Next Article in Journal
Lipids of Platelet-Rich Fibrin Reduce the Inflammatory Response in Mesenchymal Cells and Macrophages
Next Article in Special Issue
An Emerging Role for Type I Interferons as Critical Regulators of Blood Coagulation
Previous Article in Journal
SERPIN-Derived Small Peptide (SP16) as a Potential Therapeutic Agent against HIV-Induced Inflammatory Molecules and Viral Replication in Cells of the Central Nervous System
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Killer Disarmed: Natural Killer Cell Impairment in Myelodysplastic Syndrome

by
Helena Arellano-Ballestero
1,
May Sabry
1,2,3 and
Mark W. Lowdell
1,2,*
1
Department of Haematology, University College London, London NW3 5PF, UK
2
InmuneBio Inc., Boca Raton, FL 33432, USA
3
Novamune Ltd., London WC2R 1DJ, UK
*
Author to whom correspondence should be addressed.
Cells 2023, 12(4), 633; https://doi.org/10.3390/cells12040633
Submission received: 27 January 2023 / Revised: 10 February 2023 / Accepted: 13 February 2023 / Published: 16 February 2023
(This article belongs to the Special Issue Innate Immunity in Health and Disease)

Abstract

:
Myelodysplastic syndrome (MDS) treatment remains a big challenge due to the heterogeneous nature of the disease and its ability to progress to acute myeloid leukemia (AML). The only curative option is allogeneic hematopoietic stem cell transplantation (HSCT), but most patients are unfit for this procedure and are left with only palliative treatment options, causing a big unmet need in the context of this disease. Natural killer (NK) cells are attractive candidates for MDS immunotherapy due to their ability to target myeloid leukemic cells without prior sensitization, and in recent years we have seen an arising number of clinical trials in AML and, recently, MDS. NK cells are reported to be highly dysfunctional in MDS patients, which can be overcome by adoptive NK cell immunotherapy or activation of endogenous NK cells. Here, we review the role of NK cells in MDS, the contribution of the tumor microenvironment (TME) to NK cell impairment, and the most recent data from NK cell-based clinical trials in MDS.

1. Introduction

Myelodysplastic syndrome (MDS) is a group of heterogeneous clonal hematopoietic stem cell disorders characterized by cytopenias leading to ineffective hematopoiesis and increased blast production, resulting in bone marrow failure as well as risk of progression to acute myeloid leukemia (AML) [1,2,3]. MDS occurs more frequently in elderly people, which could relate to the increased inflammatory state that is associated with aging, promoting the clonal hematopoiesis that we see in patients [4,5,6]. MDS patients can be classified according to their disease risk; low-risk patients are characterized by bone marrow apoptosis and autoimmune disease manifestations, whereas high-risk patients present immune disfunction, less bone marrow apoptosis, and secondary DNA damage, accelerating the progression to AML [7,8]. On average, 30% of MDS patients progress into AML [9,10,11]. The heterogeneous nature of the disease requires a complex and personalized variety of therapeutic approaches. Among the choices for MDS treatment the only therapy with curative potential is allogenic hematopoietic stem cell transplantation (HSCT), with the remainder of therapeutic approaches involving palliative care. Allogeneic HSCT is associated with a high risk of serious complications such as infections and graft-versus-host disease (GVHD); and since MDS is a disease of the elderly, this option is unavailable to many patients [12]. Currently, the Food and Drug Administration (FDA) has only approved three drugs for the treatment of MDS, the immunomodulatory agent lenalidomide and the hypomethylating agents azacitidine and decitibine. No new drugs have been approved since 2006, and no drug has been approved for second-line treatments such as immunotherapy, occasioning a large unmet need for patients who do not respond to licensed treatments [13].
Natural Killer (NK) cells are attractive candidates for immunotherapy due to their ability to kill cancer cells without prior sensitization. They begin their development in the bone marrow and go through a step-wise process from hematopoietic stem cells (HSCs) to common lymphoid progenitors (CLPs) and NK cell precursors (NKPs) and finally into CD56+ circulating NK cells. CD122 expression whilst in the bone marrow is critical for lineage commitment. NK cells that express CD56 can also be classified according to their maturation status, characterized by the expression levels of CD56. CD56bright NK cells represent a more immature subset that can differentiate into CD56dim NK cells with the acquisition of the Fcγ receptor, CD16 [14,15,16]. NK cells belong to group 1 innate lymphoid cells and make up between 5 to 15% of the total population of circulating lymphocytes [17]. Once activated and triggered, NK cells display direct cytotoxicity against a variety of tumor targets by degranulation; the release of lytic granules containing perforins and granzymes into the synapse between NK and target cell or by induction of apoptosis through the death receptors FasL/TRAIL. Another mechanism is antibody-dependent cell-mediated cytotoxicity (ADCC), where an NK cell will bind to antibodies attached to target cells via the CD16 receptor. Cross-linkage of CD16 leads to perforin and granzyme release causing elimination of the target cell. Triggered NK cells can also secrete cytokines such as tumor necrosis factor-alpha (TNF- α ) and interferon-gamma (IFN- γ ) leading to a greater influence over immune responses (Figure 1) [18]. In the context of MDS, several studies have reported evidence of NK cell impairment, highlighting the importance of understanding their role in the context of preleukemic myelodysplasia [19,20,21,22]. In this review article, we summarize the role of NK cells in MDS, discuss how the MDS microenvironment influences NK cell function, and highlight the most recent data from clinical trials using different NK cell-based immunotherapeutic approaches for the treatment of this disease.

2. NK Cells in MDS

NK cell impairment is frequently reported in cancer to include a reduction in cell numbers, dysregulation of antigen expression, and a display of weaker functional abilities such as cytokine secretion and cytotoxicity. Although earlier studies reported similar NK cell numbers in MDS patients compared to healthy donors [19], more recent reports show that MDS patients exhibit lower numbers of NK cells [22,23,24]. In most cases, this reduction in NK cell number does not correlate with a reduction in T cell lymphocytes. Instead, it is associated with higher risk subgroups of MDS patients according to the WHO and IPSS classification [22]. This is potentially informative, since it points to a particular association between NK cells and disease severity and not a general immunosuppression of broad lymphopenia. It suggests that NK cells are involved in the immune surveillance of dysplastic clones.
The dysregulation of NK cell antigen expression in cancer usually involves the downregulation of activating receptor expression and upregulation of inhibitory receptors, leading to weaker NK cell capacity for target cell recognition and killing. NK cell activation receptors include CD16, which mediates ADCC, and two families of natural cytotoxicity receptors (NCRs): the immunoglobulin superfamily of NKp30, NKp44, NKp46 and those that are C-type lectins, NKG2D and DNAM1. The C-type lectins are particularly important in leukemic cell targeting due to their recognition of MICA/B and ULBP1-6 and PVR (CD155) and Nectin-2 (CD112), respectively [25,26].
A hallmark of NK cells is the presence of killer cell immunoglobulin-like receptors (KIR) that bind to HLA-A/B/C molecules and mostly act as inhibitors of NK cell activation. NK cells also express the CD94/NKG2A complex that recognizes HLA-E and is the main inhibitor of NK activity. The KIR family of genes is highly polymorphic and four major inhibitory KIRs have been defined: KIR2DL2/3, KIR2DL1, KIR3DL1, and KIR3DL2. Immature NK cells will undergo a process called “education” or “licensing” by which they will acquire inhibitory receptors and become mature NK cells; NK cells lacking inhibitory receptors for HLA molecules will become hyporesponsive or anergic and be eliminated. The maturation of NK cells has been described as a gradual process characterized by the loss of NKG2A together with the acquisition of multiple KIR and CD57 [27,28].
Previous studies by Kiladjian et al. investigated NK cell receptor expression in MDS patients and reported comparable expression of NCRs in peripheral blood NK cells of MDS patients versus healthy controls [19]. In a more recent study by Carlsten et al., the downregulation of NKG2D and DNAM-1 expression in bone marrow-derived NK cells from MDS patients was reportedly associated with elevated blast counts and high-risk disease, but similar to Kiladjian et al. they did not see receptor changes in the surface of NK cells from peripheral blood [21]. In contrast, other studies investigating peripheral blood NK cells from MDS patients have reported loss of NKG2D, NKp30, NKp46, CD16, and CD161 expression [20,22,23,24]. Recent studies suggest that NK cell dysfunction in MDS may be attributed to the presence of NK cells with immature phenotypes characterized by an increase in the proportion of CD56bright NK cells, higher “early” KIR expression (KIR2DL2/3), and lower “late” KIR expression (KIR2DL1 and KIR3DL) in MDS patients [22,24,29]. The expression of NK cell ligands in bone marrow aspirates of MDS patients has also been studied since these receptor–ligand interactions are crucial for lysis of MDS blasts, but reports have yielded conflicting results. Epling-Burnette et al. reported that 30% of MDS blasts expressed the NKG2D ligands MICA/B [20]. In another study by Carlsten et al., NKG2D ligands MICs and ULBPs were found to be rarely expressed in the bone marrow of MDS patients. Instead, patients expressed the DNAM1 ligands, CD155 and CD112, showing an important role for DNAM1-CD155/CD112 interactions in the killing of MDS blasts [21].
These conflicting data on NK cell receptor expression in MDS may be attributed to the source of NK cells, whether obtained from peripheral blood or bone marrow, and timing of samples especially as it relates to the administration of chemotherapeutic agents, which are known to have an effect on the state of NK cells [30,31,32]. Taken together, these studies demonstrate an NK cell phenotype characterized by lack of maturation markers and the downregulation of NK cell activation receptors in MDS patients, which is associated with NK cell functional impairment.
Several studies have reported impaired or dysfunctional NK cell cytokine secretion or cytotoxicity in MDS patients. A reduction in MDS-NK cell secretion of TNF- α and IFN- γ was previously demonstrated in response to IL-2 and K562 stimulation [19,22]. NK cell cytotoxicity through perforin and granzyme release is associated with enhanced degranulation as measured by LAMP-1/CD107a expression or by the activation of death receptor-mediated pathways TRAIL and FasL [33,34]. Carlsten et al. reported lower degranulation in MDS-NK cells following exposure to K562 leukemic target cells relative to healthy controls [21]. Hejazi et al. showed no difference in the mobilization of CD107a, a molecule expressed on the NK cell surface after degranulation, when MDS-NK cells were exposed to K562. However, they showed a significant decrease in MDS-NK cell killing activity without any associated changes in CD107a expression compared to healthy donors. This observation prompted them to investigate whether degranulating MDS-NK cells are properly armed with cytotoxic molecules. They observed a substantial reduction in perforin and granzyme B loading of granules in MDS-NK cells, which explains why NK cells might still be able to mobilize CD107a to the cell surface without effective killing of target cells [22]. Similarly, other studies have shown impaired lysis of K562 and CD34+ blasts by NK cells from MDS patients [19,20,21,22].
In a recent study, Tsirogianni et al. [35] analyzed peripheral blood NK cells from MDS patients who had been treated with azacytidine, and they tested their ability to lyse K562 leukemic target cells in vitro. Patients with higher lytic function showed significantly longer overall survival. Indeed, the association was so strong that the group was able to calculate a threshold of NK mediated lysis, which was predictive for survival beyond 2 years. Patients below the threshold showed a median overall survival of 18 months compared to those falling above the threshold with a median survival of 52 months. Collectively, these studies suggest that NK cell antitumor functions may be critical in the response to MDS. Their impairment, however, can be influenced by numerous factors in the tumor microenvironment that contribute to disease progression.

3. The Tumor Microenvironment in MDS

There is growing evidence that different components of the tumor microenvironment (TME), including other immune cells and soluble factors, contribute to NK cell impairment in MDS (Figure 2) although whether the TME is responsible for the dysfunction of NK cells isolated from the peripheral blood of MDS and other cancer patients is unknown. One possibility is that the circulating NK cells isolated from blood have trafficked through the TME and suffered long term impairment. However, Imai et al. [36] showed elegantly that healthy individuals show a range of NK cell functional abilities, and those with lower NK function have a higher lifetime risk of cancer. This supports the notion that NK cells mediate tumor immune surveillance and the first stages of impaired function occur prior to the development of cancer and a TME. So, passage through a TME is not required for impairment of NK activity, but the composition of the TME can certainly provide additional suppression of NK cell function.
Dendritic cells (DCs) in MDS patients have been reported to have an immature phenotype that is observed on a transcriptional and functional level [37,38]; this will have negative effects on DC-NK cell crosstalk and the release of activating soluble factors such as IL-12, IL-18, and type-I IFN, which prime NK cells for tumor lysis [39]. Increased levels of vascular endothelial growth factor (VEGF) have been found in the serum of MDS patients, which will have a series of effects in the TME [40,41]; increased VEGF will contribute to the inhibition of DC maturation [42,43] and induction of T regulatory cell (Treg) activation, [42,43], which is associated with an unfavorable disease prognosis in MDS [8,44,45,46,47,48]. Tregs can play two opposing roles in MDS; in the low-risk group Tregs appear to be dysfunctional, favoring the selection of dysplastic clones; in the high-risk group, Treg numbers are expanded and can suppress immune responses against transformed clones, promoting leukemia progression [8,44,45,46,47,48]. Tregs are well known to suppress NK cell function via cell-to-cell contact or through the secretion of soluble factors such as IL-10 or TGF- β [49,50]. Furthermore, in vivo murine models of AML have shown that Treg depletion allows better NK cell activation [51].
Another source of VEGF in MDS is from myeloid-derived suppressor cells (MDSCs) [52], which are expanded in the bone marrow of MDS patients and correlate with high-risk disease [24,52,53,54]. Two mechanisms have been described on how MDSCs are recruited into the bone marrow of MDS patients. The inflammatory environment in the MDS bone marrow contains inflammation associated signaling molecules, such as the danger-associated molecular pattern (DAMP) heterodimer S100A8/S100A9 that will interact with its ligand CD33 on MDSCs. S100A8/S100A9 has been reported to be overexpressed in MDS, allowing its accumulation and activation [52]. Another mechanism that has been described is via the CXCR4/CXCL12 axis, where the stromal cells in the bone marrow niche supporting MDS blasts have been shown to overexpress CXCL12. The MDSCs in these patients show an upregulation of CXCR4, a ligand for CXCL12 [54,55]. CXCL12 is an important chemokine for MDSCs, and its hyperexpression is thought to mediate the increased frequency of MDSCs in the bone marrow of MDS patients.
Furthermore, MDSCs in MDS have been described as hyperfunctional and have increased suppressive activity by secreting IL-10, TGF- β , and reactive oxygen species (ROS) that will ultimately impair NK cells [52,56,57]. In chronic myeloid leukemia, AML, and chronic myelomonocytic leukemia it has been shown that ROS triggers apoptosis of NK cells in the TME and also induce downregulation of activating receptors, leading to dysfunctional NK cells [58,59,60]. Another source of ROS is blood transfusions that MDS patients receive as part of their treatment. These often induce iron overload, causing ROS generation and therefore NK cell impairment [61,62,63].
Mesenchymal stromal cells (MSCs) are another cell type that is reportedly impaired in MDS patients. These cells exhibit reduced proliferative capacity and poor ability to differentiate into other cell types; altered morphology; and alteration of molecules and cytokines involved in hematopoietic stem and progenitor cells (HSPCs)–MSCs interactions [64,65,66]. MSCs have a very important role in supporting HSPCs in their renewal and differentiation, which will allow the production of mature blood cells, including NK cells [67,68].
Hejazi and colleagues argue in their paper that the absence of stromal support in the microenvironment in MDS patients could be leading to the lack of maturity in patient NK cells, as it is known that full maturation of NK cells that express KIR ligands requires the presence of stromal cells [22,69]. The increased secretion of the pro-inflammatory cytokine IL-6 by MSCs has also been reported in MDS [54,70], contributing to general inflammation in the bone marrow of patients. This inflammatory state in the bone marrow of MDS patients has been described by increased levels of pro-inflammatory cytokines such as IFN- γ and TNF- α , which will induce apoptosis on CD34 cells and bone marrow failure, helping in the selection of malignant CD34 cells [71,72]. Importantly, TNF- α activates the transcription factor NF- κ β , a regulator of cell signaling, proliferation, and differentiation [73]. NF- κ β levels are increased in high-risk MDS patients, corresponding with an increase of blast counts [74]. Furthermore, some studies have shown that NF- κ β signaling contributes to leukemia progression [75], which could play an important role also in transition of MDS to AML.
Hypoxia is another element that has been described to have an effect in the bone marrow of MDS patients. Hypoxic conditions have been shown to select CD34 malignant cells with stem cell potential in MDS patients [76]. Expanded CD34 malignant blasts will further contribute to inflammation in the bone marrow by secreting pro-inflammatory cytokines such as IL-8 and TGF- β into the TME [77,78,79,80,81,82], increasing levels of ROS [83,84,85], and MSCs inhibition via TGF- β secretion [82,86], ultimately affecting NK cells.
As we have seen, a broad range of different elements in the TME of MDS patients appear to be active, all of which may compromise NK cell interaction with other cell types or affect NK cells indirectly through secretion of inhibitory cytokines. Understanding the role of the TME and how it contributes to NK cell impairment is crucial in order to develop successful anticancer therapies that could include combination strategies targeting different elements of the TME, thus enhancing NK cell activity for successful blast killing.

4. NK Cell Immunotherapy in MDS

Different immunotherapeutic strategies for NK cell modulation have been developed for MDS treatment. Preclinical studies have reported enhanced MDS patient-derived NK cell proliferation, granule secretion, and cytotoxicity after exposure to IL-2, showing that NK cell dysfunction can be overcome [20,22]. The only curative option nowadays for patients with MDS is allogeneic HSCT from an HLA-identical donor. However, one of the main problems is the recurrence of underlying leukemia after the transplant, which is usually the cause of treatment failure [87]. Therefore, patients who are refractory or have relapsed are left with few options. In these cases, NK cell immunotherapy, which has shown positive results both in solid and in liquid cancers (reviewed here [88]), could be a suitable option. In the context of MDS, NK cells have been tested clinically in the past, and there are several ongoing clinical trials at the time of writing (Table 1). Two different options have been considered; the first one involves adoptive immunotherapy using allogeneic donor NK from a variety of sources: adult peripheral blood, umbilical cord blood, and hematopoietic progenitor cells from iPSC cells. The second approach has been to potentiate the patients’ own NK cells in vivo.
The infusion of NK cells from an HLA matched or unmatched donor is the most common approach, but, in most cases, this requires the infusion of cytokines such as IL-2 or IL-15 to expand them and sustain them in vivo. Some trials using IL-2 activated adoptive NK cell therapy have shown good results in patients with relapsed or refractory AML, with complete remissions in up to 50% of the patients [89]. Even though it is a widely used strategy, cytokine infusions have previously been shown to induce severe toxic side effects such as cytokine release syndrome, and the development of novel strategies that sustain NK cell proliferation and activation in vivo without toxicity will be important moving forward. Low-dose IL-2 is also known to induce Treg activation [90]. Tregs have a higher affinity for IL-2, and thus there is a great risk of starving NK of IL-2 whist simultaneously activating Treg to further suppress NK cell function [91]. The administration of donor NK cells together with IL-2 and with the TGF- β receptor I inhibitor, vactosertib, is being trialed in relapsed patients with solid and liquid cancers, including MDS (NCT05400122). It has been shown that TGF- β inhibitors such as vactosertib can help attenuate Tregs [92,93]; hence, this combination of therapies holds some promise in reducing unwanted side effects of cytokine administration.
IL-15 is another cytokine that has been used in the clinical setting, but as opposed to IL-2, IL-15 holds some advantages such as its superior role in the stimulation of NK cells and T cells and also the fact that it does not stimulate Treg proliferation [94,95]. Cooley et al. performed the first clinical trial in AML/MDS with infusion of haploidentical NK cells with IL-15 doses afterwards. Patients received either intravenous (NCT01385423) or subcutaneous (NCT02395822) IL-15 doses after NK cell infusion. In phase I, 36% of patients had robust in vivo NK cell expansion after 14 days, and 32% achieved complete remission. In phase II, 27% of patients had NK cell expansion after 14 days, and 40% achieved complete remission. IL-15 infusion induced better in vivo expansion than previous trials with IL-2, but it was associated with cytokine release syndrome in 56% of patients when given subcutaneously but not intravenously [96]. At the time of writing this manuscript, there is an ongoing trial (NCT02890758) for different types of cancers, including MDS, where patients receive NK cells from HLA-mismatched donors either alone or in combination with IL-15 infusions.
A recent development in NK cell biology is the concept of NK cells activated in vivo or ex vivo such that they obtain a memory-like phenotype (ml-NK). These ml-NKs can be generated by short-term exposure to a combination of IL-12, IL-15, and IL-18, in which case they are commonly termed “cytokine-induced ml-NK”—CIML-NK [97,98,99]. CIML-NK show better capacity to respond after a second activation with cytokines or by engagement of activating NK receptors. The first report of clinical use of CIML-NK was in patients with MDS and AML [97]. Haploidentical NK cells from donors were pre-activated for 12-16 h with IL-12, IL-15, and IL-18 ex vivo and then infused into patients who had been preconditioned with lymphodepleting chemotherapy. The infusions were supplemented with low dose IL-2. These CIML-NK were not inhibited by KIR/KIR-ligand interactions and were highly lytic to myeloid leukemic cells in vitro. Moreover, the infused NK cells expanded in vivo were associated with clinical responses in over half of the patients treated, including complete remissions in four or five patients who responded. This phase I trial transitioned to a phase II trial in which the low dose, subcutaneous IL-2 infusions were replaced with IL-15 (NCT01898793), but the trial was terminated due to insufficient funding/staff. Another trial in the use of CIML in hematologic malignancies including juvenile myelomonocytic leukemia is also ongoing (NCT04024761). In this trial, CIML NK cells will be generated and infused to the patients, but no cytokine infusions will be given afterwards. As seen here, the use of cytokines to expand and activate NK cells from patients in vivo or ex vivo has been widely used in the clinical setting showing some promising results, but cytokine release syndrome is a common and severe side effect that should not go unnoticed. Ex vivo incubation of NK cells with cytokines has achieved greater expansion numbers and activation of cells; however, cytokine infusions in the patients are still needed afterwards in order to sustain NK cells. Strategies to activate and expand NK cells that do not include cytokines should be further explored, and we may see an increase of other clinical trials in the upcoming years.
The use of feeder-cell ex vivo is another alternative for the expansion and activation of NK cells. In vitro data from the use of the irradiated K562 cell line modified to express membrane bound IL-15 and 41BB ligand (K562-mbIL15-41BBL) has shown a 26-fold expansion of NK cells and powerful antileukemic activity, with the ability to eradicate AML in mouse models [100,101,102]. Better in vitro expansion numbers have been achieved with the use of K562-mbIL21 where NK cells were expanded by up to 35,000-fold in vitro [103,104,105]. At this moment, there is an ongoing clinical trial using expanded haploidentical NK cells with K562-mbIL21 (NCT04220684).
Chimeric antigen receptor (CAR)-T cells have shown remarkable results in some blood cancers such as acute lymphoblastic leukemia, chronic lymphocytic leukemia, and lymphoma [106,107,108] and have also been used in clinical trials for AML/MDS [109,110,111,112,113]. However, despite their success in the clinics, autologous CAR-T cells present some limitations related to the fact that these drugs are personalized to only one patient, making them complex and expensive. Furthermore, in some patients CAR-T cell treatment has been linked to cytokine release syndrome and neurotoxicity [114]. Allogeneic CAR-NK cells are considered potential alternatives to autologous CAR-T since allogeneic NK cells do not induce graft-versus-host disease and may be available as an off-the-shelf alternative. NK cells can be sourced from an allogeneic donor without the need of full HLA matching; hence, they do not need to be produced individually for each patient. They also appear to be safe when infused to patients with cancer [115,116,117]. Nonetheless, one of the main challenges to develop CAR therapies for MDS/AML remains the lack of specific ligands, which are usually expressed in normal myeloid cells [118,119]. Tang et al. performed the first clinical trial using CAR-NK cells in AML, targeting CD33 and using the NK92 cell line as the source of NK cells. They reported no significant adverse effects and concluded that, even though they did not see any clinical effect, CAR-NK cells were safe to administer [120]. Several other molecules are being used in CAR-NK therapy in clinical trials in leukemia, but no results have been posted so far: CD7 (NCT02742727), CD19 (NCT02892695, NCT03056339, NCT04796675, NCT00995137) and NKG2DL (NCT04623944). There are other preclinical studies looking at the use of CAR-NK cells in AML. Sinha et al. studied a CAR construct with CD123 in NK92 cells and evaluated their efficacy; they observed how these cells exhibited higher cytotoxicity against AML cell lines expressing CD123 both in vitro and in vivo models [121]. Dong et al. engineered memory-like NK cells to express a CAR against NPM1c, an antigen expressed 35% in AML that showed enhanced cytotoxicity in in vivo models [122]. There are no current data on the use of CAR-NK cells for MDS patients, but some trials are ongoing using allogeneic CAR-NK cells targeting NKG2D ligands (NCT04623944) and CAR.70/IL15-transduced NK cells derived from cord blood (NCT05092451). The use of CAR-NK cells offers an exciting opportunity to develop an off-the-shelf product, but identifying markers that can target MDS blasts exclusively are crucial and still remain one of the biggest challenges.
The in vivo potentiation of the patient’s own NK cells is another strategy that has been considered. Bispecific and trispecific killer cell engagers (BiKE and TriKE) are single-chain variable fragment recombinant reagents that contain an anti-CD16 expressed on effector NK cells and other antigens of interest in cancer cells. Gleason et al. tested the ability of a BiKE for CD16 and CD33 to induce NK cell function in MDS patients in vitro, where CD33 engages myeloid targets including MDS. Their study showed enhanced degranulation and TNF- α and INF- γ secretion in NK cells from patients [23]. Other TriKEs have been studied in the context of MDS, where besides the anti-CD16 and the anti-CD33, a modified IL-15 linker was added in order to induce NK cell proliferation. These studies reported enhanced killing kinetics and evasion of MDSCs suppression [123,124]. At the time of writing this manuscript, there was an ongoing trial (NCT03214666) testing the TriKE GTB-3550 in MDS and other hematological malignancies, but it was terminated due to the development of the second generation TriKE GTB-3650. Overall, BiKEs and TriKEs have shown promising results both in vitro and in vivo and provide a versatile off-the-shelf option; however, safety concerns remain since IL-15 stimulation of inflammatory T cells could trigger cytokine release syndrome.
The activation of NK cells has also been studied in the context of tumor-priming. NK cells that are exposed to the ALL cell line INB16 enter a primed stage where they are ready to lyse upon exposure to a second tumor cell signal, even if the second signals arise from NK-resistant tumor cells [125,126,127,128]. Tumor-primed NK cells have also been used in clinical trials for AML/MDS, with little toxicity and some sustained clinical responses. This is of particular note since these were the first clinical trials of an adoptive NK therapy in the absence of cytokine support. Allogeneic NK cells from related, haploidentical donors were incubated overnight with a lysate of INB16 that was then removed, and the tumor-primed NK cells were cryopreserved, shipped, and infused into the patients one day after completion of lymphoreductive chemotherapy [129]. Four of the seven patients who completed treatment showed clinical responses with one patient with chemo-resistant AML achieving CR, which was sustained for over 11 months. In a second trial, 12 AML patients at high risk of recurrence were treated following the same protocol, where two patients remained relapse-free in posttrial follow-up, and three patients remained in complete remission for 32.6 to 47.6+ months [130].
The observation that MDS patients with moderate NK cell lytic function have a significantly better prognosis than those with low or undetectable NK medicated killing of K562 cells [32] led to the hypothesis that activation of endogenous NK cells in those MDS patients with low NK function might move them above the threshold for improved overall survival. Tumor-priming of endogenous NK cells in vivo is being trialed at the moment in high-risk MDS and AML patients (EudraCT 2019-004820-40) who receive three weekly infusions of a replication-incompetent preparation of the INB16 cell line. These patients receive no lymphoreductive conditioning chemotherapy and no cytokine support, resulting in a treatment that is well tolerated and compatible with an elderly patient cohort.
Table 1. Summary of NK cell-based clinical trials for MDS.
Table 1. Summary of NK cell-based clinical trials for MDS.
Approach/TreatmentTrial PhaseStatusPatient NumbersTrial IdentifierReferences
Adoptive NK cell immunotherapy
+cytokine stimulation with IL-2/-15/CIMLPhase ICompleted26NCT01385423[96]
Ongoing76NCT05400122, NCT02890758, NCT04024761[92,93]
Phase IICompleted17NCT02395822[96]
Phase I/IITerminated89NCT01898793[97,98,99]
+expanded K562-mbIL21Phase IOngoing30NCT04220684[100,101,102,103,104,105]
+CAR NK cellsPhase IOngoing90NCT04623944[120,121,122]
Phase I/IIOngoing94NCT05092451
In vivo NK cell activation
BiKE/TriKEPhase I/IITerminated12NCT03214666[23,123,124]
INB16 primingPhase IOngoing12EudraCT 2019-004820-40[125,126,127,128,129,130]
Abbreviations: IL, interleukin; CIML, cytokine-induced memory-like; mbIL21. membrane bound IL-21; CAR, chimeric antigen receptor; BiKE/TriKE, bi/trispecific killer cell engagers.
To summarize, NK cell immunotherapy in AML/MDS offers a wide range of options that show exciting opportunities and appear to be safe for the treatment of this disease, but relapse rates are still high, and side effects of concurrent cytokine treatment are common. Cytokine administration is usually a requirement in order to sustain adoptively transferred NK cells, especially those generated by extensive ex vivo expansion. The development of better strategies for NK cell expansion and persistence post infusion remains a central tenet of research, including transduction of mbIL-15 constructs into the NK product. The use of feeder-cells to increase expansion of mature NK, or iPSC as a reliable source of proliferative NK cells, promise to reduce the cost of manufacture of allogeneic NK cells for adoptive immunotherapy. This is allowing the use of multiple NK infusions and is a move away from the concept of “NK engraftment and cure” towards one of “NK injection and control” where NK cell administrations become an ongoing treatment. The success of this strategy is predicated on the assumption that repeat infusions of allogeneic NK cells will not require lymphodepleting chemotherapy before each injection, and this is being tested in trials currently. The need for adoptive NK immunotherapy may be avoided by enhancing endogenous NK function, and BiKEs/TriKEs and in vivo generation of ml-NK cells are exciting off-the-shelf products that have shown interesting results that could benefit many patients as they are relatively inexpensive, are not personalized, and appear to be well tolerated, which might result in a surge of NK cell therapies in the upcoming years.

5. Conclusions

The heterogeneous nature of MDS hinders treatment of this disease, where delaying progression to secondary lethal AML is one of the key challenges in managing patients. NK cells play a crucial role in immune surveillance, and their impairment in the context of MDS suggests that is important to understand their role in this disease. The presence of a highly suppressive TME further impairs NK cell function, making it equally important to understand. Nonetheless, exciting in vitro and in vivo data suggest that NK cell impairment can be overcome in MDS, and results from early clinical trials are encouraging. One of the main challenges remains the presence of low numbers of NK cells seen in some MDS patients and the consequent need for adoptive immunotherapy. Challenges remain to sustain adoptive NK cells in vivo, although the use of cytokines remains a common yet problematic strategy, or to develop allogeneic NK cells that do not provoke allo-immunity, and thus repeat NK doses can be given. The imatinib family of drugs has revolutionized the treatment of chronic myeloid leukemia so that patients are no longer exposed to the high risk of allogeneic hematopoietic stem cell transplants but are maintained disease-free with repeat treatments. The very minimal toxicity of allogeneic NK cell infusions suggests that these too might become routine treatments to control disease. The field of NK cell immunotherapy is still expanding and holds a lot of potential to be unraveled over the coming years, leading to yet more novel therapies and the chance for significant patient benefit.

Author Contributions

H.A.-B. and M.S.—writing and original draft preparation; M.W.L.—writing, review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

M.S. was funded by Novamune Ltd. during the drafting of this review.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

M.W.L. is a cofounder and shareholder of INmuneBio Inc., a publicly listed biotech company developing INB16 and INKmuneTM in clinical trials. M.S. was funded by Novamune, a private company that holds the rights to develop an adoptive NK immunotherapy.

References

  1. Corey, S.J.; Minden, M.D.; Barber, D.L.; Kantarjian, H.; Wang, J.C.Y.; Schimmer, A.D. Myelodysplastic Syndromes: The Complexity of Stem-Cell Diseases. Nat. Rev. Cancer 2007, 7, 118–129. [Google Scholar] [CrossRef] [PubMed]
  2. Bejar, R.; Stevenson, K.; Abdel-Wahab, O.; Galili, N.; Nilsson, B.; Garcia-Manero, G.; Kantarjian, H.; Raza, A.; Levine, R.L.; Neuberg, D.; et al. Clinical Effect of Point Mutations in Myelodysplastic Syndromes. N. Engl. J. Med. 2011, 364, 2496–2506. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Papaemmanuil, E.; Gerstung, M.; Malcovati, L.; Tauro, S.; Gundem, G.; Van Loo, P.; Yoon, C.J.; Ellis, P.; Wedge, D.C.; Pellagatti, A.; et al. Clinical and Biological Implications of Driver Mutations in Myelodysplastic Syndromes. Blood 2013, 122, 3616–3627. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Takizawa, H.; Manz, M.G. Impact of Inflammation on Early Hematopoiesis and the Microenvironment. Int. J. Hematol. 2017, 106, 27–33. [Google Scholar] [CrossRef] [Green Version]
  5. Franceschi, C.; Campisi, J. Chronic Inflammation (Inflammaging) and Its Potential Contribution to Age-Associated Diseases. J. Gerontol. A Biol. Sci. Med. Sci. 2014, 69 (Suppl. S1), S4–S9. [Google Scholar] [CrossRef]
  6. Koschmieder, S.; Mughal, T.I.; Hasselbalch, H.C.; Barosi, G.; Valent, P.; Kiladjian, J.-J.; Jeryczynski, G.; Gisslinger, H.; Jutzi, J.S.; Pahl, H.L.; et al. Myeloproliferative Neoplasms and Inflammation: Whether to Target the Malignant Clone or the Inflammatory Process or Both. Leukemia 2016, 30, 1018–1024. [Google Scholar] [CrossRef]
  7. Aggarwal, S.; van de Loosdrecht, A.A.; Alhan, C.; Ossenkoppele, G.J.; Westers, T.M.; Bontkes, H.J. Role of Immune Responses in the Pathogenesis of Low-Risk MDS and High-Risk MDS: Implications for Immunotherapy. Br. J. Haematol. 2011, 153, 568–581. [Google Scholar] [CrossRef]
  8. Kotsianidis, I.; Bouchliou, I.; Nakou, E.; Spanoudakis, E.; Margaritis, D.; Christophoridou, A.V.; Anastasiades, A.; Tsigalou, C.; Bourikas, G.; Karadimitris, A.; et al. Kinetics, Function and Bone Marrow Trafficking of CD4+ CD25+FOXP3+ Regulatory T Cells in Myelodysplastic Syndromes (MDS). Leukemia 2009, 23, 510–518. [Google Scholar] [CrossRef] [Green Version]
  9. Germing, U.; Strupp, C.; Kuendgen, A.; Isa, S.; Knipp, S.; Hildebrandt, B.; Giagounidis, A.; Aul, C.; Gattermann, N.; Haas, R. Prospective Validation of the WHO Proposals for the Classification of Myelodysplastic Syndromes. Haematologica 2006, 91, 1596–1604. [Google Scholar] [CrossRef]
  10. Breccia, M.; Latagliata, R.; Cannella, L.; Carmosino, I.; Santopietro, M.; Loglisci, G.; Federico, V.; Alimena, G. Refractory Cytopenia with Unilineage Dysplasia: Analysis of Prognostic Factors and Survival in 126 Patients. Leuk Lymphoma 2010, 51, 783–788. [Google Scholar] [CrossRef]
  11. Greenberg, P.; Cox, C.; LeBeau, M.M.; Fenaux, P.; Morel, P.; Sanz, G.; Sanz, M.; Vallespi, T.; Hamblin, T.; Oscier, D.; et al. International Scoring System for Evaluating Prognosis in Myelodysplastic Syndromes. Blood 1997, 89, 2079–2088. [Google Scholar] [CrossRef] [PubMed]
  12. Nachtkamp, K.; Kündgen, A.; Strupp, C.; Giagounidis, A.; Kobbe, G.; Gattermann, N.; Haas, R.; Germing, U. Impact on Survival of Different Treatments for Myelodysplastic Syndromes (MDS). Leuk Res. 2009, 33, 1024–1028. [Google Scholar] [CrossRef] [PubMed]
  13. Chandhok, N.S.; Boddu, P.C.; Gore, S.D.; Prebet, T. What Are the Most Promising New Agents in Myelodysplastic Syndromes? Curr. Opin. Hematol. 2019, 26, 77–87. [Google Scholar] [CrossRef] [PubMed]
  14. Briercheck, E.L.; Freud, A.G.; Caligiuri, M.A. Human Natural Killer Cell Development. Nat. Kill. Cells Basic Sci. Clin. Appl. 2009, 214, 113–122. [Google Scholar] [CrossRef]
  15. Crinier, A.; Milpied, P.; Escalière, B.; Piperoglou, C.; Galluso, J.; Balsamo, A.; Spinelli, L.; Cervera-Marzal, I.; Ebbo, M.; Girard-Madoux, M. High-Dimensional Single-Cell Analysis Identifies Organ-Specific Signatures and Conserved NK Cell Subsets in Humans and Mice. Immunity 2018, 49, 971–986. [Google Scholar] [CrossRef] [Green Version]
  16. di Vito, C.; Mikulak, J.; Mavilio, D. On the Way to Become a Natural Killer Cell. Front. Immunol. 2019, 10, 1–15. [Google Scholar] [CrossRef]
  17. Spits, H.; Artis, D.; Colonna, M.; Diefenbach, A.; di Santo, J.P.; Eberl, G.; Koyasu, S.; Locksley, R.M.; McKenzie, A.N.J.; Mebius, R.E.; et al. Innate Lymphoid Cells—A Proposal for Uniform Nomenclature. Nat. Rev. Immunol. 2013, 13, 145–149. [Google Scholar] [CrossRef]
  18. 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] [Green Version]
  19. Kiladjian, J.-J.; Bourgeois, E.; Lobe, I.; Braun, T.; Visentin, G.; Bourhis, J.-H.; Fenaux, P.; Chouaib, S.; Caignard, A. Cytolytic Function and Survival of Natural Killer Cells Are Severely Altered in Myelodysplastic Syndromes. Leukemia 2006, 20, 463–470. [Google Scholar] [CrossRef] [Green Version]
  20. Epling-Burnette, P.K.; Zhong, B.; Salih, H.R.; Wei, S.; Bai, F.; Boulware, D.; List, A.F.; Painter, J.S.; Moscinski, L.; Rollison, D.E.; et al. Reduced Natural Killer (NK) Function Associated with High-Risk Myelodysplastic Syndrome (MDS) and Reduced Expression of Activating NK Receptors. Blood 2007, 109, 4816–4824. [Google Scholar] [CrossRef]
  21. 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] [PubMed]
  22. Hejazi, M.; Manser, A.R.; Fröbel, J.; Kündgen, A.; Zhao, X.; Schönberg, K.; Germing, U.; Haas, R.; Gattermann, N.; Uhrberg, M. Impaired Cytotoxicity Associated with Defective Natural Killer Cell Differentiation in Myelodysplastic Syndromes. Haematologica 2015, 100, 643–652. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Gleason, M.K.; Ross, J.A.; Warlick, E.D.; Lund, T.C.; Verneris, M.R.; Wiernik, A.; Spellman, S.; Haagenson, M.D.; Lenvik, A.J.; Litzow, M.R.; et al. CD16xCD33 Bispecific Killer Cell Engager (BiKE) Activates NK Cells against Primary MDS and MDSC CD33+ Targets. Blood 2014, 123, 3016–3026. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Montes, P.; Bernal, M.; Campo, L.N.; González-Ramírez, A.R.; Jiménez, P.; Garrido, P.; Jurado, M.; Garrido, F.; Ruiz-Cabello, F.; Hernández, F. Tumor Genetic Alterations and Features of the Immune Microenvironment Drive Myelodysplastic Syndrome Escape and Progression. Cancer Immunol. Immunother. 2019, 68, 2015–2027. [Google Scholar] [CrossRef]
  25. Parham, P. MHC Class I Molecules and Kirs in Human History, Health and Survival. Nat. Rev. Immunol. 2005, 5, 201–214. [Google Scholar] [CrossRef]
  26. 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]
  27. Bjorkstrom, N.K.; Riese, P.; Heuts, F.; Andersson, S.; Fauriat, C.; Ivarsson, M.A.; Bjo, A.T.; Flodstro, M.; Michae, J.; Rottenberg, M.E.; et al. Expression Patterns of NKG2A, KIR, and CD57 Define a Process of CD56 Dim NK-Cell Differentiation Uncoupled from NK-Cell Education. Blood 2010, 116, 3853–3864. [Google Scholar] [CrossRef] [Green Version]
  28. Andersson, S.; Fauriat, C.; Malmberg, J.A.; Ljunggren, H.G.; Malmberg, K.J. KIR Acquisition Probabilities Are Independent of Self-HLA Class I Ligands and Increase with Cellular KIR Expression. Blood 2009, 114, 95–104. [Google Scholar] [CrossRef]
  29. Schönberg, K.; Sribar, M.; Enczmann, J.; Fischer, J.C.; Uhrberg, M. Analyses of HLA-C-Specific KIR Repertoires in Donors with Group a and B Haplotypes Suggest a Ligand-Instructed Model of NK Cell Receptor Acquisition. Blood 2011, 117, 98–107. [Google Scholar] [CrossRef] [Green Version]
  30. Markasz, L.; Stuber, G.; Vanherberghen, B.; Flaberg, E.; Olah, E.; Carbone, E.; Eksborg, S.; Klein, E.; Skribek, H.; Szekely, L. Effect of Frequently Used Chemotherapeutic Drugs on the Cytotoxic Activity of Human Natural Killer Cells. Mol. Cancer 2007, 6, 644–654. [Google Scholar] [CrossRef] [Green Version]
  31. Dauguet, N.; Récher, C.; Demur, C.; Fournié, J.-J.; Poupot, M.; Poupot, R. Pre-Eminence and Persistence of Immature Natural Killer Cells in Acute Myeloid Leukemia Patients in First Complete Remission. Am. J. Hematol. 2011, 86, 209–213. [Google Scholar] [CrossRef] [PubMed]
  32. Najima, Y.; Yoshida, C.; Iriyama, N.; Fujisawa, S.; Wakita, H.; Chiba, S.; Okamoto, S.; Kawakami, K.; Takezako, N.; Kumagai, T.; et al. Regulatory T Cell Inhibition by Dasatinib Is Associated with Natural Killer Cell Differentiation and a Favorable Molecular Response—The Final Results of the D-First Study. Leuk. Res. 2018, 66, 66–72. [Google Scholar] [CrossRef] [PubMed]
  33. 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] [PubMed]
  34. Screpanti, V.; Wallin, R.P.A.; Grandien, A.; Ljunggren, H.-G. Impact of FASL-Induced Apoptosis in the Elimination of Tumor Cells by NK Cells. Mol. Immunol. 2005, 42, 495–499. [Google Scholar] [CrossRef]
  35. Tsirogianni, M.; Grigoriou, E.; Kapsimalli, V.; Dagla, K.; Stamouli, M.; Gkirkas, K.; Konsta, E.; Karagiannidou, A.; Gkodopoulos, K.; Stavroulaki, G.; et al. Natural Killer Cell Cytotoxicity Is a Predictor of Outcome for Patients with High Risk Myelodysplastic Syndrome and Oligoblastic Acute Myeloid Leukemia Treated with Azacytidine. Leuk. Lymphoma 2019, 60, 2457–2463. [Google Scholar] [CrossRef]
  36. 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]
  37. Ma, L.; Ceuppens, J.; Kasran, A.; Delforge, M.; Boogaerts, M.; Vandenberghe, P. Immature and Mature Monocyte-Derived Dendritic Cells in Myelodysplastic Syndromes of Subtypes Refractory Anemia or Refractory Anemia with Ringed Sideroblasts Display an Altered Cytokine Profile. Leuk. Res. 2007, 31, 1373–1382. [Google Scholar] [CrossRef]
  38. Van Leeuwen-Kerkhoff, N.; Lundberg, K.; Westers, T.M.; Kordasti, S.; Bontkes, H.J.; Lindstedt, M.; de Gruijl, T.D.; van de Loosdrecht, A.A. Human Bone Marrow-Derived Myeloid Dendritic Cells Show an Immature Transcriptional and Functional Profile Compared to Their Peripheral Blood Counterparts and Separate from Slan+ Non-Classical Monocytes. Front. Immunol. 2018, 9, 1619. [Google Scholar] [CrossRef]
  39. Ferlazzo, G.; Moretta, L. Dendritic Cell Editing by Natural Killer Cells. Crit. Rev. Oncog. 2014, 19, 67–75. [Google Scholar] [CrossRef]
  40. Pruneri, G.; Bertolini, F.; Soligo, D.; Carboni, N.; Cortelezzi, A.; Ferrucci, P.F.; Buffa, R.; Lambertenghi-Deliliers, G.; Pezzella, F. Angiogenesis in Myelodysplastic Syndromes. Br. J. Cancer 1999, 81, 1398–1401. [Google Scholar] [CrossRef] [Green Version]
  41. Invernizzi, R.; Travaglino, E.; Della Porta, M.G.; Malcovati, L.; Gallì, A.; Bastia, R.; Ciola, M.; Ambaglio, I.; Boveri, E.; Rosti, V.; et al. Vascular Endothelial Growth Factor Overexpression in Myelodysplastic Syndrome Bone Marrow Cells: Biological and Clinical Implications. Leuk. Lymphoma 2017, 58, 1711–1720. [Google Scholar] [CrossRef] [PubMed]
  42. Gabrilovich, D.I.; Chen, H.L.; Girgis, K.R.; Cunningham, H.T.; Meny, G.M.; Nadaf, S.; Kavanaugh, D.; Carbone, D.P. Production of Vascular Endothelial Growth Factor by Human Tumors Inhibits the Functional Maturation of Dendritic Cells. Nat. Med. 1996, 2, 1096–1103. [Google Scholar] [CrossRef] [PubMed]
  43. Terme, M.; Tartour, E.; Taieb, J. VEGFA/VEGFR2-Targeted Therapies Prevent the VEGFA-Induced Proliferation of Regulatory T Cells in Cancer. Oncoimmunology 2013, 2, e25156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Kordasti, S.Y.; Ingram, W.; Hayden, J.; Darling, D.; Barber, L.; Afzali, B.; Lombardi, G.; Wlodarski, M.W.; Maciejewski, J.P.; Farzaneh, F.; et al. CD4+ CD25high Foxp3+ Regulatory T Cells in Myelodysplastic Syndrome (MDS). Blood 2007, 110, 847–850. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Hamdi, W.; Ogawara, H.; Handa, H.; Tsukamoto, N.; Nojima, Y.; Murakami, H. Clinical Significance of Regulatory T Cells in Patients with Myelodysplastic Syndrome. Eur. J. Haematol. 2009, 82, 201–207. [Google Scholar] [CrossRef]
  46. Kahn, J.D.; Chamuleau, M.E.D.; Westers, T.M.; van de Ven, P.M.; van Dreunen, L.; van Spronsen, M.; Ossenkoppele, G.J.; van de Loosdrecht, A.A. Regulatory T Cells and Progenitor B Cells Are Independent Prognostic Predictors in Lower Risk Myelodysplastic Syndromes. Haematologica 2015, 100, e220–e222. [Google Scholar] [CrossRef] [Green Version]
  47. Dudina, G.A.; Donetskova, A.D.; Litvina, M.M.; Mitin, A.N.; Mitina, T.A.; Polyakov, S.A. Regulatory T Cells and Profile of FOXP3 Isoforms Expression in Peripheral Blood of Patients with Myelodysplastic Syndromes. Adv. Hematol. 2018, 2018, 8487403. [Google Scholar] [CrossRef] [Green Version]
  48. Giovazzino, A.; Leone, S.; Rubino, V.; Palatucci, A.T.; Cerciello, G.; Alfinito, F.; Pane, F.; Ruggiero, G.; Terrazzano, G. Reduced Regulatory T Cells (Treg) in Bone Marrow Preferentially Associate with the Expansion of Cytotoxic T Lymphocytes in Low Risk MDS Patients. Br. J. Haematol. 2019, 185, 357–360. [Google Scholar] [CrossRef] [Green Version]
  49. Ghiringhelli, F.; Ménard, C.; Terme, M.; Flament, C.; Taieb, J.; Chaput, N.; Puig, P.E.; Novault, S.; Escudier, B.; Vivier, E.; et al. CD4+CD25+ Regulatory T Cells Inhibit Natural Killer Cell Functions in a Transforming Growth Factor–β–Dependent Manner. J. Exp. Med. 2005, 202, 1075–1085. [Google Scholar] [CrossRef]
  50. Szczepanski, M.J.; Szajnik, M.; Czystowska, M.; Mandapathil, M.; Strauss, L.; Welsh, A.; Foon, K.A.; Whiteside, T.L.; Boyiadzis, M. Increased Frequency and Suppression by Regulatory T Cells in Patients with Acute Myelogenous Leukemia. Clin. Cancer Res. 2009, 15, 3325–3332. [Google Scholar] [CrossRef] [Green Version]
  51. Hallett, W.H.D.; Ames, E.; Álvarez, M.; Barao, I.; Taylor, P.A.; Blazar, B.R.; Murphy, W.J. Combination Therapy Using IL-2 and Anti-CD25 Results in Augmented Natural Killer Cell-Mediated Antitumor Responses. Biol. Blood Marrow Transplant. 2008, 14, 1088–1099. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Chen, X.; Eksioglu, E.A.; Zhou, J.; Zhang, L.; Djeu, J.; Fortenbery, N.; Epling-Burnette, P.; van Bijnen, S.; Dolstra, H.; Cannon, J.; et al. Induction of Myelodysplasia by Myeloid-Derived Suppressor Cells. J. Clin. Investig. 2013, 123, 4595–4611. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Jiang, H.J.; Fu, R.; Wang, H.Q.; Li, L.J.; Qu, W.; Liang, Y.; Wang, G.J.; Wang, X.M.; Wu, Y.H.; Liu, H.; et al. Increased Circulating of Myeloid-Derived Suppressor Cells in Myelodysplastic Syndrome. Chin. Med. J. 2013, 126, 2582–2584. [Google Scholar] [CrossRef] [PubMed]
  54. Kittang, A.O.; Kordasti, S.; Sand, K.E.; Costantini, B.; Kramer, A.M.; Perezabellan, P.; Seidl, T.; Rye, K.P.; Hagen, K.M.; Kulasekararaj, A.; et al. Expansion of Myeloid Derived Suppressor Cells Correlates with Number of T Regulatory Cells and Disease Progression in Myelodysplastic Syndrome. Oncoimmunology 2016, 5, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Matsuda, M.; Morita, Y.; Hanamoto, H.; Tatsumi, Y.; Maeda, Y.; Kanamaru, A. CD34+ Progenitors from MDS Patients Are Unresponsive to SDF-1, despite High Levels of SDF-1 in Bone Marrow Plasma. Leukemia 2004, 18, 1038–1040. [Google Scholar] [CrossRef] [Green Version]
  56. Palumbo, G.A.; Parrinello, N.L.; Giallongo, C.; D’Amico, E.; Zanghì, A.; Puglisi, F.; Conticello, C.; Chiarenza, A.; Tibullo, D.; Raimondo, F.D.; et al. Monocytic Myeloid Derived Suppressor Cells in Hematological Malignancies. Int. J. Mol. Sci. 2019, 20, 5459. [Google Scholar] [CrossRef] [Green Version]
  57. Lv, M.; Wang, K.; Huang, X. Myeloid-Derived Suppressor Cells in Hematological Malignancies: Friends or Foes. J. Hematol. Oncol. 2019, 12, 105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Mellqvist, U.H.; Brune, M.; Dahlgren, C.; Hellstrand, K. Natural Killer Cell Dysfunction and Apoptosis Induced by CML Cells: Role of Reactive Oxygen Species and Regulation by Histamine. Blood 2000, 96, 1961–1968. [Google Scholar] [CrossRef]
  59. Aurelius, J.; Martner, A.; Riise, R.E.; Romero, A.I.; Palmqvist, L.; Brune, M.; Hellstrand, K.; Thorén, F.B. Chronic Myeloid Leukemic Cells Trigger Poly (ADP-ribose) Polymerase-dependent Inactivation and Cell Death in Lymphocytes. J. Leukoc. Biol. 2013, 93, 155–160. [Google Scholar] [CrossRef]
  60. Aurelius, J.; Hallner, A.; Werlenius, O.; Riise, R.; Moüllgård, L.; Brune, M.; Hansson, M.; Martner, A.; Thorén, F.B.; Hellstrand, K. NOX2-dependent Immunosuppression in Chronic Myelomonocytic Leukemia. J. Leukoc. Biol. 2017, 102, 459–466. [Google Scholar] [CrossRef] [Green Version]
  61. Ghoti, H.; Fibach, E.; Merkel, D.; Perez-Avraham, G.; Grisariu, S.; Rachmilewitz, E.A. Changes in Parameters of Oxidative Stress and Free Iron Biomarkers during Treatment with Deferasirox in Iron-Overloaded Patients with Myelodysplastic Syndromes. Haematologica 2010, 95, 1433–1434. [Google Scholar] [CrossRef] [PubMed]
  62. Angelucci, E.; Cianciulli, P.; Finelli, C.; Mecucci, C.; Voso, M.T.; Tura, S. Unraveling the Mechanisms behind Iron Overload and Ineffective Hematopoiesis in Myelodysplastic Syndromes. Leuk. Res. 2017, 62, 108–115. [Google Scholar] [CrossRef] [PubMed]
  63. Hua, Y.; Wang, C.; Jiang, H.; Wang, Y.; Liu, C.; Li, L.; Liu, H.; Shao, Z.; Fu, R. Iron Overload May Promote Alteration of NK Cells and Hematopoietic Stem/Progenitor Cells by JNK and P38 Pathway in Myelodysplastic Syndromes. Int. J. Hematol. 2017, 106, 248–257. [Google Scholar] [CrossRef] [PubMed]
  64. Corradi, G.; Baldazzi, C.; Očadlíková, D.; Marconi, G.; Parisi, S.; Testoni, N.; Finelli, C.; Cavo, M.; Curti, A.; Ciciarello, M. Mesenchymal Stromal Cells from Myelodysplastic and Acute Myeloid Leukemia Patients Display in Vitro Reduced Proliferative Potential and Similar Capacity to Support Leukemia Cell Survival. Stem Cell Res. 2018, 9, 271. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Geyh, S.; Öz, S.; Cadeddu, R.P.; Fröbel, J.; Brückner, B.; Kündgen, A.; Fenk, R.; Bruns, I.; Zilkens, C.; Hermsen, D.; et al. Insufficient Stromal Support in MDS Results from Molecular and Functional Deficits of Mesenchymal Stromal Cells. Leukemia 2013, 27, 1841–1851. [Google Scholar] [CrossRef] [Green Version]
  66. Poon, Z.; Dighe, N.; Venkatesan, S.S.; Cheung, A.M.S.; Fan, X.; Bari, S.; Hota, M.; Ghosh, S.; Hwang, W.Y.K. Bone Marrow MSCs in MDS: Contribution towards Dysfunctional Hematopoiesis and Potential Targets for Disease Response to Hypomethylating Therapy. Leukemia 2019, 33, 1487–1500. [Google Scholar] [CrossRef] [Green Version]
  67. Zhao, X.; Weinhold, S.; Brands, J.; Hejazi, M.; Degistirici, Ö.; Kögler, G.; Meisel, R.; Uhrberg, M. NK Cell Development in a Human Stem Cell Niche: KIR Expression Occurs Independently of the Presence of HLA Class i Ligands. Blood Adv. 2018, 2, 2452–2461. [Google Scholar] [CrossRef]
  68. Frias, A.M.; Porada, C.D.; Crapnell, K.B.; Cabral, J.M.S.; Zanjani, E.D.; Almeida-Porada, G. Generation of Functional Natural Killer and Dendritic Cells in a Human Stromal-Based Serum-Free Culture System Designed for Cord Blood Expansion. Exp. Hematol. 2008, 36, 61–68. [Google Scholar] [CrossRef] [Green Version]
  69. Miller, J.S.; McCullar, V. Human Natural Killer Cells with Polyclonal Lectin and Immunoglobulinlike Receptors Develop from Single Hematopoietic Stem Cells with Preferential Expression of NKG2A and KIR2DL2/L3/S2. Blood 2001, 98, 705–713. [Google Scholar] [CrossRef]
  70. Lopes, M.R.; Pereira, J.K.N.; de Melo Campos, P.; Machado-Neto, J.A.; Traina, F.; Saad, S.T.O.; Favaro, P. De Novo AML Exhibits Greater Microenvironment Dysregulation Compared to AML with Myelodysplasia-Related Changes. Sci. Rep. 2017, 7, 40707. [Google Scholar] [CrossRef] [Green Version]
  71. Zeng, W.; Miyazato, A.; Chen, G.; Kajigaya, S.; Young, N.S.; Maciejewski, J.P. Interferon-Gamma-Induced Gene Expression in CD34 Cells: Identification of Pathologic Cytokine-Specific Signature Profiles. Blood 2006, 107, 167–175. [Google Scholar] [CrossRef] [Green Version]
  72. Kerbauy, D.B.; Deeg, H.J. Apoptosis and Antiapoptotic Mechanisms in the Progression of Myelodysplastic Syndrome. Exp. Hematol. 2007, 35, 1739–1746. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Hayden, M.S.; Ghosh, S. Regulation of NF-ΚB by TNF Family Cytokines. Semin. Immunol. 2014, 26, 253–266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Kerbauy, D.M.B.; Lesnikov, V.; Abbasi, N.; Seal, S.; Scott, B.; Deeg, H.J. NF-ΚB and FLIP in Arsenic Trioxide (ATO)-Induced Apoptosis in Myelodysplastic Syndromes (MDSs). Blood 2005, 106, 3917–3925. [Google Scholar] [CrossRef] [PubMed]
  75. Kagoya, Y.; Yoshimi, A.; Kataoka, K.; Nakagawa, M.; Kumano, K.; Arai, S.; Kobayashi, H.; Saito, T.; Iwakura, Y.; Kurokawa, M. Positive Feedback between NF-ΚB and TNF-α Promotes Leukemia-Initiating Cell Capacity. J. Clin. Investig. 2014, 124, 528–542. [Google Scholar] [CrossRef] [Green Version]
  76. Masala, E.; Valencia-Martinez, A.; Pillozzi, S.; Rondelli, T.; Brogi, A.; Sanna, A.; Gozzini, A.; Arcangeli, A.; Sbarba, P.D.; Santini, V. Severe Hypoxia Selects Hematopoietic Progenitors with Stem Cell Potential from Primary Myelodysplastic Syndrome Bone Marrow Cell Cultures. Oncotarget 2018, 9, 10561–10571. [Google Scholar] [CrossRef] [Green Version]
  77. Schinke, C.; Giricz, O.; Li, W.; Shastri, A.; Gordon, S.; Barreyro, L.; Bhagat, T.; Bhattacharyya, S.; Ramachandra, N.; Bartenstein, M.; et al. IL8-CXCR2 Pathway Inhibition as a Therapeutic Strategy against MDS and AML Stem Cells. Blood 2015, 125, 3144–3152. [Google Scholar] [CrossRef] [Green Version]
  78. Kuett, A.; Rieger, C.; Perathoner, D.; Herold, T.; Wagner, M.; Sironi, S.; Sotlar, K.; Horny, H.P.; Deniffel, C.; Drolle, H.; et al. IL-8 as Mediator in the Microenvironment-Leukaemia Network in Acute Myeloid Leukaemia. Sci. Rep. 2015, 5, 18411. [Google Scholar] [CrossRef] [Green Version]
  79. Zhou, L.; Nguyen, A.N.; Sohal, D.; Ying Ma, J.; Pahanish, P.; Gundabolu, K.; Hayman, J.; Chubak, A.; Mo, Y.; Bhagat, T.D.; et al. Inhibition of the TGF-Beta Receptor I Kinase Promotes Hematopoiesis in MDS. Blood 2008, 112, 3434–3443. [Google Scholar] [CrossRef] [Green Version]
  80. Bhagat, T.D.; Zhou, L.; Sokol, L.; Kessel, R.; Caceres, G.; Gundabolu, K.; Tamari, R.; Gordon, S.; Mantzaris, I.; Jodlowski, T.; et al. MiR-21 Mediates Hematopoietic Suppression in MDS by Activating TGF-β Signaling. Blood 2013, 121, 2875–2881. [Google Scholar] [CrossRef] [Green Version]
  81. Zhou, L.; McMahon, C.; Bhagat, T.; Alencar, C.; Yu, Y.; Fazzari, M.; Sohal, D.; Heuck, C.; Gundabolu, K.; Ng, C.; et al. Reduced SMAD7 Leads to Overactivation of TGF-Beta Signaling in MDS That Can Be Reversed by a Specific Inhibitor of TGF-Beta Receptor I Kinase. Cancer Res. 2011, 71, 955–963. [Google Scholar] [CrossRef] [Green Version]
  82. Geyh, S.; Rodríguez-Paredes, M.; Jäger, P.; Koch, A.; Bormann, F.; Gutekunst, J.; Zilkens, C.; Germing, U.; Kobbe, G.; Lyko, F.; et al. Transforming Growth Factor Β1-Mediated Functional Inhibition of Mesenchymal Stromal Cells in Myelodysplastic Syndromes and Acute Myeloid Leukemia. Haematologica 2018, 103, 1462–1471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Peddie, C.M.; Wolf, C.R.; McLellan, L.I.; Collins, A.R.; Bowen, D.T. Oxidative DNA Damage in CD34+ Myelodysplastic Cells Is Associated with Intracellular Redox Changes and Elevated Plasma Tumour Necrosis Factor-Alpha Concentration. Br. J. Haematol. 1997, 99, 625–631. [Google Scholar] [CrossRef] [PubMed]
  84. Gonçalves, A.C.; Cortesão, E.; Oliveiros, B.; Alves, V.; Espadana, A.I.; Rito, L.; Magalhães, E.; Lobão, M.J.; Pereira, A.; Nascimento Costa, J.M.; et al. Oxidative Stress and Mitochondrial Dysfunction Play a Role in Myelodysplastic Syndrome Development, Diagnosis, and Prognosis: A Pilot Study. Free. Radic. Res. 2015, 49, 1081–1094. [Google Scholar] [CrossRef] [PubMed]
  85. Picou, F.; Vignon, C.; Debeissat, C.; Lachot, S.; Kosmider, O.; Gallay, N.; Foucault, A.; Estienne, M.H.; Ravalet, N.; Bene, M.C.; et al. Bone Marrow Oxidative Stress and Specific Antioxidant Signatures in Myelodysplastic Syndromes. Blood Adv. 2019, 3, 4271–4279. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Chen, S.; Zambetti, N.A.; Bindels, E.M.J.; Kenswill, K.; Mylona, A.M.; Adisty, N.M.; Hoogenboezem, R.M.; Sanders, M.A.; Cremers, E.M.P.; Westers, T.M.; et al. Massive Parallel RNA Sequencing of Highly Purified Mesenchymal Elements in Low-Risk MDS Reveals Tissue-Context-Dependent Activation of Inflammatory Programs. Leukemia 2016, 30, 1938–1942. [Google Scholar] [CrossRef] [Green Version]
  87. Choi, I.; Yoon, S.R.; Park, S.Y.; Kim, H.; Jung, S.J.; Kang, Y.L.; Lee, J.H.; Lee, J.H.; Kim, D.Y.; Lee, J.L.; et al. Donor-Derived Natural Killer Cell Infusion after Human Leukocyte Antigen–Haploidentical Hematopoietic Cell Transplantation in Patients with Refractory Acute Leukemia. Biol. Blood Marrow Transplant. 2016, 22, 2065–2076. [Google Scholar] [CrossRef] [Green Version]
  88. Sabry, M.; Lowdell, M.W. Killers at the Crossroads: The Use of Innate Immune Cells in Adoptive Cellular Therapy of Cancer. Stem Cells Transl. Med. 2020, 9, 974–984. [Google Scholar] [CrossRef]
  89. Bachanova, V.; Cooley, S.; Defor, T.E.; Verneris, M.R.; Zhang, B.; Mckenna, D.H.; Curtsinger, J.; Panoskaltsis-mortari, A.; Lewis, D.; Hippen, K.; et al. Clearance of Acute Myeloid Leukemia by Haploidentical Natural Killer Cells Is Improved Using IL-2 Diphtheria Toxin Fusion Protein. Blood 2014, 123, 3855–3863. [Google Scholar] [CrossRef]
  90. Ito, S.; Bollard, C.M.; Carlsten, M.; Melenhorst, J.J.; Biancotto, A.; Wang, E.; Chen, J.; Kotliarov, Y.; Cheung, F.; Xie, Z.; et al. Ultra-Low Dose Interleukin-2 Promotes Immune-Modulating Function of Regulatory T Cells and Natural Killer Cells in Healthy Volunteers. Mol. Ther. 2014, 22, 1388–1395. [Google Scholar] [CrossRef] [Green Version]
  91. Geller, M.A.; Cooley, S.; Judson, P.L.; Ghebre, R.; Carson, L.F.; Argenta, P.A.; Jonson, A.L.; Panoskaltsis-Mortari, A.; Curtsinger, J.; McKenna, D.; et al. A Phase II Study of Allogeneic Natural Killer Cell Therapy to Treat Patients with Recurrent Ovarian and Breast Cancer. Cytotherapy 2011, 13, 98–107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Alvarez, M.; Bouchlaka, M.N.; Sckisel, G.D.; Sungur, C.M.; Chen, M.; Murphy, W.J. Increased Antitumor Effects Using IL-2 with Anti–TGF-β Reveals Competition between Mouse NK and CD8 T Cells. J. Immunol. 2014, 193, 1709–1716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Alvarez, M.; Dunai, C.; Khuat, L.T.; Aguilar, E.G.; Barao, I.; Murphy, W.J. IL-2 and Anti-TGF-β Promote NK Cell Reconstitution and Anti-Tumor Effects after Syngeneic Hematopoietic Stem Cell Transplantation. Cancers 2020, 12, 3189. [Google Scholar] [CrossRef] [PubMed]
  94. Tian, Z.; Wei, H. Developmental and Functional Control of Natural Killer Cells by Cytokines. Front. Immunol. 2017, 8, 930. [Google Scholar] [CrossRef] [Green Version]
  95. Yang, Y.; Lundqvist, A. Immunomodulatory E Ff Ects of IL-2 and IL-15; Implications for Cancer Immunotherapy. Cancers 2020, 2, 3586. [Google Scholar] [CrossRef]
  96. Cooley, S.; He, F.; Bachanova, V.; Vercellotti, G.M.; Defor, T.E.; Curtsinger, J.M.; Robertson, P.; Grzywacz, B.; Conlon, K.C.; Waldmann, T.A.; et al. First-in-Human Trial of RhIL-15 and Haploidentical Natural Killer Cell Therapy for Advanced Acute Myeloid Leukemia. Blood Adv. 2019, 3, 1970–1980. [Google Scholar] [CrossRef]
  97. Cooper, M.A.; Elliott, J.M.; Keyel, P.A.; Yang, L.; Carrero, J.A.; Yokoyama, W.M. Cytokine-Induced Memory-like Natural Killer Cells. Proc. Natl. Acad. Sci. USA 2009, 106, 1915–1919. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Romee, R.; Schneider, S.E.; Leong, J.W.; Chase, J.M.; Keppel, C.R.; Sullivan, R.P.; Cooper, M.A.; Fehniger, T.A. Cytokine Activation Induces Human Memory-like NK Cells. Blood 2012, 120, 4751–4760. [Google Scholar] [CrossRef] [Green Version]
  99. 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]
  100. 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]
  101. Imai, C.; Iwamoto, S.; Campana, D. Genetic Modification of Primary Natural Killer Cells Overcomes Inhibitory Signals and Induces Specific Killing of Leukemic Cells. Blood 2005, 106, 376–383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Fujisaki, H.; Kakuda, H.; Imai, C.; Mullighan, C.G.; Campana, D. Replicative Potential of Human Natural Killer Cells. Br. J. Haematol 2009, 145, 606–613. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Ciurea, S.O.; Schafer, J.R.; Bassett, R.; Denman, C.J.; Cao, K.; Willis, D.; Rondon, G.; Chen, J.; Soebbing, D.; Kaur, I.; et al. Phase 1 Clinical Trial Using MbIL21 Ex Vivo–Expanded Donor-Derived NK Cells after Haploidentical Transplantation. Blood 2017, 130, 1857–1868. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Denman, C.J.; Senyukov, V.V.; Somanchi, S.S.; Phatarpekar, P.V.; Kopp, L.M.; Johnson, J.L.; Singh, H.; Hurton, L.; Maiti, S.N.; Huls, M.H.; et al. Membrane-Bound IL-21 Promotes Sustained Ex Vivo Proliferation of Human Natural Killer Cells. PLoS ONE 2012, 7, e30264. [Google Scholar] [CrossRef]
  105. Vasu, S.; Bhatnagar, B.; Blachly, J.S.; Szuminski, N.; O’Donnell, L.; Lee, D.A. A Phase I Clinical Trial Testing the Safety of IL-21-Expanded, Off-the-Shelf, Third-Party Natural Killer Cells for Relapsed/Refractory Acute Myeloid Leukemia and Myelodysplastic Syndrome. Blood 2020, 136, 44. [Google Scholar] [CrossRef]
  106. Brentjens, R.J.; Davila, M.L.; Riviere, I.; Park, J.; Wang, X.; Cowell, L.G.; Bartido, S.; Stefanski, J.; Taylor, C.; Olszewska, M.; et al. CD19-Targeted T Cells Rapidly Induce Molecular Remissions in Adults with Chemotherapy-Refractory Acute Lymphoblastic Leukemia. Sci. Transl. Med. 2013, 5, 177ra38. [Google Scholar] [CrossRef] [Green Version]
  107. Kochenderfer, J.N.; Wilson, W.H.; Janik, J.E.; Dudley, M.E.; Stetler-Stevenson, M.; Feldman, S.A.; Maric, I.; Raffeld, M.; Nathan, D.-A.N.; Lanier, B.J.; et al. Eradication of B-Lineage Cells and Regression of Lymphoma in a Patient Treated with Autologous T Cells Genetically Engineered to Recognize CD19. Blood 2010, 116, 4099–4102. [Google Scholar] [CrossRef]
  108. 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]
  109. Ritchie, D.S.; Neeson, P.J.; Khot, A.; Peinert, S.; Tai, T.; Tainton, K.; Chen, K.; Shin, M.; Wall, D.M.; Hönemann, D.; et al. Persistence and Efficacy of Second Generation CAR T Cell against the LeY Antigen in Acute Myeloid Leukemia. Mol. Ther. 2013, 21, 2122–2129. [Google Scholar] [CrossRef] [Green Version]
  110. Wang, Q.S.; Wang, Y.; Lv, H.Y.; Han, Q.W.; Fan, H.; Guo, B.; Wang, L.L.; Han, W.D. Treatment of CD33-Directed Chimeric Antigen Receptor-Modified T Cells in One Patient with Relapsed and Refractory Acute Myeloid Leukemia. Mol. Ther. 2015, 23, 184–191. [Google Scholar] [CrossRef] [Green Version]
  111. Baumeister, S.H.; Murad, J.; Werner, L.; Daley, H.; Trebeden-Negre, H.; Gicobi, J.K.; Schmucker, A.; Reder, J.; Sentman, C.L.; Gilham, D.E.; et al. Phase I Trial of Autologous CAR T Cells Targeting NKG2D Ligands in Patients with AML/MDS and Multiple Myeloma. Cancer Immunol. Res. 2019, 7, 100–112. [Google Scholar] [CrossRef] [PubMed]
  112. Zhang, H.; Gan, W.-T.; Hao, W.-G.; Wang, P.-F.; Li, Z.-Y.; Chang, L.-J. Successful Anti-CLL1 CAR T-Cell Therapy in Secondary Acute Myeloid Leukemia. Leukemia. Front. Oncol. 2020, 10, 685. [Google Scholar] [CrossRef] [PubMed]
  113. Zhang, H.; Wang, P.; Li, Z.; He, Y.; Gan, W.; Jiang, H. Anti-CLL1 Chimeric Antigen Receptor T-Cell Therapy in Children with Relapsed/Refractory Acute Myeloid Leukemia. Clin. Cancer Res. 2021, 27, 3549–3555. [Google Scholar] [CrossRef] [PubMed]
  114. Neelapu, S.S.; Tummala, S.; Kebriaei, P.; Wierda, W.; Gutierrez, C.; Locke, F.L.; Komanduri, K.V.; Lin, Y.; Jain, N.; Daver, N.; et al. Chimeric Antigen Receptor T-Cell Therapy-Assessment and Management of Toxicities. Nat. Rev. Clin. Oncol. 2018, 15, 47–62. [Google Scholar] [CrossRef] [PubMed]
  115. Siegler, E.L.; Zhu, Y.; Wang, P.; Yang, L. Off-the-Shelf CAR-NK Cells for Cancer Immunotherapy. Cell Stem Cell 2018, 23, 160–161. [Google Scholar] [CrossRef] [Green Version]
  116. Xie, G.; Dong, H.; Liang, Y.; Ham, J.D.; Rizwan, R.; Chen, J. CAR-NK Cells: A Promising Cellular Immunotherapy for Cancer. EBioMedicine 2020, 59, 102975. [Google Scholar] [CrossRef]
  117. Wang, W.; Jiang, J.; Wu, C. CAR-NK for Tumor Immunotherapy: Clinical Transformation and Future Prospects. Cancer Lett. 2020, 472, 175–180. [Google Scholar] [CrossRef]
  118. Gurney, M.; O’dwyer, M.; Fracchiolla, S.; Onida, F. Realizing Innate Potential: CAR-NK Cell Therapies for Acute Myeloid Leukemia. Cancers 2020, 12, 1752. [Google Scholar] [CrossRef]
  119. Haubner, S.; Perna, F.; Köhnke, T.; Schmidt, C.; Berman, S.; Augsberger, C.; Schnorfeil, F.M.; Krupka, C.; Lichtenegger, F.S.; Liu, X.; et al. Coexpression Profile of Leukemic Stem Cell Markers for Combinatorial Targeted Therapy in AML. Leukemia 2019, 33, 64–74. [Google Scholar] [CrossRef]
  120. Tang, X.; Yang, L.; Li, Z.; Nalin, A.P.; Dai, H.; Xu, T.; Yin, J.; You, F.; Zhu, M.; Shen, W.; et al. First-in-Man Clinical Trial of CAR NK-92 Cells: Safety Test of CD33-CAR NK-92 Cells in Patients with Relapsed and Refractory Acute Myeloid Leukemia. Am. J. Cancer Res. 2018, 8, 1899. [Google Scholar]
  121. Sinha, C.; Seth, A.; Kahali, B.; Cunningham, L. Development and Evaluation of NK-CD123 CAR Against High Risk Acute Myeloid Leukemia. Biol. Blood Marrow Transplant. 2017, 23, S253. [Google Scholar] [CrossRef] [Green Version]
  122. Dong, H.; Xie, G.; Liang, Y.; Dongjoo Ham, J.; Vergara, J.; Chen, J.; Ritz, J.; Romee, R. Engineered Memory-like NK Cars Targeting a Neoepitope Derived from Intracellular NPM1c Exhibit Potent Activity and Specificity Against Acute Myeloid Leukemia. Blood 2020, 136, 3–4. [Google Scholar] [CrossRef]
  123. Felices, M.; Sarhan, D.; Brandt, L.; Guldevall, K.; McElmurry, R.; Lenvik, A.; Chu, S.; Tolar, J.; Taras, E.; Spellman, S.R.; et al. CD16-IL15-CD33 Trispecific Killer Engager (TriKE) Overcomes Cancer-Induced Immune Suppression and Induces Natural Killer Cell-Mediated Control of MDS and AML Via Enhanced Killing Kinetics. Blood 2016, 128, 4291. [Google Scholar] [CrossRef]
  124. Sarhan, D.; Brandt, L.; Felices, M.; Guldevall, K.; Lenvik, T.; Hinderlie, P.; Curtsinger, J.; Warlick, E.; Spellman, S.R.; Blazar, B.R.; et al. 161533 TriKE Stimulates NK-Cell Function to Overcome Myeloid-Derived Suppressor Cells in MDS. Blood Adv. 2018, 2, 1459–1469. [Google Scholar] [CrossRef] [Green Version]
  125. Sabry, M.; Lowdell, M.W. Tumor-Primed NK Cells: Waiting for the Green Light. Front. Immunol. 2013, 4, 00408. [Google Scholar] [CrossRef] [Green Version]
  126. North, J.; Bakhsh, I.; Marden, C.; Pittman, H.; Addison, E.; Navarrete, C.; Anderson, R.; Lowdell, M.W. Tumor-Primed Human Natural Killer Cells Lyse NK-Resistant Tumor Targets: Evidence of a Two-Stage Process in Resting NK Cell Activation. J. Immunol. 2007, 178, 85–94. [Google Scholar] [CrossRef] [Green Version]
  127. Sabry, M.; Tsirogianni, M.; Bakhsh, I.A.; North, J.; Sivakumaran, J.; Giannopoulos, K.; Anderson, R.; Mackinnon, S.; Lowdell, M.W. Leukemic Priming of Resting NK Cells Is Killer Ig-like Receptor Independent but Requires CD15-Mediated CD2 Ligation and Natural Cytotoxicity Receptors. J. Immunol. 2011, 187, 6227–6234. [Google Scholar] [CrossRef] [Green Version]
  128. Sabry, M.; Zubiak, A.; Hood, S.P.; Simmonds, P.; Arellano-Ballestero, H.; Cournoyer, E.; Mashar, M.; Graham Pockley, A.; Lowdell, M.W. Tumor- And Cytokine-Primed Human Natural Killer Cells Exhibit Distinct Phenotypic and Transcriptional Signatures. PLoS ONE 2018, 14, e0218674. [Google Scholar] [CrossRef] [Green Version]
  129. Kottaridis, P.D.; North, J.; Tsirogianni, M.; Marden, C.; Samuel, E.R.; Jide-Banwo, S.; Grace, S.; Lowdell, M.W. Two-Stage Priming of Allogeneic Natural Killer Cells for the Treatment of Patients with Acute Myeloid Leukemia: A Phase I Trial. PLoS ONE 2015, 10, e0123416. [Google Scholar] [CrossRef]
  130. Fehniger, T.A.; Miller, J.S.; Stuart, R.K.; Cooley, S.; Salhotra, A.; Curtsinger, J.; Westervelt, P.; DiPersio, J.F.; Hillman, T.M.; Silver, N.; et al. A Phase 1 Trial of CNDO-109–Activated Natural Killer Cells in Patients with High-Risk Acute Myeloid Leukemia. Biol. Blood Marrow Transplant. 2018, 24, 1581–1589. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Stages of NK cell development and function. Schematic representation of the different stages of NK cell differentiation and maturation in humans. HSCs differentiate into CPLs and then into NKPs. The acquisition of CD56 will determine the last step into NK cells. CD56bright NK cells represent a more immature subset that can develop into CD56dim NK cells. Both subsets are capable of cytokine secretion, but CD56dim NK cells are more cytotoxic. Direct cytotoxicity can be exerted via granule secretion, death receptor ligation, or ADCC.
Figure 1. Stages of NK cell development and function. Schematic representation of the different stages of NK cell differentiation and maturation in humans. HSCs differentiate into CPLs and then into NKPs. The acquisition of CD56 will determine the last step into NK cells. CD56bright NK cells represent a more immature subset that can develop into CD56dim NK cells. Both subsets are capable of cytokine secretion, but CD56dim NK cells are more cytotoxic. Direct cytotoxicity can be exerted via granule secretion, death receptor ligation, or ADCC.
Cells 12 00633 g001
Figure 2. MDS impairs NK cell function through TME factors. The presence of immature DCs directly affects NK cell activation. Increased levels of VEGF contribute to DC immaturity and also Treg expansion. Increased levels of hyperactive Tregs secrete IL-10 and TGF-β into the TME, suppressing NK cell activation. A source of VEGF in MDS are MDSCs, which are expanded and hyperactive and contribute to NK cell impairment by secreting IL-10, TGF-β, and ROS into the TME. Another source of ROS are blood transfusions received by MDS patients as part of their treatment. MSCs are also reported to be reduced in numbers and have an impaired phenotype in MDS; impaired MSCs are not able to support NK cell maturation and contribute to general inflammation by secreting pro-inflammatory cytokines such as IL-6. The general inflammatory state in the bone marrow of MDS patients is characterized by an increase of IFN-γ and TNF-α and increased hypoxia, inducing bone marrow apoptosis that helps in the selection of malignant CD34 cells. Those cells further secrete ROS and pro-inflammatory cytokines such as IL-8 and TGF-β, impairing directly or indirectly NK cell function and activation.
Figure 2. MDS impairs NK cell function through TME factors. The presence of immature DCs directly affects NK cell activation. Increased levels of VEGF contribute to DC immaturity and also Treg expansion. Increased levels of hyperactive Tregs secrete IL-10 and TGF-β into the TME, suppressing NK cell activation. A source of VEGF in MDS are MDSCs, which are expanded and hyperactive and contribute to NK cell impairment by secreting IL-10, TGF-β, and ROS into the TME. Another source of ROS are blood transfusions received by MDS patients as part of their treatment. MSCs are also reported to be reduced in numbers and have an impaired phenotype in MDS; impaired MSCs are not able to support NK cell maturation and contribute to general inflammation by secreting pro-inflammatory cytokines such as IL-6. The general inflammatory state in the bone marrow of MDS patients is characterized by an increase of IFN-γ and TNF-α and increased hypoxia, inducing bone marrow apoptosis that helps in the selection of malignant CD34 cells. Those cells further secrete ROS and pro-inflammatory cytokines such as IL-8 and TGF-β, impairing directly or indirectly NK cell function and activation.
Cells 12 00633 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Arellano-Ballestero, H.; Sabry, M.; Lowdell, M.W. A Killer Disarmed: Natural Killer Cell Impairment in Myelodysplastic Syndrome. Cells 2023, 12, 633. https://doi.org/10.3390/cells12040633

AMA Style

Arellano-Ballestero H, Sabry M, Lowdell MW. A Killer Disarmed: Natural Killer Cell Impairment in Myelodysplastic Syndrome. Cells. 2023; 12(4):633. https://doi.org/10.3390/cells12040633

Chicago/Turabian Style

Arellano-Ballestero, Helena, May Sabry, and Mark W. Lowdell. 2023. "A Killer Disarmed: Natural Killer Cell Impairment in Myelodysplastic Syndrome" Cells 12, no. 4: 633. https://doi.org/10.3390/cells12040633

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