotypic changes and functional impairment following repeated exposure to antigens under pathological conditions, such as cancer or chronic infection. Induction of this state is associated with extensive alterations in T lymphocytes, including the induction of the inhibitory immune checkpoints, metabolic changes, epigenetic changes, and changes in molecular signaling pathways [5]. It should be noted that the exhaustion process and features are different in T and NK lymphocytes, which may be due to the fundamental differences between these two cells. While T cells have very diverse antigen receptors and identify antigens depending on MHC molecules [6, 7].

The evidence indicates that the occurrence of exhausted NK cells in leukemic patients has significantly increased, and their anti-leukemic activity has been dramatically inhibited [8, 9]. On the other hand, leukemia treatments based on targeting these cells have been associated with promising results, which indicate the introduction of a new treatment method. It seems necessary to mention that using NK cells in treating hematopoietic malignancies has been more successful than solid tumors. This issue is probably due to the difficulty of infiltrating NK cells to the tumor site and exposure to inhibitory signals in the tumor microenvironment, suppressing NK cell activity and inducing their exhaustion [10].

Despite efforts, many ambiguous and unknown issues in this field still require detailed and comprehensive studies. In this review article, we will try to discuss the immunobiology of NK cells, exhaustion of NK cells, the role of exhausted NK cells in leukemia, and targeting these cells for the treatment of leukemia.

Immunobiology of NK cells

NK cells are the most important components of the innate immune system that originate from the bone marrow and are present in the bloodstream and tissues such as lymph nodes, liver, thymus, and uterus [11].

Different subtypes of NK cells have been identified in humans and mice. The expression of molecules such as NK.1, CD49b, and NKp46 determine NK cells in mice. In humans, the CD16+CD56+ phenotype represents these cells [12]. This classification is partly derived from the differentiation stages of these cells. Accordingly, four stages of differentiation have been observed in murine NK cells. Primary immature NK cells have a CD27CD11b phenotype. In the second stage, these cells obtain the expression of molecules such as CD27, NK1.1, NKp46, and NKGD2. The third step starts with the expression of CD27, CD11b, and S1P5 molecules on the cell surface. In the fourth stage, mature cells will have a CD27CD11b+KLRG1+ phenotype that exhibits cytotoxic function.

On the other hand, the differentiation of NK cells in humans includes five stages. In the first stage in the bone marrow, pre-NK cells originate from lymphoid progenitors. In the second stage, these cells express the IL-15 receptor to ensure their survival during this stage until stage four. CD3CD56brightCD16 cells are the product of the fourth stage of differentiation, which resides mainly in the lymph nodes, produces a high amount of cytokines, and has low toxicity. The outcome of the fifth stage of differentiation is CD3CD56dimCD16+ cells, which are mainly present in blood circulation or inflammatory tissues. These cells exert cytotoxic function through the production of perforin and interferon (IFN)-gamma [13, 14].

According to another classification, within CD56dim cells, there are two types, including conventional NK cells and adaptive NK cells [15]. Adaptive NK cells have a different metabolic profile and epigenetic characteristics similar to effector CD8+ T cells [16]. These cells are long-lived; many express CD94/NKG2C [17]. These cells, mainly identified in mice, can survive over six months and self-renew. So far, three categories of adaptive NK cells have been identified, including hepatic-liver-resident NK, cytokine-induced memory-like NK, and cytomegalovirus (CMV)-specific NK (Specific NK cells against CMV) [18]. Adaptive NK cells exhibit antigenic specificity levels and memory recall properties. While the lifespan of conventional NK cells is less than ten days, adaptive NK cells can survive for months and even years [19].

Functionally, NK cells have a high ability to identify and kill virus-infected cells and transformed cells. These cells do not need prior exposure to the antigen to recognize the target and identify antigens through germline-encoded receptors. NK cells express various inhibitory and activating receptors, whose interaction with different ligands leads to activating or inhibiting their activity following target cell recognition. It is essential to mention that in the case of the simultaneous connection of activating and inhibitory receptors with their ligands, inhibitory signals will dominate, probably to maintain homeostasis and prevent self-directed responses [20]. NK cells express several inhibitory receptors, including T-cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domains (TIGIT), programmed death protein 1 (PD-1), T-cell immunoglobulin and mucin domain-containing protein 3 (TIM3), CD96, CD112R, interleukin (IL)-1R8, NKG2A, killer-cell immunoglobulin-like receptors (KIRs), and lymphocyte-activation gene 3 (LAG-3). The activating receptors of these cells also include NKG2, NKG2D, and CD226 [21]. NK cells express many inhibitory checkpoints of T cells, which led many researchers to think that it is possible to enhance the activity of NK cells by inhibiting these checkpoints and preventing exhaustion, similar to what has been experienced with T lymphocytes.

NK cells use a variety of mechanisms to activate and kill virus-infected or transformed cells. Identifying cells that lack MHC I molecules can lead to the non-activation of inhibitory checkpoints and, as a result, the dominance of NK cell activating receptors.Antibody-dependent cell-mediated cytotoxicity (ADCC) is another important mechanism of NK cell activation, in which NK cells bind to antibody-mediated opsonized particles through CD16 receptor. Moreover, some inflammatory cytokines can also activate NK cells [22, 23].

NK cell exhaustion

Exhaustion of immune cells is a state in which immune cells are defective regarding function and proliferative capacity. Exhaustion associated with a significant transcriptional profile alteration which is a consequence of extensive phenotypic, metabolic, and epigenetic alterations. Exhaustion primarily transpires in the presence of antigens and through recurrent stimulation, exemplified by chronic infections and malignancies. Considering the lifespan of NK cells, it seems that the exhaustion is mainly related to adaptive NK cells, which have long-term survival and can be chronically and repeatedly exposed to infectious or tumor antigens. However, there is evidence indicating that conventional natural killer (NK) cells can also experience an exhaustion process following repeated stimulation with cytokines or infectious agents [24]. As mentioned earlier, the exhaustion of NK cells is associated with various phenotypic, metabolic, functional, and epigenetic changes, which we describe below (as shown in Fig. 1).

Fig. 1
figure 1

Phenotypic and functional properties of NK cells in cancer. While immunosuppressive cells or cytokines, exosomes, and hypoxia promote exhaustion of NK cells, chemotherapy, stimulatory cytokines, blockade of immune checkpoint molecules, and stimulating activating receptors prevent exhaustion

The augmentation of the expression of inhibitory receptors serves as a critical marker for the state of exhaustion in natural killer cells. Accordingly, increased expression of the PD-1 molecule is considered one of the most critical indicators of exhausted cells in both T and NK cells. The PD-1 molecule is not only an indicator of exhausted NK cells, but its signaling plays a significant role in the exhaustion process [25]. The upregulation of PD-1 in CD56dimNKG2AKIR+CD57+ NK cells has been observed in both solid tumors and hematopoietic malignancies. These elevated PD-1 levels were accompanied by reduced cytokine secretion, defective degranulation, and decreased proliferation [1, 26, 27].

It should be noted that some studies have suggested that the expression of TIGIT and, to a lesser extent, LAG-3 and TIM-3 are more critical compared to PD-1 as an indicator of NK cell exhaustion [28, 29]. Another molecule whose signaling is mentioned as one of the influential factors in inducing NK exhaustion is NKG2A. CD56dim NKG2Ahigh NK cells were also increased in hepatocellular carcinoma patients, which was associated with a poor prognosis [30]. KIR receptors family and CD96 are other inhibitory molecules increased in exhausted NK cells.

Moreover, the decrease in the expression of stimulatory receptors is correlated with the exhaustion of NK cells. There has been a regular observation of the diminished presence of the stimulatory receptor NKG2D in different types of solid tumors or leukemia [31]. The reduced expression of NKG2D was also correlated with the decreased DAP10 expression [32]. Furthermore, decreased expression of several other NK cell activating receptors, including NKp30, NKp44, NKp46, CD16, 2B4, and CD226, has also been demonstrated in cancer patients, associated with poor prognosis [1]. Given that the equilibrium between the signals obtained from the inhibitory and activating receptors of NK cells will ultimately dictate the outcome of cellular activity, it appears that the decrease in expression levels of activating receptors in individuals with cancer results in the promotion of inhibitory signals prevailing in these cells.

From a functional point of view, it has been determined that NK cells have an incomplete function in a tumor environment or chronic infection and lose the ability to perform cytotoxic activities. For example, cancer progression in murine models is associated with the decreased frequency and function of NK cells [33, 34]. Furthermore, adoptively transferred NK cells to leukemic mice also lost their cytotoxic ability after exposure to malignant cells [22]. Also, tumor-infiltrating NK cells exhibit impaired cytotoxic functions, which is in part related to the downregulation of IFN-γ, FasL, perforin, CD107a, granzyme B, and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) [30, 31].

In addition to phenotypic and functional modifications, there seem to be some transcription profile changes in exhausted NK cells in the tumor area. It has been reported that transcription factors T-bet and eomesodermin (Eomes), which play an essential role in NK cells' activity, differentiation, and maturation, are significantly reduced in exhausted NK cells [22, 35]. Adoptively transferred NK cells to the murine leukemia model exhibited the reduced expression of these transcription factors, which was associated with impaired NK cell function. Interestingly, the induction of Eomes expression in these cells partially restored the function of NK cells [22]. This finding shows that the reduction of Eomes indicates exhaustion in NK cells and part of its induction process.

To understand the induction of NK cell exhaustion, it is necessary to examine the influential factors implicated in this process. One of the influential factors in NK exhaustion is the immunosuppressive microenvironment of the tumor. The most important immunosuppressive cells include regulatory T cells (Tregs) [36], myeloid-derived suppressor cells (MDSCs) [37], tumor-associated fibroblasts [38], tumor-associated neutrophils [39], and tumor-associated macrophages [40]. The upregulation of immunosuppressive cells in cancer patients is associated with decreased frequency and function of NK cells. These inhibitory cells can inhibit NK cells and induce exhaustion with various mechanisms dependent on cell–cell contact or independent of cell attachment (through secretion of immunosuppressive factors) [41,42,43]. Immune inhibitory cytokines such as transforming growth factor-β (TGF-β) and IL-10 are also essential in NK cell exhaustion [44, 45]. The hypoxic condition of the tumor microenvironment is an important factor in the suppression and exhaustion of NK cells. The hypoxia downregulates NKG2D, NKp46, NKp30, and NKp44 in NK cells through the expression of hypoxia-inducible factor 1α (HIF-1α) [46, 47].

The upregulation or downregulation of certain factors on cancer cells, which act as ligands for inhibitory or activating receptors of NK cells, is considered to be one of the most crucial elements that lead to NK cell exhaustion. As mentioned, the balance between the signals received from activating and inhibitory receptors of NK cells will determine their function. This balance will be towards inhibitory signals in the vicinity of malignant cells. Numerous investigations have demonstrated the notable presence of diverse molecules on the surface of malignant cells, which serve as the ligands for the suppressive receptors of NK cells. Consequently, this phenomenon has resulted in the impairment of NK cells and, ultimately, their exhaustion. For example, the increased expression of CD200 and galectin-9 on cancer cells (ligands of CD200R and TIM-3 on NK cells, respectively) has been demonstrated in acute myeloid leukemia (AML) [48, 49]. On the other hand, the downregulation of CD48 on leukemic cells (the ligand for activating receptor 2B4 on NK cells, was observed in AML [50].

Finally, exosomes secreted from cancer cells are also effective factors in the inhibition and exhaustion of NK cells. Exosomes with different mechanisms such as the secretion of inhibitory cytokines such as TGF-β, providing microRNAs involved in NK suppression or the expression of inhibitory receptor ligands of NK cells can lead to defects in the cytotoxic activity of these cells and facilitate the exhaustion process [51, 52].

Exhausted NK cells and leukemia

The inactivation of NK cells and their failure to eradicate leukemic cells is a prevailing occurrence noted in nearly all leukemic malignancies. In this section, we will review the studies on the inefficiency of NK cells in various leukemias, the mechanisms of NK exhaustion, and the therapeutic strategies used to strengthen NK cells.

CLL

The ability of ammonium chloride to inhibit the cytotoxic activity of NK cells cultured with the leukemia cell line K562 (chronic myelogenous leukemia) was one of the first experiences related to the inhibition of these cells in leukemia. This suppression was reversible (after 15 h) and dose-dependent [53].

One of the most common ways to study exhausted NK cells is to examine their frequency in patients and their correlation with disease prognosis. Accordingly, an increased frequency of exhausted NK cells has been reported in chronic lymphocytic leukemia (CLL) patients. By studying the peripheral blood of 24 CLL patients and 19 normal individuals, Hadadi et al. have shown that the frequency of CD56+CD3Tim-3+ cells has increased significantly in these patients, which was associated with the downregulation of CD56+CD3NKp30+ cells. They indicated that these dysregulated frequencies were correlated with poor prognostic factors such as high absolute lymphocyte count, decreased hemoglobin, and elevated serum C reactive protein (CRP) concentration [54].

Some studies have shown the effect of some common treatments on NK cell exhaustion in leukemia patients. For example, the anti-CD20 monoclonal (ofatumumab or rituximab) antibody could promote NK cell exhaustion by binding to CD16 on NK cells. This interaction could impair the cytotoxic activity of NK cells. Following this interaction, some NK cells activating receptors such as NKp46, NKG2D, 2B4, and DNAM-1 could not phosphorylate essential signaling molecules involved in the cytotoxic function of NK cells, such as phospholipase C (PLC)γ2, SH2-domain-containing leukocyte protein of 76 kDa (SLP-76), and Vav1. Furthermore, this ligation could also recruit the inhibitory phosphatase Src homology region 2 domain-containing phosphatase-1 (SHP-1) to the cytoplasmic tail of CD16, leading to further suppression of NK cells. These findings interestingly show the dual role of the CD16 receptor in the activation or inhibition of NK cells and provide a new mechanism for the exhaustion of these cells because the pharmacological blockade of this receptor led to the recovery of the cytotoxic activity following binding to anti-CD20 antibody [55]. Although these findings imply that rituximab can inhibit some anti-leukemia responses, it should be evaluated in further studies to evaluate its advantages and disadvantages.

On the contrary, there exists information suggesting that certain therapies can impede the exhaustion of NK cells and enhance the cytotoxic capabilities of these cells. Accordingly, NK cells derived from healthy subjects after treatment with drugs such as sunitinib, sorafenib, or the pan-RAF inhibitor ZM336372 had a high ability to secrete cytokines and kill target cells and prevent exhaustion in the RAS/RAF/ERK signaling pathway-dependent manner [56]. Similarly, treatment of 55 relapsed/refractory and 50 untreated CLL patients with Ibrutinib was associated with increased effective NK cells compared to 20 normal subjects [57]. Likewise, during four years of follow-up of 31 CLL patients and 20 normal subjects, Ibrutinib could enhance the frequency and function of NK cells, which was associated with good prognosis [58].

The expression of checkpoints on NK cells and their engagement with the corresponding receptors on leukemic cells stands as a highly crucial means by which NK cells experience suppression and exhaustion. Likewise, the endeavor to target inhibitory checkpoints on NK cells has been recognized as a preeminent immunotherapeutic approach for targeting exhausted NK cells. The study of circulating NK cells in 17 CLL patients showed that leukemic B cells express significantly higher levels of Siglec-7 ligands than normal individuals, which was associated with poor disease prognosis. Moreover, the blockade of the Siglec-7 ligand markedly enhanced the sensitivity of leukemic cells to the cytotoxic effects of NK cells [59]. In another study, the expression of LAG3 was significantly increased in leukemic and NK cells of 61 untreated CLL patients, which was associated with a poor prognosis. In addition, the blockade of LAG3 using monoclonal antibodies significantly preserved the cytotoxic function of NK cells against leukemic cells [60]. B and T lymphocyte attenuator (BTLA) is another inhibitory checkpoint its upregulation is detected on both leukemic and NK cells in 46 untreated CLL patients associated with a poor prognosis. Interestingly, ex vivo blockade of BTLA led to the depletion of leukemic cells and enhanced cytotoxic function and cytokine secretion by NK cells [61]. Human inhibitory receptors Ig-like transcript 2 (ILT2) (also known as LIR-1 or LILRB1) is also an immune checkpoint investigated in CLL patients regarding its role in suppressing NK cells. While B-CLL cells exhibited low levels of this ILT2, it was upregulated in the NK cells of CLL patients (n = 60) compared to normal individuals (n = 25), which was associated with the severity of disease. It has been reported that inhibiting ILT2 with Lenalidomide significantly activates NK cells and eliminates leukemic cells [62]. In contrast, blockade of PD-1 and TIM-3 checkpoints in NK cells derived from the peripheral blood of 18 early-stage CLL patients did not affect the cytotoxic function and secretion of tumor necrosis factor (TNF)-α and IFN-γ [63].

As we reviewed in this section (summarized in Table 1), despite the interesting clues regarding the role of exhausted NK cells in CLL patients, there are still many unknowns in this field. In future studies, different subtypes of exhausted NK cells should be investigated in patients with varying degrees of disease progression. Future investigations should comprehensively address the differences between peripheral blood and bone marrow NK cells. Also, many other checkpoints have not yet been investigated in CLL patients, which should be comprehensively evaluated. Furthermore, the efficacy of targeting exhausted NK cells in CLL patients as a therapeutic target should be precisely investigated in preclinical and clinical studies.

Table 1 Studies related to the role of exhausted NK cells in CLL

AML

Among the various malignancies of leukemia, the majority of research regarding the significance of exhausted NK cells has been conducted in patients with AML. A particular investigation within this context investigated the impact of genetic polymorphism in the coding sequences of receptors responsible for the inhibition or activation of NK cells, and its potential association with the susceptibility to AML. While AML patients (n = 169) exhibit high expression of KIR activating receptors, especially KIR3DS1, normal subjects (n = 167) express KIR inhibitory receptors, especially KIR2DL1 and KIR3DL1. It has been proposed that the increased expression of KIR-activating receptors in patients probably leads to the hyperactivation of these cells following the encounter with leukemic cells and further the exhaustion of NK cells and the progression of cancer [64]. Interestingly, some fungal infections, such as A. fumigatus, promote NK cell exhaustion in AML patients leading to their reduced cytotoxic function against leukemic cells. Exhaustion was associated with the reduced secretion of inflammatory cytokines such as TNF-α, IFN-γ, regulated upon activation, normal T cell expressed and presumably secreted (RANTES), macrophage inflammatory protein (MIP)-1α, and MIP-1β, decreased expression of activating receptors such as NKG2D and NKp46, and impaired NK cell degranulation [65]. Therefore, some infections in leukemic patients can also be considered one of the causes of NK cell exhaustion.

The frequency and absolute number of peripheral CD56+CD16+ NK cells were also markedly decreased in myelodysplastic syndromes (MDS) and AML secondary to MDS patients (n = 130) compared to normal subjects (n = 40). It was also shown that NK cells expressing NKG2D, NKp46, and CD161 were significantly reduced in patients compared to normal subjects [66]. Zeng and colleagues also proposed that NK cells in the peripheral blood of AML patients are in an exhaustion state. In contrast, NK cells in the bone marrow mainly have a terminally differentiated phenotype, which correlates with low patient survival. They showed that the frequency of CD16CD56dim NK cells is reduced in the peripheral blood of novo AML and AML patients with complete remission after chemotherapy, whereas there was no change in the bone marrow. In addition, while the frequency of killer cell lectin-like receptor G1 (KLRG1)- and TIGIT-expressing NK cells was increased in peripheral blood of novo patients, it was recovered in patients after chemotherapy. Moreover, the frequency of terminally differentiated NK cells (CD56dimCD16+CD57+), which had a potent cytotoxic function and low replication capacity, was increased in the bone marrow of novo AML patients and correlated with a lower survival rate [67]. Tang et al. also demonstrated that peripheral NK cells of AML patients (n = 79) had extensive functional impairments, which were correlated with disease relapse and resistance to treatment. Also, in patients who responded to chemotherapy, the functional responses of NK cells were restored [68]. By studying the bone marrow samples of 37 newly diagnosed AML patients, it was found that the expression of TIM-3 checkpoint on NK cells and blasts can be used as a prognosis marker [69]. The review of these three studies shows that the exhaustion status of NK cells in AML patients with various degrees of progression is different. Another point that can be taken from these studies is the difference between exhausted NK cells in peripheral blood and bone marrow. Finally, it seems that the exhaustion status in NK cells is reversible, and after treatment with drugs such as chemotherapy, the activity of these cells returns to normal conditions.

Bou-Tayeh and coworkers demonstrated that the deregulation of signaling pathways induced by cytokines in NK cells of AML patients might constitute one of the mechanisms implicated in the exhaustion process. They provided evidence that the advancement of AML correlated with impairments in the maturation and functionality of NK cells in murine models of AML. While the IL-15/mammalian target of rapamycin (mTOR) and type I interferon (IFN) signaling pathways were constitutively active in NK cells purified from the leukemia murine model, these cells could not respond to stimulation with IL-15, in vitro. Therefore, chronic activation of NK cells through the IL-15/mTOR pathway is one of the NK cell exhaustion mechanisms in AML models. Similar results were observed in NK cells derived from AML patients in vitro. There were low expression levels of IL-2/15Rβ and decreased response to stimulation with IL-15 in these cells [70].

Targeting checkpoint molecules has been introduced as an effective immunotherapy method for targeting exhausted NK cells. Recently, poliovirus receptor-related immunoglobulin domain-containing (PVRIG) has been introduced as one of the new effective checkpoints inhibiting NK cells' activity, especially CD56bright cells. By studying the blasts of 20 AML patients, it has been shown that these cells continuously express the ligand of this checkpoint, poliovirus receptor-related 2, PVRL2. Interestingly, blockade of the PVRIG/PVRL2 axis using an anti-PVRIG antibody markedly activated NK cells to kill PVRL2-expressing leukemic cells. In contrast to peripheral NK cells, bone marrow NK cells had no upregulated PVRIG expression. Activation of NK cells following ligation of activating receptors (NKp46 and CD16), leukemic cell recognition, or cytokine stimulation (IL-12 and IL-2) could suppress the expression of PVRIG in NK cells [71].

PD-1/PD-L1 axis is another checkpoint that was well investigated in various cancers. An exciting recently published finding showed that NK cells in contact with leukemic cells in the C1498 murine AML model acquire the PD-1 molecule from leukemic cells through the Trogocytosis mechanism in a SLAM receptor-mediated manner. Trogocytosis denotes a biological phenomenon characterized by the physical extraction and ingestion of cellular material, referred to as "bites," from one cell by another cell [72]. Therefore, PD-1 checkpoint expression on NK cells seems intrinsic and can originate from leukemic cells.

Moreover, exhausted NK cells are worthy targets for the treatment of PD-L1 tumors. Dong and coworkers showed that the ameliorative effect of anti-PD-L1 antibody in PD-L1 AML (n = 79) is related in part to the targeting of PD-L1+ exhausted NK cells. Malignant cells induce PD-L1 in NK cells through the AKT signaling pathway. The impact of anti-PD-L1 antibody on NK cells is also through the P38 pathway. Furthermore, the combination use of anti-PD-L1 antibody and NK cell-stimulating cytokines had a more significant impact on leukemic cells compared to monotherapy [73]. These findings imply that the rationale for using an anti-PD-L1 antibody in PD-L1 negative tumors can be targeting exhausted NK cells. The mouse model of AML has yielded comparable findings with regard to the inhibition of PD-L1, underscoring the significance of checkpoint inhibition in averting NK cell exhaustion and enhancing their efficacy against leukemia [74].

Evaluation of NK cells in peripheral blood of 100 AML patients showed increased expression of B7-H3 checkpoint, which was associated with poor prognosis. Also, inhibition of this checkpoint using monoclonal antibody had a significant impact on the cytotoxic function of NK cells, both in vitro (in HL-60, Kasumi-1, THP-1, MV4-11, MOLM-13, U937, OCI-AML3, OCI-AML2, and MOLM-14 cells) and in vivo. Treatment of AML patient-derived xenografts with an anti-B7-H3 antibody had similar results [75].

Another checkpoint that plays a role in inhibiting the function of NK cells is TIGIT, which binds to its ligands, including CD155 and CD112, on leukemic cells. In an in vitro study conducted on AML cell lines (MOLM-13, MV-4–11, NB-4, THP-1, and KG-1), inhibition of CD155/CD122 on leukemic cells by Flt3 inhibitors enhanced the cytotoxic function of NK cells against these cells [76]. TIGIT was also upregulated on NK cells and bone marrow samples in AML patients following allogeneic transplantation, which was associated with the downregulation of NK cells in the bone marrow [77]. Likewise, blockade of TIGIT, CD39, and A2AR checkpoints on NK cells purified from peripheral blood (n = 15) and bone marrow (n = 25) of AML patients increased the cytotoxic function of NK cells [78].

Furthermore, the examination of TIM-3 expression in NK cells in 47 newly diagnosed AML patients showed that the expression of this checkpoint is associated with a poor prognosis and has prognostic significance [79]. In a similar study, newly diagnosed AML patients (n = 23) had a higher frequency of TIGIT+PD-1+TIM3+ NK cells compared to normal subjects, which was associated with poor prognosis. The expression of these checkpoints was associated with low cytotoxicity, and their inhibition increased the function of NK cells [80]. In contrast, in a study on 150 AML patients, Rakova and coworkers reported that TIM-3 expression in NK cells correlates with high functional capacity and better clinical outcomes. Moreover, in contrast to M1 and M2 patients, M4 and M5 patients had a lower frequency of NK cells compared to normal subjects. They also showed intact TIGIT and significant downregulation of PD-1 in patients-derived NK cells compared to healthy donors. Intriguingly, the blockade of TIM-3 (but not PD-1 and TIGIT) inhibited the secretion of IFN-γ in NK cells following stimulation [81].

The use of anti-NKG2A antibodies in the mouse model of AML led also to significant activation of NK cells and tumor regression [82].

In addition to the inhibitory receptors, investigating the activating receptors of NK cells can also be considered a solution to prevent the exhaustion of these cells and increase their anti-leukemic activity. Accordingly, by studying 111 AML patients, it has been determined that the interaction of the OX40 molecule on leukemic cells with the OX40L checkpoint causes the activation of NK cells and eliminates leukemic cells [83].

The design and production of CAR-NK cells is another advanced and innovative method of treating leukemia. Gurney et al., using transposon-engineered CAR-NK cells, could efficiently target and destroy AML cell lines with expression of CLL-1 molecule and primary AML cells, even leukemia stem cells. They further engineered CAR-NK cells by silencing NK cell cytokine checkpoint cytokine-inducible SH2-containing protein (CIS) using the CRISPR/Cas9 system, which was associated with increased cytotoxicity of these cells [84].

Taken together, it seems that the progression of AML is associated with an increase in the frequency of exhausted NK cells, accompanied by a high expression of inhibitory checkpoints and a decrease in the expression of activating receptors and functional defects in these cells (Table 2). Also, although the results of the studies are not sufficient and comprehensive, it seems that exhausted NK cells in the peripheral blood and bone marrow of AML patients do not have similar conditions; however, this field should be studied comprehensively. The results of the studies that have used the inhibition of NK cell checkpoints for treating AML are promising. Still, they should be evaluated in more detail in mouse models and clinical efficacy.

Table 2 Studies related to the role of exhausted NK cells in AML

Acute lymphocytic leukemia (ALL)

In a study conducted by Duault et al. on many B-ALL and T-ALL patients, they suggested that the characterization of NK cells can be used to predict patients' clinical outcomes. They showed that although the cytotoxic function of NK cells in leukemic patients is significantly reduced compared to normal controls, activation markers such as high expression of CD69 and CD56 molecules, production of cytokines, and Calcium signaling are increased. The results also showed that the incomplete maturation of NK cells into effector cells prevents the lysis of leukemic cells by NK cells. They proposed that chronic activation may lead to NK cell exhaustion in ALL patients. The increase of cytokine-producing activated NK cells was associated with disease progression and independently indicated a poor prognosis for ALL [85].

Interestingly, NK cells in the bone marrow and peripheral blood samples of leukemic patients had a similar status. In a study that Bailur and colleagues conducted on bone marrow samples of 35 B-ALL and 26 AML patients, they showed that the activity of NK cells was significantly impaired compared to normal subjects (n = 11). While CD16+CD57+ NK cells were significantly decreased in AML patients, these cells did not change in B-ALL patients. Also, while granzyme secretion and NKG2D expression were reduced in AML patient cells and TIM3 expression was increased, NK cells of B-ALL patients were not different from normal individuals [86]. Moreover, by conducting next-generation sequencing and evaluating gene expression profiles and gene polymorphisms, it has been demonstrated that B-ALL and T-ALL might differentially regulate NK cell exhaustion [87].

In ALL patients, the tumor microenvironment in the bone marrow has been mentioned as one of the influential factors in inducing NK cell exhaustion. Ramírez-Ramírez and colleagues have suggested that molecular CRTAM/Necl-2 interaction in bone marrow niches leads to NK cell exhaustion. They reported that lymphoid progenitor cells in the bone marrow with phenotype CD34+CD56+CD3+CD19+CRTAM+ are likely to be in the first activation phase and are adjacent to niches with high expression of the ligand nectin-like-2. On the other hand, the bone marrow of ALL patients had a high abundance of CD56highCRTAM+ NK cells with an exhausted phenotype, which was functionally defective and could produce IL-10 and TGF-β [88].

The phenotypic and functional properties of CD56 NK cells have also been investigated in ALL and chronic myelogenous leukemia (CML) patients treated with dasatinib by Ishiyama and colleagues. This study was conducted on 36 CML or Ph + ALL (Philadelphia chromosome-positive ALL), 26 imatinib or nilotinib‐treated patients, and 15 normal subjects. Their results showed that the frequency of CD56 NK cells was exclusively augmented in dasatinib-treated patients who were cytomegalovirus‐seropositive, and expansion of these cells was accompanied by upregulation of specific NK cells against CMV. The expression of differentiation and activation markers such as CD57, NKG2D, NKG2C, NKp46, NKp30, perforin, and granzyme B was significantly reduced in CD56 NK cells. A comparison of NK cells showed that the characteristics of CD56 and CD56dim cells were relatively similar, but these two cell groups were completely different from CD56bright cells; however, it should be noted that the functional properties of CD56 NK cells were significantly lower than CD56dim cells. Also, inhibition of PD-1 could increase the activity of NK cells, especially CD56dim cells, which was proportional to the expression level of PD-1. This study demonstrated that expansion of CD56PD-1+ NK cells indicates chronic activation of NK cells in dasatinib-treated patients who were cytomegalovirus‐seropositive. Further, combination therapy by blockade of the PD-1/PD-L1 axis and dasatinib was suggested as a potential treatment approach for leukemia [89].

A study on leukemic cells in 5 patients with B-cell precursor ALL (BCP-ALL) also showed that microRNA582 inhibits the cytotoxic effects of NK cells on leukemic cells by inducing the CD276 (B7-H3) checkpoint. Therefore, it is suggested that blockade of CD276 or its ligand on NK cells may be a potential therapeutic approach in leukemia [90]. In addition to the discussed inhibitory checkpoints, Rothfelder et al., by studying 44 ALL patients and ALL cell lines (NALM-16, JURKAT, SD-1, REH, SUP -B15, TOM-1, and), have shown that the expression of the OX40L checkpoint on NK cells and its interaction with OX40 on ALL cells causes the activation of NK cells and depletion of Leukemic cells [91].

The use of cytokines that stimulate the activity of NK cells has been proposed as one of the exciting immunotherapy methods that prevent their exhaustion by activating the cytotoxic activity of these cells. Accordingly, a study conducted on a 70Z/3 murine pre-B cell leukemia model (murine model of immune-mediated rejection of the acute lymphoblastic leukemia) showed that treating mice by injecting IL-15-secreting leukemic cells induces and activates NK1.1+ cells, accompanied by increased mouse survival time [92].

Few studies have been conducted regarding the exhaustion of NK cells in ALL patients, which causes the lack of accurate conclusions from the results (Table 3). More studies are needed regarding the expression and role of different factors in the exhaustion of NK cells. Studies suggest that the progression of ALL is associated with the exhaustion of NK cells, and targeting exhaustion markers has been associated with promising results; however, there is a need for more comprehensive studies.

Table 3 Studies related to the role of exhausted NK cells in ALL

Conclusion

The importance of the anti-cancer function of NK cells in innate immunity has encouraged several investigators to investigate the phenotypic and functional characteristics of these cells in hematopoietic malignancies to evaluate their targeting worth for leukemia immunotherapy.

The phenotype and function of NK cells in almost all leukemia malignancies shifted towards exhausted cells, associated with a poor prognosis. Also, the presence and increase of exhausted NK cells were associated with a poor prognosis. Another point that can be mentioned about exhausted NK cells in leukemic malignancies is the importance of targeting them as a new treatment method. Except for a few exceptions, in almost all studies that targeted the inhibitory checkpoints of exhausted NK cells, all therapeutic studies showed that this treatment method is associated with the recovery of the anti-leukemic function of NK cells and the elimination of leukemic cells.

Regarding the commonality of NK cell exhaustion in different subtypes of leukemia, it seems that leukemia occurrence and progression are associated with the reduced frequency and function of NK cells, hallmarks of the exhaustion process. The upregulation of inhibitory checkpoints and reduced secretion of inflammatory cytokines and toxic mediators are other common NK cell exhaustion characteristics observed in the majority of studies.

Despite the above-indicated deduction, there are unknown issues related to exhausted NK cells in leukemic malignancies, which require comprehensive studies in the future. While most of the studies on exhausted NK cells have been conducted in AML patients, little is known regarding the immunobiology and targeting potential of these cells in other leukemic malignancies, especially CML.

The comparison of exhausted NK cells in the peripheral and bone marrow of leukemia patients is another important issue that should be further studied. In the few studies that have evaluated exhausted NK cells in both peripheral blood and bone marrow, comprehensive information has not been obtained. In some cases, there were even contradictory data. Therefore, extensive studies must investigate these cells in various leukemias in peripheral blood and bone marrow samples. Consideration of disease stage and previously received treatments are also critical issues that should be considered in studies since they can significantly affect the exhaustion process. The defect observed in many studies was that either a proper classification of patients was not provided or patients with different disease stages were not studied simultaneously.

A critical question that future studies should seek to answer is whether the development of cancer leads to the creation of exhausted cells or whether the emergence of exhausted NK cells is one of the reasons for cancer development. Available evidence supports both theories; although these two events may coincide, other factors cause these events. The observation that chemotherapy or anti-leukemia treatments that target the leukemic cells themselves restore the function of NK cells further strengthens the theory that other factors are involved in this issue. Several checkpoints have not yet been investigated in various leukemia diseases, which should be considered in future studies. Also, the molecular mechanisms of NK cell exhaustion have not yet been clearly defined and should be studied further studies.

Availability of data and materials

No data was used for the preparation of this manuscript.

References

  1. Bi J, Tian Z. NK cell exhaustion. Front Immunol. 2017;8:760.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Zhang Q, et al. Blockade of the checkpoint receptor TIGIT prevents NK cell exhaustion and elicits potent anti-tumor immunity. Nat Immunol. 2018;19(7):723–32.

    Article  CAS  PubMed  Google Scholar 

  3. Ishigami S, et al. Clinical impact of intratumoral natural killer cell and dendritic cell infiltration in gastric cancer. Cancer Lett. 2000;159(1):103–8.

    Article  CAS  PubMed  Google Scholar 

  4. Peng LS, et al. Tumor-Associated Monocytes/Macrophages Impair NK-Cell Function via TGFβ1 in Human Gastric Cancer. Cancer Immunol Res. 2017;5(3):248–56.

    Article  CAS  PubMed  Google Scholar 

  5. Merino AM, et al. Unraveling exhaustion in adaptive and conventional NK cells. J Leukoc Biol. 2020;108(4):1361–8.

    Article  CAS  PubMed  Google Scholar 

  6. Blank CU, et al. Defining ‘T cell exhaustion.’ Nat Rev Immunol. 2019;19(11):665–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Wu S-Y, et al. Natural killer cells in cancer biology and therapy. Mol Cancer. 2020;19(1):1–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Sun H, Sun C. The rise of NK cell checkpoints as promising therapeutic targets in cancer immunotherapy. Front Immunol. 2019;10:2354.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Liu X, et al. NK and NKT cells have distinct properties and functions in cancer. Oncogene. 2021;40(27):4521–37.

    Article  CAS  PubMed  Google Scholar 

  10. Russick J, et al. NK cells in the tumor microenvironment: Prognostic and theranostic impact. Recent advances and trends. Semin Immunol. 2020;48:101407. https://doi.org/10.1016/j.smim.2020.101407.

  11. Shimasaki N, Jain A, Campana D. NK cells for cancer immunotherapy. Nat Rev Drug Discovery. 2020;19(3):200–18.

    Article  CAS  PubMed  Google Scholar 

  12. Morvan MG, Lanier LL. NK cells and cancer: you can teach innate cells new tricks. Nat Rev Cancer. 2016;16(1):7–19.

    Article  CAS  PubMed  Google Scholar 

  13. Perera Molligo Adarachchige AS. Human NK cells: From development to effector functions. Innate immunity. 2021;27(3):212–29.

    Article  Google Scholar 

  14. Zhang Y, Huang B. The development and diversity of ILCs, NK cells and their relevance in health and diseases. Regulation of Inflammatory Signaling in Health and Disease. 2017. p. 225–44.

    Google Scholar 

  15. Liu LL, et al. Critical Role of CD2 Co-stimulation in Adaptive Natural Killer Cell Responses Revealed in NKG2C-Deficient Humans. Cell Rep. 2016;15(5):1088–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Cichocki F, et al. ARID5B regulates metabolic programming in human adaptive NK cells. J Exp Med. 2018;215(9):2379–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Béziat V, et al. NK cell responses to cytomegalovirus infection lead to stable imprints in the human KIR repertoire and involve activating KIRs. Blood. 2013;121(14):2678–88.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Zhang C, Hu Y, Shi C. Targeting Natural Killer Cells for Tumor Immunotherapy. Front Immunol. 2020;11:60.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Roe K. NK-cell exhaustion, B-cell exhaustion and T-cell exhaustion-the differences and similarities. Immunology. 2022;166(2):155–68.

    Article  CAS  PubMed  Google Scholar 

  20. Martinet L, Smyth MJ. Balancing natural killer cell activation through paired receptors. Nat Rev Immunol. 2015;15(4):243–54.

    Article  CAS  PubMed  Google Scholar 

  21. Cao Y, et al. Immune checkpoint molecules in natural killer cells as potential targets for cancer immunotherapy. Signal Transduct Target Ther. 2020;5(1):250.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Gill S, et al. Rapid development of exhaustion and down-regulation of eomesodermin limit the antitumor activity of adoptively transferred murine natural killer cells. Blood. 2012;119(24):5758–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Chen Y, et al. Research Progress on NK Cell Receptors and Their Signaling Pathways. Mediators Inflamm. 2020;2020:6437057.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Alvarez M, et al. Regulation of murine NK cell exhaustion through the activation of the DNA damage repair pathway. JCI Insight. 2019;5(14):e127729. https://doi.org/10.1172/jci.insight.127729.

  25. Lee EHC, Wong DCP, Ding JL. NK Cells in a Tug-of-War With Cancer: The Roles of Transcription Factors and Cytoskeleton. Front Immunol. 2021;12:734551.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Beldi-Ferchiou A, et al. PD-1 mediates functional exhaustion of activated NK cells in patients with Kaposi sarcoma. Oncotarget. 2016;7(45):72961–77.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Wiesmayr S, et al. Decreased NKp46 and NKG2D and elevated PD-1 are associated with altered NK-cell function in pediatric transplant patients with PTLD. Eur J Immunol. 2012;42(2):541–50.

    Article  CAS  PubMed  Google Scholar 

  28. Judge SJ, Murphy WJ, Canter RJ. Characterizing the Dysfunctional NK Cell: Assessing the Clinical Relevance of Exhaustion, Anergy, and Senescence. Front Cell Infect Microbiol. 2020;10:49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. da Silva IP, et al. Reversal of NK-cell exhaustion in advanced melanoma by Tim-3 blockade. Cancer Immunol Res. 2014;2(5):410–22.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Sun C, et al. High NKG2A expression contributes to NK cell exhaustion and predicts a poor prognosis of patients with liver cancer. Oncoimmunology. 2017;6(1):e1264562.

    Article  PubMed  Google Scholar 

  31. Peng YP, et al. Comprehensive analysis of the percentage of surface receptors and cytotoxic granules positive natural killer cells in patients with pancreatic cancer, gastric cancer, and colorectal cancer. J Transl Med. 2013;11:262.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Sun C, et al. TGF-β1 down-regulation of NKG2D/DAP10 and 2B4/SAP expression on human NK cells contributes to HBV persistence. PLoS Pathog. 2012;8(3):e1002594.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Guillerey C, et al. Immunosurveillance and therapy of multiple myeloma are CD226 dependent. J Clin Invest. 2015;125(5):2077–89.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Paul S, et al. Intratumoral natural killer cells show reduced effector and cytolytic properties and control the differentiation of effector Th1 cells. Oncoimmunology. 2016;5(12):e1235106.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Simonetta F, Pradier A, Roosnek E. T-bet and Eomesodermin in NK Cell Development, Maturation, and Function. Front Immunol. 2016;7:241.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Jadidi-Niaragh F, et al. Regulatory T cells in chronic lymphocytic leukemia: implication for immunotherapeutic interventions. Tumor Biology. 2013;34:2031–9.

    Article  CAS  PubMed  Google Scholar 

  37. Yazdani Y, et al. Myeloid-derived suppressor cells in B cell malignancies. Tumor Biology. 2015;36:7339–53.

    Article  CAS  PubMed  Google Scholar 

  38. Salimifard S, et al. Cancer associated fibroblasts as novel promising therapeutic targets in breast cancer. Pathology-Research and Practice. 2020;216(5):152915.

    Article  CAS  PubMed  Google Scholar 

  39. Hajizadeh F, et al. Tumor-associated neutrophils as new players in immunosuppressive process of the tumor microenvironment in breast cancer. Life Sci. 2021;264:118699.

    Article  CAS  PubMed  Google Scholar 

  40. Yousefzadeh Y, et al. Tumor associated macrophages in the molecular pathogenesis of ovarian cancer. Int Immunopharmacol. 2020;84:106471.

    Article  CAS  PubMed  Google Scholar 

  41. Ghiringhelli F, et al. CD4+CD25+ regulatory T cells inhibit natural killer cell functions in a transforming growth factor-beta-dependent manner. J Exp Med. 2005;202(8):1075–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Li H, et al. Cancer-expanded myeloid-derived suppressor cells induce anergy of NK cells through membrane-bound TGF-beta 1. J Immunol. 2009;182(1):240–9.

    Article  CAS  PubMed  Google Scholar 

  43. Balsamo M, et al. Melanoma-associated fibroblasts modulate NK cell phenotype and antitumor cytotoxicity. Proc Natl Acad Sci U S A. 2009;106(49):20847–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Castriconi R, et al. Transforming growth factor beta 1 inhibits expression of NKp30 and NKG2D receptors: consequences for the NK-mediated killing of dendritic cells. Proc Natl Acad Sci U S A. 2003;100(7):4120–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Donatelli SS, et al. TGF-β-inducible microRNA-183 silences tumor-associated natural killer cells. Proc Natl Acad Sci U S A. 2014;111(11):4203–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Sarkar S, et al. Hypoxia induced impairment of NK cell cytotoxicity against multiple myeloma can be overcome by IL-2 activation of the NK cells. PLoS ONE. 2013;8(5):e64835.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Balsamo M, et al. Hypoxia downregulates the expression of activating receptors involved in NK-cell-mediated target cell killing without affecting ADCC. Eur J Immunol. 2013;43(10):2756–64.

    Article  CAS  PubMed  Google Scholar 

  48. Coles SJ, et al. CD200 expression suppresses natural killer cell function and directly inhibits patient anti-tumor response in acute myeloid leukemia. Leukemia. 2011;25(5):792–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Ndhlovu LC, et al. Tim-3 marks human natural killer cell maturation and suppresses cell-mediated cytotoxicity. Blood. 2012;119(16):3734–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Zhang T, et al. Heme oxygenase 1 overexpression induces immune evasion of acute myeloid leukemia against natural killer cells by inhibiting CD48. J Transl Med. 2022;20(1):394.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Clayton A, et al. Human tumor-derived exosomes down-modulate NKG2D expression. J Immunol. 2008;180(11):7249–58.

    Article  CAS  PubMed  Google Scholar 

  52. Szczepanski MJ, et al. Blast-derived microvesicles in sera from patients with acute myeloid leukemia suppress natural killer cell function via membrane-associated transforming growth factor-beta1. Haematologica. 2011;96(9):1302–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Brander C, et al. Inhibition of human NK cell-mediated cytotoxicity by exposure to ammonium chloride. J Immunol Methods. 2001;252(1–2):1–14.

    Article  CAS  PubMed  Google Scholar 

  54. Hadadi L, et al. Dysregulated Expression of Tim-3 and NKp30 Receptors on NK Cells of Patients with Chronic Lymphocytic Leukemia. Oncol Res Treat. 2019;42(4):202–8.

    Article  PubMed  Google Scholar 

  55. Capuano C, et al. Anti-CD20 Therapy Acts via FcγRIIIA to Diminish Responsiveness of Human Natural Killer Cells. Cancer Res. 2015;75(19):4097–108.

    Article  CAS  PubMed  Google Scholar 

  56. Lohmeyer J, et al. Sorafenib paradoxically activates the RAS/RAF/ERK pathway in polyclonal human NK cells during expansion and thereby enhances effector functions in a dose- and time-dependent manner. Clin Exp Immunol. 2018;193(1):64–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Solman IG, et al. Ibrutinib restores immune cell numbers and function in first-line and relapsed/refractory chronic lymphocytic leukemia. Leuk Res. 2020;97:106432.

    Article  CAS  PubMed  Google Scholar 

  58. Solman IG, et al. Impact of long-term ibrutinib treatment on circulating immune cells in previously untreated chronic lymphocytic leukemia. Leuk Res. 2021;102:106520.

    Article  CAS  PubMed  Google Scholar 

  59. Chang LY, et al. Molecular Basis and Role of Siglec-7 Ligand Expression on Chronic Lymphocytic Leukemia B Cells. Front Immunol. 2022;13:840388.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Sordo-Bahamonde C, et al. LAG-3 Blockade with Relatlimab (BMS-986016) Restores Anti-Leukemic Responses in Chronic Lymphocytic Leukemia. Cancers (Basel). 2021;13(9):2112. https://doi.org/10.3390/cancers13092112.

  61. Sordo-Bahamonde C, et al. BTLA/HVEM Axis Induces NK Cell Immunosuppression and Poor Outcome in Chronic Lymphocytic Leukemia. Cancers (Basel). 2021;13(8):1766. https://doi.org/10.3390/cancers13081766.

  62. Villa-Álvarez M, et al. Ig-Like Transcript 2 (ILT2) Blockade and Lenalidomide Restore NK Cell Function in Chronic Lymphocytic Leukemia. Front Immunol. 2018;9:2917.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Astaneh M, et al. Tim-3 and PD-1 blocking cannot restore the functional properties of natural killer cells in early clinical stages of chronic lymphocytic leukemia: An in vitro study. J Cancer Res Ther. 2022;18(3):704–11.

    Article  CAS  PubMed  Google Scholar 

  64. Alavianmehr A, et al. Association of killer-cell immunoglobulin-like receptor genes with acute myelogenous leukaemia. Int J Immunogenet. 2020;47(6):512–21.

    Article  CAS  PubMed  Google Scholar 

  65. Santiago V, et al. Human NK Cells Develop an Exhaustion Phenotype During Polar Degranulation at the Aspergillus fumigatus Hyphal Synapse. Front Immunol. 2018;9:2344.

    Article  PubMed  PubMed Central  Google Scholar 

  66. Montes P, et al. Tumor genetic alterations and features of the immune microenvironment drive myelodysplastic syndrome escape and progression. Cancer Immunol Immunother. 2019;68(12):2015–27.

    Article  CAS  PubMed  Google Scholar 

  67. Zeng X, et al. Terminal differentiation of bone marrow NK cells and increased circulation of TIGIT(+) NK cells may be related to poor outcome in acute myeloid leukemia. Asia Pac J Clin Oncol. 2022;18(4):456–64.

    Article  PubMed  Google Scholar 

  68. Tang L, et al. Characterization of Immune Dysfunction and Identification of Prognostic Immune-Related Risk Factors in Acute Myeloid Leukemia. Clin Cancer Res. 2020;26(7):1763–72.

    Article  CAS  PubMed  Google Scholar 

  69. Chajuwan T, et al. Characteristics and clinical correlation of TIM-3 and PD-1/PD-L1 expressions in leukemic cells and tumor microenvironment in newly diagnosed acute myeloid leukemia. Leuk Lymphoma. 2022;63(2):450–6.

    Article  CAS  PubMed  Google Scholar 

  70. Bou-Tayeh B, et al. Chronic IL-15 Stimulation and Impaired mTOR Signaling and Metabolism in Natural Killer Cells During Acute Myeloid Leukemia. Front Immunol. 2021;12:730970.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Li J, et al. PVRIG is a novel natural killer cell immune checkpoint receptor in acute myeloid leukemia. Haematologica. 2021;106(12):3115–24.

    Article  CAS  PubMed  Google Scholar 

  72. Hasim MS, et al. When killers become thieves: Trogocytosed PD-1 inhibits NK cells in cancer. Sci Adv. 2022;8(15):eabj3286.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Dong W, et al. The Mechanism of Anti-PD-L1 Antibody Efficacy against PD-L1-Negative Tumors Identifies NK Cells Expressing PD-L1 as a Cytolytic Effector. Cancer Discov. 2019;9(10):1422–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Shen W, et al. Immunovirotherapy with vesicular stomatitis virus and PD-L1 blockade enhances therapeutic outcome in murine acute myeloid leukemia. Blood. 2016;127(11):1449–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Tyagi A, et al. Evidence supporting a role for the immune checkpoint protein B7–H3 in NK cell-mediated cytotoxicity against AML. Blood. 2022;139(18):2782–96.

    Article  CAS  PubMed  Google Scholar 

  76. Kaito Y, et al. CD155 and CD112 as possible therapeutic targets of FLT3 inhibitors for acute myeloid leukemia. Oncol Lett. 2022;23(2):51.

    Article  CAS  PubMed  Google Scholar 

  77. Hattori N, et al. Monitoring TIGIT/DNAM-1 and PVR/PVRL2 Immune Checkpoint Expression Levels in Allogeneic Stem Cell Transplantation for Acute Myeloid Leukemia. Biol Blood Marrow Transplant. 2019;25(5):861–7.

    Article  CAS  PubMed  Google Scholar 

  78. Brauneck F, et al. Combined Blockade of TIGIT and CD39 or A2AR Enhances NK-92 Cell-Mediated Cytotoxicity in AML. Int J Mol Sci. 2021;22(23):12919. https://doi.org/10.3390/ijms222312919.

  79. Wang J, et al. Prognostic significance of TIM-3 expression pattern at diagnosis in patients with t(8;21) acute myeloid leukemia. Leuk Lymphoma. 2022;63(1):152–61.

    Article  PubMed  Google Scholar 

  80. Liu G, et al. Increased TIGIT expressing NK cells with dysfunctional phenotype in AML patients correlated with poor prognosis. Cancer Immunol Immunother. 2022;71(2):277–87.

    Article  CAS  PubMed  Google Scholar 

  81. Rakova J, et al. TIM-3 levels correlate with enhanced NK cell cytotoxicity and improved clinical outcome in AML patients. Oncoimmunology. 2021;10(1):1889822.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Ruggeri L, et al. Effects of anti-NKG2A antibody administration on leukemia and normal hematopoietic cells. Haematologica. 2016;101(5):626–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Nuebling T, et al. The Immune Checkpoint Modulator OX40 and Its Ligand OX40L in NK-Cell Immunosurveillance and Acute Myeloid Leukemia. Cancer Immunol Res. 2018;6(2):209–21.

    Article  CAS  PubMed  Google Scholar 

  84. Gurney M, et al. Concurrent transposon engineering and CRISPR/Cas9 genome editing of primary CLL-1 chimeric antigen receptor-natural killer cells. Cytotherapy. 2022;24(11):1087–94. https://doi.org/10.1016/j.jcyt.2022.07.008.

  85. Duault C, et al. Activated natural killer cells predict poor clinical prognosis in high-risk B- and T-cell acute lymphoblastic leukemia. Blood. 2021;138(16):1465–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Bailur JK, et al. Risk-associated alterations in marrow T cells in pediatric leukemia. JCI Insight. 2020;5(16):e140179. https://doi.org/10.1172/jci.insight.140179.

  87. Lee LJ, et al. Differential Regulation of NK Cell Receptors in Acute Lymphoblastic Leukemia. J Immunol Res. 2022;2022:7972039.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Ramírez-Ramírez D, et al. CRTAM(+) NK cells endowed with suppressor properties arise in leukemic bone marrow. J Leukoc Biol. 2019;105(5):999–1013.

    Article  PubMed  Google Scholar 

  89. Ishiyama KI, et al. Programmed cell death 1-expressing CD56-negative natural killer (NK) cell expansion is a hallmark of chronic NK cell activation during dasatinib treatment. Cancer Sci. 2021;112(2):523–36.

    Article  CAS  PubMed  Google Scholar 

  90. Li X, et al. miR-582 Suppresses the Proliferation of B-Cell Precursor Acute Lymphoblastic Leukemia (BCP-ALL) Cells and Protects Them From Natural Killer Cell-Mediated Cytotoxicity. Front Immunol. 2022;13:853094.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Rothfelder K, et al. Expression of the Immune Checkpoint Modulator OX40 in Acute Lymphoblastic Leukemia Is Associated with BCR-ABL Positivity. Neoplasia. 2018;20(11):1150–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Berger A, et al. Interleukin-15 in cancer immunotherapy: IL-15 receptor complex versus soluble IL-15 in a cancer cell-delivered murine leukemia model. J Immunother Cancer. 2019;7(1):355.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

None.

Funding

We would like to thank Tabriz University of Medical Sciences for financial support of this study (grant number: 68995).

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to the study’s conception and design. Data collection, manuscript editing, and reviewing were performed by FE, FA, AAS, and MHF. The first draft of the manuscript was written by AB, KA, and FJN. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Farhad Jadidi-Niaragh.

Ethics declarations

Ethics approval and consent to participate

This study is a review article and does not need ethics approval.

Informed consent was not required for this article.

Consent for publication

This article has no human data.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Barshidi, A., Ardeshiri, K., Ebrahimi, F. et al. The role of exhausted natural killer cells in the immunopathogenesis and treatment of leukemia. Cell Commun Signal 22, 59 (2024). https://doi.org/10.1186/s12964-023-01428-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12964-023-01428-2

Keywords