Next Article in Journal
Deep Learning-Based Artificial Intelligence System for Automatic Assessment of Glomerular Pathological Findings in Lupus Nephritis
Next Article in Special Issue
The Current Role of the Heavy/Light Chain Assay in the Diagnosis, Prognosis and Monitoring of Multiple Myeloma: An Evidence-Based Approach
Previous Article in Journal
Tracking SARS-CoV-2: Novel Trends and Diagnostic Strategies
Previous Article in Special Issue
Harmonization of Flow Cytometric Minimal Residual Disease Assessment in Multiple Myeloma in Centers of Polish Myeloma Consortium
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

From Immune Dysregulations to Therapeutic Perspectives in Myelodysplastic Syndromes: A Review

by
Thibault Comont
1,2,3,*,
Emmanuel Treiner
3,4,5 and
François Vergez
2,3,6
1
Department of Internal Medicine, IUCT-Oncopole, Toulouse University Hospital (CHU-Toulouse), 31300 Toulouse, France
2
Cancer Research Center of Toulouse, Unité Mixte de Recherche (UMR) 1037 INSERM, ERL5294 Centre National de La Recherche Scientifique, 31100 Toulouse, France
3
School of Medicine, Université Toulouse III—Paul Sabatier, 31062 Toulouse, France
4
Laboratory of Immunology, Toulouse University Hospital (CHU-Toulouse), 31300 Toulouse, France
5
Infinity, Inserm UMR1291, 31000 Toulouse, France
6
Laboratory of Hematology, IUCT-Oncopole, Toulouse University Hospital (CHU-Toulouse), 31300 Toulouse, France
*
Author to whom correspondence should be addressed.
Diagnostics 2021, 11(11), 1982; https://doi.org/10.3390/diagnostics11111982
Submission received: 30 September 2021 / Revised: 21 October 2021 / Accepted: 22 October 2021 / Published: 26 October 2021
(This article belongs to the Special Issue Hematological Neoplasms)

Abstract

:
The pathophysiology of myelodysplastic syndromes (MDSs) is complex and often includes immune dysregulation of both the innate and adaptive immune systems. Whereas clonal selection mainly involves smoldering inflammation, a cellular immunity dysfunction leads to increased apoptosis and blast proliferation. Addressing immune dysregulations in MDS is a recent concept that has allowed the identification of new therapeutic targets. Several approaches targeting the different actors of the immune system have therefore been developed. However, the results are very heterogeneous, indicating the need to improve our understanding of the disease and interactions between chronic inflammation, adaptive dysfunction, and somatic mutations. This review highlights current knowledge of the role of immune dysregulation in MDS pathophysiology and the field of new drugs.

1. Introduction

Myelodysplastic syndromes (MDSs) are acquired clonal myeloid malignancies characterized by ineffective hematopoiesis resulting in peripheral cytopenia and a risk of progression to acute myeloid leukemia (AML) [1]. MDSs are a heterogeneous group of diseases with differences in clinical presentation, biological features, prognosis, and treatment. Severity is defined by the Revised International Prognostic Scoring System (IPSS-R) [2] but some studies identified other prognostic factors such as patient characteristics (comorbidities) [3] or somatic mutations [4] and new scores are actually being developed [5].
In Low-risk MDS (LR-MDS), the objectives of treatments are to correct peripheral cytopenia. In contrast, blastic progression and risk of AML transformation are the major concerns of High-Risk MDS (HR-MDS) patients, and treatments target the malignant clone [6]. In both situations, however, available therapeutic options are limited, and a better comprehension of MDS pathophysiology is needed to identify new targets [7].
The central role of the immune system in tumor surveillance is well known and targeted in solid tumors [8] but its implication in myeloid malignancies pathogenesis, especially in MDS, is less well described. However, recent studies tried to explore dysregulations in both innate and adaptive compartments of immunity and their consequences on MDS presentation, prognosis, and risk of AML transformation. Moreover, T-cells from patients with MDS may originate from the malignant clone [9]. Together, these studies showed that MDS is associated with several immune alterations leading to cytopenia and/or disease progression; further, this immune dysregulation is a dynamic process, evolving during the disease [10].
Thus, an understanding of these immunological disorders may allow for improved initial classification of patients but also for innovative and more targeted treatments.
In this review, we will describe the current knowledge on the immune dysregulations involved in the early or late steps of MDS pathophysiology, their clinical implications, prognosis, and their present and potential therapeutic actions.

2. Inflamm-Aging as a Risk for Clonal Hematopoiesis

MDS diagnoses increase with advanced age [11]. Indeed, whereas prevalence is about 4/100,000 in the US population, it increases after 60 years to 35/100,000, suggesting the role of aging in disease initiation. Aging is now a well-understood process with several alterations in different systems including immune and hematopoietic compartments. Consequences include increased risk of infections, auto-immunity, and/or malignancies. Several molecular mechanisms are involved in aging, including genomic damage, telomere curtailment, or epigenetic alterations [12]. Furthermore, aging is associated with an increase in the number of somatic mutations, which, in the hematopoietic compartment, create a broad array of genetically distinct stem cells (named clones).
Clonal hematopoiesis is characterized by the over-representation of blood cells derived from a single clone. Several studies have identified recurrent somatic mutations in several genes driving clonal hematopoiesis. These loss-of-function mutations mostly concern genes involved in DNA methylation (DNA methyl-transferase 3A, DNMT3A and Ten-eleven-translocation 2, TET2), chromatin regulation (ASXL1), or splicing factors (Splicing Factor 3b Subunit 1, SF3B1, Serine and Arginine Rich Splicing Factor 2, SRSF2, and U2 Small Nuclear RNA Auxiliary Factor 1, U2AF1) [13,14,15,16,17,18,19,20,21]. The term “clonal hematopoiesis of indeterminate potential” (CHIP) was then introduced to distinguish non-malignant clonal hematopoiesis, which is clearly linked to cancer-associated mutations from other forms of clonal hematopoiesis. CHIP is defined in a subset of individuals with clonal hematopoiesis based on a variant allele frequency ≥ 2% of a somatic mutation in a hematologic malignancy-associated gene. Individuals with CHIP have a modestly increased long-term risk of developing hematologic malignancies. Somatic clones increase in frequency with each decade of life (from 1% of healthy individuals under the age of 40 to 10–20% after 70 years old). CHIP affects 20–40% of individuals > 80 years, and is associated with an increased risk for transformation to MDS or AML [20,22,23,24,25].
Whereas the molecular defects driving CHIP are well studied, how these alterations occur is less well understood. Environmental stimuli and stressors, including chronic infections and inflammatory processes, may play a key role in CHIP [26]. Indeed, aging is also characterized by immune senescence with quantitative and qualitative modifications in several cell types. A major characteristic of immune-aging is chronic inflammation (by persistent infection or a sterile inflammatory process) [27]. This phenotype notably includes elevated levels of pro-inflammatory cytokines (TNF-α, interleukin(IL)-1β, IL-6, RANTES). Some studies explored the consequences of chronic inflammation in hematopoietic stem cells (HSCs) in the context of aging and showed a depletion of HSCs [28,29] or functional alterations [30,31]. Moreover, chronic inflammation favors the expansion of HSCs with CHIP mutations [32]. Inflammation can initiate the CHIP process but also its progression. Indeed, in vivo studies showed that CHIP-associated mutations, such as TET2 or DNMT3A, could confer a pro-inflammatory profile, with high levels of TNF-α, IL-1, IL-6, IL-8, or IFN-γ that could drive the expansion of clonal HSCs [32,33].
Taken together, chronic inflammation could participate in CHIP initiation, and mutant clones promote this pro-inflammatory profile, driving clonal expansion.

3. Smoldering Inflammation Acts Early in MDS Pathophysiology

We previously described the role of inflammation in clonal selection and progression, but dysregulations of innate immune system components are also involved in MDS pathophysiology.

3.1. Proinflammatory Cytokines

Several studies observed high levels of proinflammatory cytokines in MDS patients [34], including TNF-α [35,36], IFN-γ [37], TGF-β, IL-6 [38], or IL-8 [39] with a potential independent risk factor of progression [40]. Moreover the levels of TNF-α, IFN-γ, and IL-6 are associated with apoptosis induction in bone marrow (BM) [41,42]. This can explain why this cytokine profile is more frequently observed in LR-MDS, while immunosuppressive cytokines such as IL-10 are more prevalent in HR-MDS [43].

3.2. NLRP3 Inflammasome

Inflammasomes play a key role in MDS pathophysiology, particularly in LR-MDS [44,45,46]. In an activated NLR family, NOD-like receptor pyrin domain-containing 3 (NLRP3) complexes are implicated in the pyroptosis of MDS cells, which is an inflammatory cell death process [47]. The NLRP3–pyroptosis axis is found to be activated in MDS BM cells [48] and is responsible for LR-MDs features such as macrocytosis and ineffective hematopoiesis resulting in cytopenia. Activation of the NLRP3 inflammasome is initiated by damage-associated molecular pattern signals (DAMP) (e.g., S100A8/9 [49]), and involves signal transducers such as serine/threonine kinases IRAK1/4 [50] and the E3 ubiquitin ligase TRAF6. Once activated, NLRP3 recruits and activates pro-caspase-1, which then activates pro-IL-1β, pro-IL-18, and the pore-forming protein gasdermin D [51], followed by pyroptotic cell death with the release of active IL-1β, IL-18, and other intracellular proteins that contribute to local inflammation [48,52]. Interestingly, S100A8 and S100A9 proteins are increased in the HSCs or blood of MDS patients, especially in LR-MDS [53]. These proteins also expand Myeloid-Derived Suppressor Cells (MDSCs) in the BM of patients with MDS [54].

3.3. Toll-like-Receptors (TLR)

The TLR family contains 10 subtypes in humans, and these are expressed on a variety of hematopoietic cell types [55]. TLR signaling regulates hematopoietic stem and progenitor cell function [56]. MDS HSCs are particularly sensitive to pyroptosis because of high levels of TLR (TLR 1-2-6) expression and activation [42,57,58,59]. Moreover, while the overexpression of TLR-2 or TLR-4 is observed in LR-MDS, increased expression of TLR-6 correlates with HR disease [60]. HSCs of LR-MDS patients are more sensitive to TLR ligands (DAMP) and some of these ligands are more abundant in the BM and/or serum of MDS patients compared to healthy controls (ex HMGB1 [61], S100A8/9 [53]). In LR-MDS, S100A8/A9 is more abundant (also secreted by MDSCs) and binds to TLR-4 and CD33, stimulating inflammasome assembly and pyroptotic cell death by promoting the production of inflammasome components, Reactive Oxygen Species (ROS), and proinflammatory cytokines such as IL-1β and IL-18, and the expansion of MDSCs. The consequence is the suppression of normal hematopoiesis via the production of cytokines such as IL-10 and TGF-β.

3.4. Immune Cells

MDSCs [62] are increased in the BM and blood of MDS patients compared to healthy donors, especially in HR-MDS, and are involved in ineffective hematopoiesis. MDSCs also induce T-cell immunosuppression via the secretion of IL-10 and TGF-β, leading to MDS progression. MDSC expansion is driven by the interaction of S100A9 with CD33.
Monocytes: The number of monocytes in the blood or BM of MDS patients is increased, especially in HR-MDS, but their ability to differentiate into macrophages to then use their phagocytic function is decreased [63,64]. Cytokines, chemokines, and TLR genes are also downregulated in BM monocytes from MDS patients. Moreover, as the disease progresses, the number of abnormal monocytes increases in the BM of the patients [65]. In addition, the ratio of M2 macrophages to monocytes is higher in patients with MDS whereas the ratio of M1 to M2 macrophages is lower in MDS patients. Because monocytes play a key role in the defense against microbial agents, these alterations may explain the susceptibility to infections observed in MDS patients.
Dendritic cells (DCs): Some studies described quantitative or qualitative defects of DCs in MDS patients [66] with, for example, an altered cytokine profile [67]. High secretion of IL-10 may contribute to the immunosuppressive phenotype. Interestingly, it was shown that cytogenetic abnormalities in DCs from MDS patients were similar to the malignant clones of MDS, suggesting that they originated from the malignant clones [68]. DCs derived from MDS monocytes also have morphologic and phenotypic abnormalities [64].
Lymphocytes: A reduced number of lymphocytes, or their dysfunction, can also have prognostic implications. For example, the number of Tregs could be a prognostic factor in LR-MDS and predict the severity of anemia, AML transformation, and overall survival [69]. A low number of NK-cells, their reduced cytotoxic capacity, or their Killer-cell immunoglobulin-like receptors (KIR) gene haplotype are also associated with poor prognosis and AML transformation [70,71,72].

4. Positive and Negative Immune Regulators

Immune cell signaling includes co-stimulatory and co-inhibitory (named Immune check points or ICPs) molecules that are differentially expressed by a variety of cell types (such as antigen-presenting cells, B-cells, macrophages, and T-cells) and may be regulated by activation [73]. Some of these molecules were studied in MDS and appeared to be potential therapeutic targets.

4.1. Co-Stimulatory Molecules

-
OX40, Inducible T-cell COStimulator (ICOS) and 4-1BB+: Tumor protein 3 (TP53) MDS patients display reduced numbers of OX40+ cytotoxic T-cells and helper T-cells, as well as reduced ICOS+ and 4-1BB+ NK-cells [74].
-
Other positive regulators, such as CD244, CD80, and CD40, have been studied but their implication is not clearly validated and they are not yet interesting therapeutic leads [75,76].

4.2. Coinhibitory Molecules

Cytotoxic T-Lymphocyte Antigen 4 (CTLA-4): Upregulation of CTLA-4 has been observed in CD34+ cells from HR-MDS, and levels further increased in the post hypomethylating agents (HMA) failure setting [77].
Program cell death-1, and L1 (PD-1 and PD-L1): The PD-1/PD-L1 axis is well studied. Upregulation of PD-1 and PDL-1 was seen in CD34+ BM cells from patients with MDS and in T-cells, with further upregulation seen in those previously treated with HMAs [77,78]. Moreover, a higher rate of PD-L1 expression on BM blasts and HSCs from TP53-mutated AML and MDS patients is observed [74]. The expression of ICPs could be upregulated in the presence of pro-inflammatory cytokines [79] or in response to S100A9 signaling, contributing to MDSC-induced HSC death [80].
T-cell immunoglobulin and mucin-containing protein-3 (TIM3): Some studies showed that TIM3 is overexpressed in HSCs, blasts, and CD8-T-cells from MDS patients [81,82]. Excessive TIM3+ HSCs are closely related to disease severity. TIM3+ HSCs and T-cells displayed aberrant functions [83]. Moreover, MDSCs suppress CD8+ T cells through the TIM3/Galectin 9 pathway [84].
T cell immunoreceptor with Ig and ITIM domains (TIGIT): TIGIT was found to be highly expressed in NK-cells and T-cells from the blood of MDS patients with an association with disease progression [85]. The overexpression of TIGIT can be associated with decreased NK and T-cell functions, and lower secretions of activating cytokines such as IFN-γ and TNF-α.
ICPs expression is also associated with resistance to hypomethylating agents [77].
Moreover, the impact of ICP expression on MDS prognosis was assessed in a recent study, which evaluated the mutation burden in genes coding for ICP molecules (LAG-3, CTLA-4, B7H3, PD-1, PD-L1, etc.). The authors observed a high prevalence of mutations in these genes, with an impact on overall survival [86].

5. Opposite Adaptive Dysfunctions in LR and HR-MDS

In the past few years, several studies have explored the role of adaptive immunity in MDS pathogenesis and have highlighted differences in immune profiles between LR and HR-MDS. While LR-MDS presents with a more inflammatory and cytotoxic profile, HR-MDS is characterized by a more suppressive microenvironment [10]. For example, whereas IL-17-producing CD4+ T-cells from LR-MDS patients are increased and Tregs number and function are reduced [87], HR-MDS patients display quantitative and qualitative alterations of CD8+ T-cells [88], NK-cells [70], and an expansion of Tregs [89,90] and MDSCs. Moreover, HR-MDS patients overexpress ICPs on their blasts (such as PD-L1 or TIM-3) [83,91,92], their HSCs (PD-L1, TIM-3, or CD47) [77,78,81,93], their T and NK-cells (PD-1, TIGIT, or TIM-3) [78,82,84,85], or their macrophages [94].

6. Immune Phenotypes Can Be Associated with Somatic Mutations

Clonal hematopoiesis and MDS progression are driven by the acquisition of somatic mutations. However, these mutations not only act on clonal selection but also on the immune microenvironment. Then, mutations affecting DNA methylation (TET-2 and DNMT3A), histone modification (ASXL1), or splicing factors (SF3B1, SRSF2, U2AF1) have been associated with increased inflammation in patients with CHIP or MDS.
TET2 mutation seems to promote clonal HSC dominance by creating an inflammatory environment [32,95]. Interestingly, TET2 mutant CHIP is more highly associated with elevated IL-6, while DNMT3A mutant CHIP is more highly associated with elevated TNF-α or IFN-γ [96,97]. Recently Zhao et al. found that TET2 or IDH 1/2 mutations were more frequent in MDS patients presenting with systemic autoinflammatory or autoimmune diseases [98]. In this study, TET2/IDH mutant patients’ phenotyping showed a reduction of Tregs and deep alterations in CD8+ T-cell distribution.
Spliceosome mutations (such SF3B1, U2AF1, and SRSF2) have also been associated with immune dysregulation such as TRL activity [99,100] or proinflammatory cytokine production [101].
As previously described, Sallman et al. recently described a distinct immune phenotype associated with TP53 mutated MDS/AML patients [74]. These patients overexpressed ICPs PD-L1 (especially in HSCs) and CTLA-4, associated with poor survival. Moreover, TP53 mutant patients displayed reduced numbers of BM OX40+ cytotoxic T-cells and helper T-cells, reduced ICOS+ and 4-1BB+ NK cells, and an increased number of immunosuppressive regulatory T-cells and MDSCs. Finally, they found that a higher proportion of BM infiltrating ICOShigh/PD-1neg Tregs was a highly significant predictor of overall survival.
Somatic mutations not involved in clonal hematopoiesis can also drive inflammatory manifestations and MDS. Recently, an autoinflammatory disease characterized by a somatic mutation of UBA1 has been described and named VEXAS (Vacuoles, E1 Enzyme, X-linked, Autoinflammatory, Somatic syndrome) [102]. Systemic manifestations often include neutrophilic skin lesions, polychondritis, pulmonary infiltrates, or thrombosis [103]. VEXAS can be associated with hematological disorders, especially MDS in 25–55% of cases [102,104,105,106,107]. Implication of ubiquitin-like modifier activating enzyme 1 (UBA1) mutation in MDS pathophysiology is not well studied but could be explained by chronic inflammation.
Finally, driver mutations have been found in lymphoid precursors, raising the question of the potential impact of these mutations on immune responses in the MDS context [108].

7. Immune Strategies in MDS: Past, Present and Future

Targeting the immune system in MDS is not a novel concept, but modern approaches such as ICP inhibitors (ICPi) or an adoptive T-cell transfer may be promising and are still under evaluation (Table 1).

7.1. “Old” Treatments

Immunosuppressive and immunomodulatory therapies have been used in selected MDS patients.
First, allogenic Hematopoietic stem-cell transplantation (HSCT) is actually the only curative treatment for MDS. It is considered an immune approach because of the graft versus leukemia effect but also because of its immune modulatory properties, especially in the context of reduced-intensity conditioning transplant programs [109].
The first and most commonly used immunosuppressive treatments were antithymocyte globulin and ciclosporin. Studies have reported hematologic improvement in 20–60% of selected MDS patients (young age, hypoplastic LR-MDS, normal karyotype), especially when used in combination [110,111,112]. These responses seem to be associated with lower rates of AML transformation [111,113,114,115].
In del(5q) LR-MDS, lenalidomide, an immunomodulatory drug, has shown a high rate of efficacy with sustained responses [116]. It acts on T-cell activity via several mechanisms, including reduced T-cell tolerance and increased effector functions [117]. Other studies have shown its role in the ubiquitination and degradation of specific substrates [118]. Lenalidomide is currently used in LR-MDS with del(5) patients who are still transfusion-dependent despite treatment with erythropoietic-stimulating agents [119].
Alemtuzumab is a humanized monoclonal antibody directed against CD52, a glycosyl phosphatidyl inositol-anchored cell surface marker abundantly expressed on B and T-cells and at low levels on effector cells of the innate immune system. It was evaluated in a subset of MDS patients and showed good efficacy and a good safety profile [120].
Targeting pro-inflammatory cytokines is also an option in MDS, especially in LR-MDS patients. Infliximab, an antibody targeting TNF-α, was evaluated in LR-MDS and showed a moderate hematologic response but a good safety profile [121]. Other agents such as Etanercept, were associated with HMA and showed interesting results [122]. Studies using the anti-TNF-α antibody in combination with other drugs are ongoing. Other biologics are actually used in MDS-associated systemic inflammatory or auto-immune disorders (such as anti-IL-1, IL-6 antibodies) but are also evaluated in combination with other drugs [123].

7.2. Targeting TGF-β Superfamily in LR-MDS with Anemia

Increased concentrations of TGF-β superfamily ligands, including growth differentiation factor 11 (GDF11) in bone marrow have been linked to ineffective erythropoiesis in MDS [124,125]. Luspatercept (ACE-536) is a soluble fusion protein that binds to GDF11 and other TGF-β superfamily ligands. It acts as an activin receptor type II ligand trap and allows the restoration of terminal maturation of erythroid progenitors by diminishing Smad2/3 signaling. It was recently evaluated in a phase III, randomized, double blind, placebo-controlled study for LR-MDS patients with ring sideroblasts who were refractory/intolerant or ineligible for ESA, and transfusion dependent. Results are encouraging, and luspartecept obtained FDA and EMA approbation in this indication [126,127].

7.3. Targeting the Inflammasomes

As stated before, inflammasomes play a key role in dysregulated innate immune signaling in MDS, and targeting key hub mediators could be a promising option. New molecules such as CA-4948 (IRAK4 inhibitor), ibrutinib (BTK inhibitor acting as NRLP3 inhibitor), bortezomib (NFKb inhibitor), Cx-01 (TLR-4 inhibitor), or OPN-305 (TLR-2 inhibitor) are actually being evaluated in early-stage clinical studies for MDS and AML [50,128,129,130,131,132]. Other targets, including S100A9, CD33, and IL-1 Receptor Accessory Protein will be studied.

7.4. Checkpoint Inhibitors

Following the successes with ICPi in solid tumors, these therapies are being evaluated in hematologic malignancies, including AML and MDS [133]. The first ICPi molecules challenged in clinical trials for HR-MDS patients are ipilimumab [134,135,136] (CTLA-4 inhibitor), nivolumab [137,138] or pembrolizumab [139,140] (PD-1 inhibitors), and atezolizumab or durvalumab [141,142,143] (PD-L1 inhibitors). Targeting TIGIT, CD70, or TIM-3 also seems to be an interesting option [84,85,144,145,146]. Response rates in monotherapy approaches are actually very low and combined strategies are more promising, especially with HMA. Indeed, treatment with HMA has been shown to upregulate several of these ICPs (such as PD-L1, TIM-3, and CD47) on MDS cells [77]. Moreover, the association of two ICPis (ipilimumab and nivolumab) has been also evaluated, finding the same efficacy as the results for monotherapy [135]. More recently, new ICPs have been identified as CD47, especially expressed by macrophages and acting as a “do not eat me” signal [147,148,149]. MDS cells from HR-MDS patients overexpress CD47 and this is associated with poor survival. Thus, the CD47–SIRPa interaction could be targeted by specific inhibitors [150]. Two anti-CD47 inhibitors have been evaluated in HR-MDS patients. First, CC-90002 was tested in phase 1, but was associated with a major toxicity profile and poor preliminary efficacy [151]. Another CD47 inhibitor, Magrolimab (HuF9-G4 or 5F9), seems to be less toxic than CC-90002 in a phase I trial [152]. Investigators reported high efficacy of magrolimab in association with HMA and a phase III evaluation of this association is currently in the recruitment stage (NCT04313881) [152].
Together, ICPis showed moderate and variable efficacy in early-phase trials with HR-MDS patients. The association with HMA seems to be more interesting than monotherapy, especially in the front line. Responders must be characterized more to identify the predictive factors of response with each ICPi.

7.5. Targeting NK-Cells

Similar to T-cells, NK-cells can also be exhausted in HR-MDS with the loss of their cytotoxic capacity [85,153,154]. Restoring their functional activity could help to avoid MDS progression. To address this hypothesis, new molecules are actually being evaluated, such as GTB-3550 TriKE™, a novel CD16/IL-15/CD33 Tri-Specific Killer Engager (TriKE) [155,156]. Pre-clinical data show specific NK cell activation and targeted cytolytic activity in xenogeneic AML mouse models. Another way to improve NK cell activity consists of blocking interactions between KIRs and human leukocyte antigen-C (HLA-C) molecules. Lirilumab is a human IgG4 monoclonal antibody that blocks KIR/HLA-C interaction [157].

7.6. Vaccination Strategies

Several tumor-associated antigens (TAA) have been identified and found to be overexpressed in MDS patients, leading to the development of specific vaccines to stimulate tumor-specific T cells [158,159,160]. TAAs targeted by peptide vaccines include Wilm’s tumor-1 (WT1), Proteinase-3+ neutrophil elastase (PR-1), or NY-ESO-1. The expression of WT1 is correlated with the marrow blast percentage, poor response to HMA, and poor OS [159,161,162,163,164]. The first studies of the peptide vaccine of WT1 have demonstrated an acceptable safety profile, good biological response (expansion of WT1 reactive T cells), but with no significant sustained clinical response [159,165,166,167,168]. PR-3 is another potential target, and a PR-1 vaccine (an HLA-A2-restricted peptide-based PR3 in combination with neutrophil elastase) was recently evaluated with the same profile of response as the WT1 vaccine [160]. NY-ESO1, a cancer testis antigen, has been found expressed on some solid tumors [169]. NY-ESO1 expression is low in MDS due to the silencing of TAA genes by the methylation mechanism [170]. As expected, early studies showed an increase in NY-ESO1 expression after treatment with HMA [171] and the results in MDS patients are encouraging [172]. New multiepitope vaccines are also being evaluated in combination with HMA, but the first results are disappointing (negative results and AML progression) [173].
To conclude, vaccine strategies in MDS have actually shown a good safety profile and interesting biological results but with poor clinical responses. Because dendritic cells play a key role in vaccine response and because these cells present some dysfunction in MDS patients [64,67,174], it could be important to consider this issue. Other vaccine strategies are also evaluated [175].

7.7. Adoptive T-Cell Transfer Therapy

Chimeric antigen receptor: MDS cells have been shown to overexpress the ligands for Natural killer group 2 (NKG2D) receptors [176,177], and clinical trials with NKG2D receptors CAR-T cells have been evaluated, but without favorable outcomes [178]. The expansion process could be improved by lymphodepleting chemotherapy prior to infusion, but a major issue seems to be the inability of CAR-T to persist as memory lymphocytes. Other studies evaluating new CAR-T cells are ongoing [179,180]. Because CD123 delineates MDS stem cells in HR-MDS patients, CD123 CARs have been generated and have demonstrated promising results in vitro and in vivo [181].
Donor-directed lymphocytes: While HSCT is the only curative treatment for MDS, relapses can occur and are associated with poor outcomes. In these cases, unmanipulated DLIs may be an option but these are associated with severe GVHD. DLI could be stimulated ex vivo by TAA, which selects for an enriched, polyclonal CD4+ and CD8+ specifically directed against MDS. TTA-DLI appears to be safer, with respect to GVHD, than unselected DLI [182,183].
Table 1. Current and potential immune targets in MDS.
Table 1. Current and potential immune targets in MDS.
MoleculePatientsNCT/Phase/StatusRef.
Cytokines
TGF-βLuspaterceptLR-MDS with anemiaNCT02604433/Phase III/Authorization[126]
TNF-αEtanerceptHR-MDS (+Aza)NCT00118287/I-II/Completed[122]
IL-1CanakinumabLR-MDS (+Aza)
LR-MDS (+luspa, +TIM3inh)
NCT04239157/II/Recruiting
NCT04810611/Ib/Recruiting
Inflammasome
IRAK-4CA-4948HR-MDS (+aza and/or Ven)NCT04278768/I-II/Recruiting[128]
NLRP3ibrutinibHR-MDS (+Aza)
HR-MDS (+Len)
NCT02553941/I/Recruiting
NCT03359460/I/Recruiting
[129]
TLR
TLR-4CX-01HR-MDS (+Aza)NCT02995655/I/Completed[131]
TLR-2OPN-305 LR-MDSNCT02363491/I-II/Completed[132]
NF-kB BortezomibHR-MDS (+Len)NCT00580242/I/completed[130]
MDSC
CD33 inhBI 836858LR-MDSNCT02240706/II/Terminated
Immune check-points
CTLA-4ipilimumabHR-MDS
HR-MDS (and/or nivolumab +/−Aza)
HR-MDS (+Dec)
NCT01757639/I/Completed
NCT02530463/II/recruiting
NCT02890329/I/Recruiting
[134,135]
PD-1Nivolumab

Prembrolizumab

PDR001
HR-MDS (and/or ipi +/−Aza)
HR-MDS (+chemo)
HR-MDS (+Aza)
HR-MDS(+Dec)
HR-MDS (+Aza+/Tim3 inh)
NCT02530463/II/recruiting
NCT02464657/II/completed
NCT03094637/II/recruiting
NCT03969446/I/recruiting
NCT03066648/I/active
[135,137,139]
PD-L1Durvalumab
Atezolizumab
HR-MDS (+Aza)
HR-MDS (+Guadecitabine)
HR-MDS (+/−Aza)
NCT02775903/II/active
NCT02935361/I-II/active
NCT02508870/I/completed
[141,143,179]
TIM3Sabatolimab (MBG453)HR-MDS (+Aza+/−PD-1 inh)
HR-MDS (+Aza)
HR-MDS (+Aza+Ven)
NCT03066648/I/active
NCT04266301/III/Recruiting
NCT04812548/II/recruiting
[145,146]
CD70Cusatuzumab (ARGX-110)HR-MDS (+Aza)
HR-MDS (+Aza)
NCT04241549/I/Active
NCT03030612/I-II/Active
[146]
CD47AK117
Hu5F9-G4
CC-90002
ALX148
Magrolimab
HR-MDS (+Aza)
HR-MDS
HR-MDS
HR-MDS (+Aza)
HR-MDS (+/−Aza)
HR-MDS (+Aza)
NCT04900350/I-II/Recruiting
NCT02678338/I/completed
NCT02641002/I/Terminated
NCT04417517/I-II/recruiting
NCT03248479/I/completed
NCT04313881/III/recruiting
[152,153]
NK-cells
CD16/IL-15/CD33GTB-3550 TriKE™HR-MDSNCT03214666/I-II/recruiting[156]
KIR inhLirilumabHR-MDS (+/−Aza)NCT02599649/II/terminated[157]
CAR-T-cells
Cyad-O1/02Cyad-O1/02HR-MDSNCT03466320/I-II/completed, NCT04167696/I/recruiting[178]
NKX101NKX101HR-MDSNCT04623944/I/Recruiting[179]
Prgn-3006 Prgn-3006HR-MDSNCT03927261/I/recruiting[180]
Vaccine
DSP-7888LR and HR-MDSNCT02436252/I-II/completed[158]
K562/GM-CSF/CD40LHR-MDNCT00840931/I/Completed[175]
NPMWHR-MDSNCT02750995/I/completed[173]
Aza: Azacitidine; CD: Cluster of Differentiation; CTLA-4: Cytotoxic T-Lymphocyte Antigen 4; HR-MDS: High-risk myelodysplastic syndrome; IL: Interleukin; inh: Inhibitor; Len: Lenalidomide; luspa: Luspatercept; LR-MDS: Low-risk myelodysplastic syndrome; MDSC: Myeloid-Derived Suppressor Cells; NKG2D: Natural killer group 2 member; NLRP3: NOD-like receptor family, pyrin domain containing 3; PD-1: Program cell death-1; PD-L1: Program cell death-Ligand; TIM3: T-cell immunoglobulin and mucin containing protein-3; TGF-β: Transforming Growth Factor-β; TLR: Toll Like Receptor; TNF-α: Tumor Necrosis Factor-α; Ven: Venetoclax.

8. Conclusions

Recent interest for alterations in the immune system in MDS has led to an improved understanding of its pathogenesis (Figure 1). Both the innate and adaptive immune systems have been shown to participate in MDS pathogenesis and progression, which has allowed the development of different therapeutic approaches in LR-MDS and HR-MDS. However, results are heterogeneous and future investigations should further explore the MDS immune landscape. In parallel, future studies will have to determine which patients will respond to a predefined treatment, in order to achieve a personalized approach.

Author Contributions

Conceptualization, T.C. and F.V.; writing—original draft preparation, T.C. and E.T. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

Thibault Comont received honoraria and/or research or educational support from AbbVie, AstraZeneca, Bristol Myers Squibb (Celgene), Novartis, and Takeda. All other authors have no conflict of interest to declare.

References

  1. Cazzola, M. Myelodysplastic Syndromes. N. Engl. J. Med. 2020, 383, 1358–1374. [Google Scholar] [CrossRef] [PubMed]
  2. Greenberg, P.L.; Tuechler, H.; Schanz, J.; Sanz, G.; Garcia-Manero, G.; Solé, F.; Bennett, J.M.; Bowen, D.; Fenaux, P.; Dreyfus, F.; et al. Revised International Prognostic Scoring System for Myelodysplastic Syndromes. Blood 2012, 120, 2454–2465. [Google Scholar] [CrossRef] [PubMed]
  3. Thomopoulos, T.P.; Pappa, V.; Papageorgiou, S.G. Comorbidities and frailty predict outcome of patients with myelodysplastic syndromes. Should we integrate them in novel prognostic scoring systems? J. Geriatr. Oncol. 2021, 12, 1122–1129. [Google Scholar] [CrossRef]
  4. Haferlach, T. The Molecular Pathology of Myelodysplastic Syndrome. Pathobiology 2018, 86, 24–29. [Google Scholar] [CrossRef] [Green Version]
  5. Shreve, J.; Nazha, A. The Evolving Landscape of Myelodysplastic Syndrome Prognostication. Clin. Hematol. Int. 2020, 2, 43–48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Garcia-Manero, G.; Chien, K.S.; Montalban-Bravo, G. Myelodysplastic syndromes: 2021 update on diagnosis, risk stratification and management. Am. J. Hematol. 2020, 95, 1399–1420. [Google Scholar] [CrossRef]
  7. Fozza, C. Retuning the immune system in myelodysplastic syndromes: From immunomodulatory approaches to vaccination strategies and non myeloablative hemopoietic cell transplant. Crit. Rev. Oncol. 2019, 133, 112–119. [Google Scholar] [CrossRef]
  8. Sharma, P.; Wagner, K.; Wolchok, J.D.; Allison, J.P. Novel cancer immunotherapy agents with survival benefit: Recent successes and next steps. Nat. Rev. Cancer 2011, 11, 805–812. [Google Scholar] [CrossRef] [PubMed]
  9. Vercauteren, S.M.; Starczynowski, D.T.; Sung, S.; McNeil, K.; Salski, C.; Jensen, C.-L.; Bruyere, H.; Lam, W.L.; Karsan, A. T cells of patients with myelodysplastic syndrome are frequently derived from the malignant clone. Br. J. Haematol. 2011, 156, 409–412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Wang, C.; Yang, Y.; Gao, S.; Chen, J.; Yu, J.; Zhang, H.; Li, M.; Zhan, X.; Li, W. Immune dysregulation in myelodysplastic syndrome: Clinical features, pathogenesis and therapeutic strategies. Crit. Rev. Oncol. 2018, 122, 123–132. [Google Scholar] [CrossRef] [PubMed]
  11. Rollison, D.E.; Howlader, N.; Smith, M.T.; Strom, S.S.; Merritt, W.D.; Ries, L.A.; Edwards, B.K.; List, A. Epidemiology of myelodysplastic syndromes and chronic myeloproliferative disorders in the United States, 2001–2004, using data from the NAACCR and SEER programs. Blood 2008, 112, 45–52. [Google Scholar] [CrossRef] [PubMed]
  12. López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The Hallmarks of Aging. Cell 2013, 153, 1194–1217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Haferlach, T.; Nagata, Y.; Grossmann, V.; Okuno, Y.; Bacher, U.; Nagae, G.; Schnittger, S.; Sanada, M.; Kon, A.; Alpermann, T.; et al. Landscape of genetic lesions in 944 patients with myelodysplastic syndromes. Leukemia 2013, 28, 241–247. [Google Scholar] [CrossRef] [Green Version]
  14. Papaemmanuil, E.; Gerstung, M.; Malcovati, L.; Tauro, S.; Gundem, G.; Van Loo, P.; Yoon, C.J.; Ellis, P.; Wedge, D.; Pellagatti, A.; et al. Clinical and biological implications of driver mutations in myelodysplastic syndromes. Blood 2013, 122, 3616–3627. [Google Scholar] [CrossRef]
  15. Abdel-Wahab, O.; E Figueroa, M. Interpreting new molecular genetics in myelodysplastic syndromes. Hematology 2012, 2012, 56–64. [Google Scholar] [CrossRef] [Green Version]
  16. 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]
  17. Yoshida, K.; Sanada, M.; Shiraishi, Y.; Nowak, D.; Nagata, Y.; Yamamoto, R.; Sato, Y.; Sato-Otsubo, A.; Kon, A.; Nagasaki, M.; et al. Frequent pathway mutations of splicing machinery in myelodysplasia. Nature 2011, 478, 64–69. [Google Scholar] [CrossRef]
  18. Kon, A.; Shih, L.-Y.; Minamino, M.; Sanada, M.; Shiraishi, Y.; Nagata, Y.; Yoshida, K.; Okuno, Y.; Bando, M.; Nakato, R.; et al. Recurrent mutations in multiple components of the cohesin complex in myeloid neoplasms. Nat. Genet. 2013, 45, 1232–1237. [Google Scholar] [CrossRef] [PubMed]
  19. Gao, T.; Ptashkin, R.; Bolton, K.L.; Sirenko, M.; Fong, C.; Spitzer, B.; Menghrajani, K.; Ossa, J.E.A.; Zhou, Y.; Bernard, E.; et al. Interplay between chromosomal alterations and gene mutations shapes the evolutionary trajectory of clonal hematopoiesis. Nat. Commun. 2021, 12, 338. [Google Scholar] [CrossRef]
  20. Jaiswal, S.; Fontanillas, P.; Flannick, J.; Manning, A.; Grauman, P.V.; Mar, B.; Lindsley, C.; Mermel, C.; Burtt, N.; Chavez, A.; et al. Age-Related Clonal Hematopoiesis Associated with Adverse Outcomes. N. Engl. J. Med. 2014, 371, 2488–2498. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Kwok, B.; Hall, J.M.; Witte, J.S.; Xu, Y.; Reddy, P.; Lin, K.; Flamholz, R.; Dabbas, B.; Yung, A.; Al-Hafidh, J.; et al. MDS-associated somatic mutations and clonal hematopoiesis are common in idiopathic cytopenias of undetermined significance. Blood 2015, 126, 2355–2361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Heuser, M.; Thol, F.; Ganser, A. Clonal Hematopoiesis of Indeterminate Potential. Dtsch. Aerzteblatt Online 2016, 113, 317–322. [Google Scholar] [CrossRef] [Green Version]
  23. Steensma, D.P.; Bejar, R.; Jaiswal, S.; Lindsley, R.C.; Sekeres, M.; Hasserjian, R.P.; Ebert, B.L. Clonal hematopoiesis of indeterminate potential and its distinction from myelodysplastic syndromes. Blood 2015, 126, 9–16. [Google Scholar] [CrossRef] [Green Version]
  24. Genovese, G.; Kähler, A.K.; Handsaker, R.; Lindberg, J.; Rose, S.; Bakhoum, S.; Chambert, K.; Mick, E.; Neale, B.M.; Fromer, M.; et al. Clonal Hematopoiesis and Blood-Cancer Risk Inferred from Blood DNA Sequence. N. Engl. J. Med. 2014, 371, 2477–2487. [Google Scholar] [CrossRef] [Green Version]
  25. Xie, M.; Lu, C.; Wang, J.; McLellan, M.D.; Johnson, K.J.; Wendl, M.C.; McMichael, J.F.; Schmidt, H.K.; Yellapantula, V.; Miller, C.A.; et al. Age-related mutations associated with clonal hematopoietic expansion and malignancies. Nat. Med. 2014, 20, 1472–1478. [Google Scholar] [CrossRef]
  26. Barreyro, L.; Chlon, T.; Starczynowski, D.T. Chronic immune response dysregulation in MDS pathogenesis. Blood 2018, 132, 1553–1560. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Rea, I.M.; Gibson, D.; McGilligan, V.; McNerlan, S.E.; Alexander, H.D.; Ross, O. Age and Age-Related Diseases: Role of Inflammation Triggers and Cytokines. Front. Immunol. 2018, 9, 586. [Google Scholar] [CrossRef] [PubMed]
  28. Bogeska, R.; Kaschutnig, P.; Fawaz, M.; Mikecin, A.-M.; Büchler-Schäff, M.; Paffenholz, S.; Asada, N.; Frauhammer, F.; Buettner, F.; Ball, M.; et al. Hematopoietic Stem Cells Fail to Regenerate Following Inflammatory Challenge. bioRxiv 2020. [Google Scholar]
  29. Matatall, K.A.; Jeong, M.; Chen, S.; Sun, D.; Chen, F.; Mo, Q.; Kimmel, M.; King, K.Y. Chronic Infection Depletes Hematopoietic Stem Cells through Stress-Induced Terminal Differentiation. Cell Rep. 2016, 17, 2584–2595. [Google Scholar] [CrossRef] [Green Version]
  30. Esplin, B.L.; Shimazu, T.; Welner, R.S.; Garrett, K.P.; Nie, L.; Zhang, Q.; Humphrey, M.B.; Yang, Q.; Borghesi, L.A.; Kincade, P.W. Chronic Exposure to a TLR Ligand Injures Hematopoietic Stem Cells. J. Immunol. 2011, 186, 5367–5375. [Google Scholar] [CrossRef] [Green Version]
  31. Mann, M.; Mehta, A.; de Boer, C.; Kowalczyk, M.S.; Lee, K.; Haldeman, P.; Rogel, N.; Knecht, A.R.; Farouq, D.; Regev, A.; et al. Heterogeneous Responses of Hematopoietic Stem Cells to Inflammatory Stimuli Are Altered with Age. Cell Rep. 2018, 25, 2992–3005.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Abegunde, S.O.; Buckstein, R.; Wells, R.A.; Rauh, M.J. An inflammatory environment containing TNFα favors Tet2 -mutant clonal hematopoiesis. Exp. Hematol. 2018, 59, 60–65. [Google Scholar] [CrossRef] [PubMed]
  33. Leoni, C.; Montagner, S.; Rinaldi, A.; Bertoni, F.; Polletti, S.; Balestrieri, C.; Monticelli, S. Dnmt3arestrains mast cell inflammatory responses. Proc. Natl. Acad. Sci. USA 2017, 114, E1490–E1499. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Shi, X.; Zheng, Y.-H.; Xu, L.; Cao, C.; Dong, B.; Chen, X. The inflammatory cytokine profile of myelodysplastic syndromes. Medicine 2019, 98, e15844. [Google Scholar] [CrossRef]
  35. Molnár, L.; Berki, T.; Hussain, A.; Németh, P.; Losonczy, H. Detection of TNFα expression in the bone marrow and determination of TNFα production of peripheral blood mononuclear cells in myelodysplastic syndrome. Pathol. Oncol. Res. 2000, 6, 18–23. [Google Scholar] [CrossRef]
  36. Stifter, G.; Heiss, S.; Gastl, G.; Tzankov, A.; Stauder, R. Over-expression of tumor necrosis factor-alpha in bone marrow biopsies from patients with myelodysplastic syndromes: Relationship to anemia and prognosis. Eur. J. Haematol. 2005, 75, 485–491. [Google Scholar] [CrossRef] [PubMed]
  37. Zhang, Z.; Li, X.; Guo, J.; Xu, F.; He, Q.; Zhao, Y.; Yang, Y.; Gu, S.; Zhang, Y.; Wu, L.; et al. Interleukin-17 enhances the production of interferon-γ and tumour necrosis factor-α by bone marrow T lymphocytes from patients with lower risk myelodysplastic syndromes. Eur. J. Haematol. 2013, 90, 375–384. [Google Scholar] [CrossRef]
  38. Pardanani, A.; Finke, C.; Lasho, T.L.; Al-Kali, A.; Begna, K.H.; A Hanson, C.; Tefferi, A. IPSS-independent prognostic value of plasma CXCL10, IL-7 and IL-6 levels in myelodysplastic syndromes. Leukemia 2011, 26, 693–699. [Google Scholar] [CrossRef] [PubMed]
  39. De Matos, A.G.; Junior, H.L.R.; Borges, D.D.P.; Okubo, B.M.; De Sousa, J.C.; Barbosa, M.C.; De Castro, M.F.; Gonçalves, R.P.; Pinheiro, R.F.; Magalhães, S.M.M. Interleukin-8 and nuclear factor kappa B are increased and positively correlated in myelodysplastic syndrome. Med. Oncol. 2017, 34, 168. [Google Scholar] [CrossRef] [PubMed]
  40. Tsimberidou, A.M.; Estey, E.; Wen, S.; Pierce, S.; Kantarjian, H.; Albitar, M.; Kurzrock, R. The prognostic significance of cytokine levels in newly diagnosed acute myeloid leukemia and high-risk myelodysplastic syndromes. Cancer 2008, 113, 1605–1613. [Google Scholar] [CrossRef]
  41. Shetty, V.; Mundle, S.; Alvi, S.; Showel, M.; Broady-Robinson, L.; Dar, S.; Borok, R.; Showel, J.; Gregory, S.; Rifkin, S.; et al. Measurement of apoptosis, proliferation and three cytokines in 46 patients with myelodysplastic syndromes. Leuk. Res. 1996, 20, 891–900. [Google Scholar] [CrossRef]
  42. Zeng, Q.; Shu, J.; Hu, Q.; Zhou, S.-H.; Qian, Y.-M.; Hu, M.-H.; Hu, L.-Y.; Wang, Y.-G.; Zhou, Y.-M.; Lu, J.-H. Apoptosis in human myelodysplastic syndrome CD34+ cells is modulated by the upregulation of TLRs and histone H4 acetylation via a β-arrestin 1 dependent mechanism. Exp. Cell Res. 2016, 340, 22–31. [Google Scholar] [CrossRef] [PubMed]
  43. Kordasti, S.Y.; Afzali, B.; Lim, Z.; Ingram, W.; Hayden, J.; Barber, L.; Matthews, K.; Chelliah, R.; Guinn, B.; Lombardi, G.; et al. IL-17-producing CD4+T cells, pro-inflammatory cytokines and apoptosis are increased in low risk myelodysplastic syndrome. Br. J. Haematol. 2009, 145, 64–72. [Google Scholar] [CrossRef]
  44. Ratajczak, M.Z.; Bujko, K.; Cymer, M.; Thapa, A.; Adamiak, M.; Ratajczak, J.; Abdel-Latif, A.K.; Kucia, M. The Nlrp3 inflammasome as a “rising star” in studies of normal and malignant hematopoiesis. Leukemia 2020, 34, 1512–1523. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Sallman, D.A.; List, A. The central role of inflammatory signaling in the pathogenesis of myelodysplastic syndromes. Blood 2019, 133, 1039–1048. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Banerjee, T.; Calvi, L.M.; Becker, M.W.; Liesveld, J.L. Flaming and fanning: The Spectrum of inflammatory influences in myelodysplastic syndromes. Blood Rev. 2019, 36, 57–69. [Google Scholar] [CrossRef] [PubMed]
  47. Bergsbaken, T.; Fink, S.; Cookson, B.T. Pyroptosis: Host cell death and inflammation. Nat. Rev. Genet. 2009, 7, 99–109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Basiorka, A.A.; McGraw, K.L.; Eksioglu, E.A.; Chen, X.; Johnson, J.; Zhang, L.; Zhang, Q.; Irvine, B.A.; Cluzeau, T.; Sallman, D.A.; et al. The NLRP3 inflammasome functions as a driver of the myelodysplastic syndrome phenotype. Blood 2016, 128, 2960–2975. [Google Scholar] [CrossRef] [PubMed]
  49. Simard, J.-C.; Cesaro, A.; Chapeton-Montes, J.; Tardif, M.; Antoine, F.; Girard, D.; Tessier, P.A. S100A8 and S100A9 Induce Cytokine Expression and Regulate the NLRP3 Inflammasome via ROS-Dependent Activation of NF-κB1. PLoS ONE 2013, 8, e72138. [Google Scholar] [CrossRef] [Green Version]
  50. Rhyasen, G.W.; Bolanos, L.; Fang, J.; Jerez, A.; Wunderlich, M.; Rigolino, C.; Mathews, L.; Ferrer, M.; Southall, N.; Guha, R.; et al. Targeting IRAK1 as a Therapeutic Approach for Myelodysplastic Syndrome. Cancer Cell 2013, 24, 90–104. [Google Scholar] [CrossRef] [Green Version]
  51. Shi, J.; Zhao, Y.; Wang, K.; Shi, X.; Wang, Y.; Huang, H.; Zhuang, Y.; Cai, T.; Wang, F.; Shao, F. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature 2015, 526, 660–665. [Google Scholar] [CrossRef]
  52. Liu, X.; Zhang, Z.; Ruan, J.; Pan, Y.; Magupalli, V.G.; Wu, H.; Lieberman, J. Inflammasome-activated gasdermin D causes pyroptosis by forming membrane pores. Nature 2016, 535, 153–158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Giudice, V.; Wu, Z.; Kajigaya, S.; Ibanez, M.D.P.F.; Rios, O.; Cheung, F.; Ito, S.; Young, N.S. Circulating S100A8 and S100A9 protein levels in plasma of patients with acquired aplastic anemia and myelodysplastic syndromes. Cytokine 2018, 113, 462–465. [Google Scholar] [CrossRef]
  54. 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]
  55. Nagai, Y.; Garrett, K.P.; Ohta, S.; Bahrun, U.; Kouro, T.; Akira, S.; Takatsu, K.; Kincade, P.W. Toll-like Receptors on Hematopoietic Progenitor Cells Stimulate Innate Immune System Replenishment. Immunity 2006, 24, 801–812. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Paracatu, L.C.; Schuettpelz, L.G. Contribution of Aberrant Toll Like Receptor Signaling to the Pathogenesis of Myelodysplastic Syndromes. Front. Immunol. 2020, 11, 1236. [Google Scholar] [CrossRef]
  57. Wei, Y.; Dimicoli, S.; Bueso-Ramos, C.; Chen, R.; Yang, H.; Neuberg, N.; Pierce, S.; Jia, Y.; Zheng, H.; Wang, H.; et al. Toll-like receptor alterations in myelodysplastic syndrome. Leukemia 2013, 27, 1832–1840. [Google Scholar] [CrossRef] [Green Version]
  58. Maratheftis, C.I.; Andreakos, E.; Moutsopoulos, H.M.; Voulgarelis, M. Toll-like Receptor-4 Is Up-Regulated in Hematopoietic Progenitor Cells and Contributes to Increased Apoptosis in Myelodysplastic Syndromes. Clin. Cancer Res. 2007, 13, 1154–1160. [Google Scholar] [CrossRef] [Green Version]
  59. Dimicoli, S.; Wei, Y.; Bueso-Ramos, C.; Yang, H.; Dinardo, C.; Jia, Y.; Zheng, H.; Fang, Z.; Nguyen, M.; Pierce, S.; et al. Overexpression of the Toll-Like Receptor (TLR) Signaling Adaptor MYD88, but Lack of Genetic Mutation, in Myelodysplastic Syndromes. PLoS ONE 2013, 8, e71120. [Google Scholar] [CrossRef]
  60. Monlish, D.A.; Greenberg, Z.J.; Bhatt, S.T.; Leonard, K.M.; Romine, M.P.; Dong, Q.; Bendesky, L.; Duncavage, E.J.; Magee, J.A.; Schuettpelz, L.G. TLR2/6 signaling promotes the expansion of premalignant hematopoietic stem and progenitor cells in the NUP98–HOXD13 mouse model of MDS. Exp. Hematol. 2020, 88, 42–55. [Google Scholar] [CrossRef] [PubMed]
  61. Velegraki, M.; Papakonstanti, E.; Mavroudi, I.; Psyllaki, M.; Tsatsanis, C.; Oulas, A.; Iliopoulos, I.; Katonis, P.; Papadaki, H.A. Impaired clearance of apoptotic cells leads to HMGB1 release in the bone marrow of patients with myelodysplastic syndromes and induces TLR4-mediated cytokine production. Haematologica 2013, 98, 1206–1215. [Google Scholar] [CrossRef] [Green Version]
  62. Kapor, S.; Santibanez, J. Myeloid-Derived Suppressor Cells and Mesenchymal Stem/Stromal Cells in Myeloid Malignancies. J. Clin. Med. 2021, 10, 2788. [Google Scholar] [CrossRef] [PubMed]
  63. Han, Y.; Wang, H.; Shao, Z. Monocyte-Derived Macrophages Are Impaired in Myelodysplastic Syndrome. J. Immunol. Res. 2016, 2016, 5479013. [Google Scholar] [CrossRef]
  64. Bento, L.C.; Bacal, N.S.; Rocha, F.A.; Severino, P.; Marti, L.C. Bone Marrow Monocytes and Derived Dendritic Cells from Myelodysplastic Patients Have Functional Abnormalities Associated with Defective Response to Bacterial Infection. J. Immunol. 2020, 204, 2098–2109. [Google Scholar] [CrossRef]
  65. Zhang, G.; Yang, L.; Han, Y.; Niu, H.; Yan, L.; Shao, Z.; Xing, L.; Wang, H. Abnormal Macrophage Polarization in Patients with Myelodysplastic Syndrome. Mediat. Inflamm. 2021, 2021, 9913382. [Google Scholar] [CrossRef]
  66. Saft, L.; Björklund, E.; Berg, E.; Hellström-Lindberg, E.; Porwit, A. Bone marrow dendritic cells are reduced in patients with high-risk myelodysplastic syndromes. Leuk. Res. 2012, 37, 266–273. [Google Scholar] [CrossRef]
  67. 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] [PubMed]
  68. Ma, L.; Delforge, M.; Van Duppen, V.; Verhoef, G.; Emanuel, B.; Boogaerts, M.; Hagemeijer, A.; Vandenberghe, P. Circulating myeloid and lymphoid precursor dendritic cells are clonally involved in myelodysplastic syndromes. Leukemia 2004, 18, 1451–1456. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Mailloux, A.W.; Sugimori, C.; Komrokji, R.S.; Yang, L.; Maciejewski, J.P.; Sekeres, M.A.; Paquette, R.; Loughran, T.P.; List, A.F.; Epling-Burnette, P.K. Expansion of effector memory regulatory T cells represents a novel prognostic factor in lower risk myelodysplastic syndrome. J. Immunol. 2012, 189, 3198–3208. [Google Scholar] [CrossRef] [Green Version]
  70. Epling-Burnette, P.K.; Bai, F.; Painter, J.S.; Rollison, D.E.; Salih, H.R.; Krusch, M.; Zou, J.; Ku, E.; Zhong, B.; Boulware, D.; 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]
  71. 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]
  72. Stringaris, K.; Marin, D.; Barrett, A.J.; Hills, R.; Sobieski, C.; Cao, K.; Saltarrelli, J.G.; Daher, M.; Shaim, H.; Smith, N.; et al. KIR gene haplotype: An independent predictor of clinical outcome in MDS patients. Blood 2016, 128, 2819–2823. [Google Scholar] [CrossRef] [Green Version]
  73. Williams, P.; Basu, S.; Garcia-Manero, G.; Hourigan, C.S.; Oetjen, K.A.; Cortes, J.E.; Ravandi, F.; Jabbour, E.J.; Al-Hamal, Z.; Konopleva, M.; et al. The distribution of T-cell subsets and the expression of immune checkpoint receptors and ligands in patients with newly diagnosed and relapsed acute myeloid leukemia. Cancer 2018, 125, 1470–1481. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Sallman, D.A.; McLemore, A.F.; Aldrich, A.L.; Komrokji, R.S.; McGraw, K.L.; Dhawan, A.; Geyer, S.; Hou, H.-A.; Eksioglu, E.A.; Sullivan, A.; et al. TP53 mutations in myelodysplastic syndromes and secondary AML confer an immunosuppressive phenotype. Blood 2020, 136, 2812–2823. [Google Scholar] [CrossRef]
  75. Epling-Burnette, P.K.; Painter, J.S.; E Rollison, D.; Ku, E.; Vendron, D.; Widen, R.; Boulware, D.; Zou, J.X.; Bai, F.; List, A. Prevalence and clinical association of clonal T-cell expansions in Myelodysplastic Syndrome. Leukemia 2007, 21, 659–667. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Tcvetkov, N.; Gusak, A.; Morozova, E.; Moiseev, I.; Baykov, V.; Barabanshikova, M.; Lepik, K.; Bakin, E.; Vlasova, J.; Osipova, A.; et al. Immune checkpoints bone marrow expression as the predictor of clinical outcome in myelodysplastic syndrome. Leuk. Res. Rep. 2020, 14, 100215. [Google Scholar] [CrossRef]
  77. Yang, H.; Bueso-Ramos, C.; Dinardo, C.D.; Estecio, M.; Davanlou, M.; Geng, Q.-R.; Fang, Z.; Nguyen, M.; Pierce, S.; Wei, Y.; et al. Expression of PD-L1, PD-L2, PD-1 and CTLA4 in myelodysplastic syndromes is enhanced by treatment with hypomethylating agents. Leukemia 2013, 28, 1280–1288. [Google Scholar] [CrossRef] [PubMed]
  78. Moskorz, W.; Cosmovici, C.; Jäger, P.S.; Cadeddu, R.P.; Timm, J.; Haas, R. Myelodysplastic syndrome patients display alterations in their immune status reflected by increased PD-L1-expressing stem cells and highly dynamic exhausted T-cell frequencies. Br. J. Haematol. 2021, 193, 941–945. [Google Scholar] [CrossRef] [PubMed]
  79. Kondo, A.; Yamashita, T.; Tamura, H.; Zhao, W.; Tsuji, T.; Shimizu, M.; Shinya, E.; Takahashi, H.; Tamada, K.; Chen, L.; et al. Interferon-γ and tumor necrosis factor-α induce an immunoinhibitory molecule, B7-H1, via nuclear factor-κB activation in blasts in myelodysplastic syndromes. Blood 2010, 116, 1124–1131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Cheng, P.; Eksioglu, E.A.; Chen, X.; Kandell, W.; Le Trinh, T.; Cen, L.; Qi, J.; Sallman, D.A.; Zhang, Y.; Tu, N.; et al. S100A9-induced overexpression of PD-1/PD-L1 contributes to ineffective hematopoiesis in myelodysplastic syndromes. Leukemia 2019, 33, 2034–2046. [Google Scholar] [CrossRef] [PubMed]
  81. Tao, J.-L.; Li, L.-J.; Fu, R.; Wang, H.-Q.; Jiang, H.-J.; Yue, L.-Z.; Zhang, W.; Liu, H.; Ruan, E.-B.; Qu, W.; et al. Elevated TIM3+ hematopoietic stem cells in untreated myelodysplastic syndrome displayed aberrant differentiation, overproliferation and decreased apoptosis. Leuk. Res. 2014, 38, 714–721. [Google Scholar] [CrossRef]
  82. Tao, J.; Li, L.; Wang, Y.; Fu, R.; Wang, H.; Shao, Z. Increased TIM3+CD8+T cells in Myelodysplastic Syndrome patients displayed less perforin and granzyme B secretion and higher CD95 expression. Leuk. Res. 2016, 51, 49–55. [Google Scholar] [CrossRef] [PubMed]
  83. Asayama, T.; Tamura, H.; Ishibashi, M.; Kuribayashi-Hamada, Y.; Onodera-Kondo, A.; Okuyama, N.; Yamada, A.; Shimizu, M.; Moriya, K.; Takahashi, H.; et al. Functional expression of Tim-3 on blasts and clinical impact of its ligand galectin-9 in myelodysplastic syndromes. Oncotarget 2017, 8, 88904–88917. [Google Scholar] [CrossRef] [Green Version]
  84. Tao, J.; Han, D.; Gao, S.; Zhang, W.; Yu, H.; Liu, P.; Fu, R.; Li, L.; Shao, Z. CD8 + T cells exhaustion induced by myeloid-derived suppressor cells in myelodysplastic syndromes patients might be through TIM3/Gal-9 pathway. J. Cell. Mol. Med. 2019, 24, 1046–1058. [Google Scholar] [CrossRef] [Green Version]
  85. Meng, F.; Li, L.; Lu, F.; Yue, J.; Liu, Z.; Zhang, W.; Fu, R. Overexpression of TIGIT in NK and T Cells Contributes to Tumor Immune Escape in Myelodysplastic Syndromes. Front. Oncol. 2020, 10, 1595. [Google Scholar] [CrossRef]
  86. Moiseev, I.S.; Tcvetkov, N.Y.; Barkhatov, I.M.; Barabanshikova, M.V.; Bug, D.S.; Petuhova, N.V.; Tishkov, A.V.; Bakin, E.A.; Izmailova, E.A.; Shakirova, A.I.; et al. High mutation burden in the checkpoint and micro-RNA processing genes in myelodysplastic syndrome. PLoS ONE 2021, 16, e0248430. [Google Scholar] [CrossRef]
  87. Bouchliou, I.; Miltiades, P.; Nakou, E.; Spanoudakis, E.; Goutzouvelidis, A.; Vakalopoulou, S.; Garypidou, V.; Kotoula, V.; Bourikas, G.; Tsatalas, C.; et al. Th17 and Foxp3+ T regulatory cell dynamics and distribution in myelodysplastic syndromes. Clin. Immunol. 2011, 139, 350–359. [Google Scholar] [CrossRef] [PubMed]
  88. Lopes, M.R.; Traina, F.; Campos, P.D.M.; Pereira, J.K.N.; Machado-Neto, J.A.; Machado, H.D.C.; Gilli, S.C.O.; Saad, S.T.O.; Favaro, P. IL10 inversely correlates with the percentage of CD8+ cells in MDS patients. Leuk. Res. 2013, 37, 541–546. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Kordasti, S.Y.; Ingram, W.; Hayden, J.; Darling, D.; Barber, L.; Afzali, B.; Lombardi, G.; Wlodarski, M.; 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]
  90. 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 2008, 23, 510–518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Haroun, F.; Solola, S.A.; Nassereddine, S.; Tabbara, I. PD-1 signaling and inhibition in AML and MDS. Ann. Hematol. 2017, 96, 1441–1448. [Google Scholar] [CrossRef]
  92. Kikushige, Y.; Shima, T.; Takayanagi, S.-I.; Urata, S.; Miyamoto, T.; Iwasaki, H.; Takenaka, K.; Teshima, T.; Tanaka, T.; Inagaki, Y.; et al. TIM-3 Is a Promising Target to Selectively Kill Acute Myeloid Leukemia Stem Cells. Cell Stem Cell 2010, 7, 708–717. [Google Scholar] [CrossRef] [Green Version]
  93. Jiang, H.; Fu, R.; Wang, H.; Li, L.; Liu, H.; Shao, Z. CD47 is expressed abnormally on hematopoietic cells in myelodysplastic syndrome. Leuk. Res. 2013, 37, 907–910. [Google Scholar] [CrossRef] [PubMed]
  94. Dong, X.; Han, Y.; Liu, Y.; Yang, L.; Niu, H.; Yan, L.; Liu, C.; Shao, Z.; Xing, L.; Wang, H. Phagocytosis checkpoints on hematopoietic stem cells in patients with myelodysplastic syndromes. Asia-Pac. J. Clin. Oncol. 2021. [Google Scholar] [CrossRef]
  95. Fuster, J.J.; MacLauchlan, S.; Zuriaga, M.A.; Polackal, M.N.; Ostriker, A.C.; Chakraborty, R.; Wu, C.-L.; Sano, S.; Muralidharan, S.; Rius, C.; et al. Clonal hematopoiesis associated with TET2 deficiency accelerates atherosclerosis development in mice. Science 2017, 355, 842–847. [Google Scholar] [CrossRef] [Green Version]
  96. Zhang, C.R.; Nix, D.; Gregory, M.; Ciorba, M.A.; Ostrander, E.L.; Newberry, R.D.; Spencer, D.H.; Challen, G.A. Inflammatory cytokines promote clonal hematopoiesis with specific mutations in ulcerative colitis patients. Exp. Hematol. 2019, 80, 36–41.e3. [Google Scholar] [CrossRef]
  97. Cook, E.K.; Izukawa, T.; Young, S.; Rosen, G.; Jamali, M.; Zhang, L.; Johnson, D.; Bain, E.; Hilland, J.; Ferrone, C.K.; et al. Comorbid and inflammatory characteristics of genetic subtypes of clonal hematopoiesis. Blood Adv. 2019, 3, 2482–2486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Zhao, L.-P.; Boy, M.; Azoulay, C.; Clappier, E.; Sébert, M.; Amable, L.; Klibi, J.; Benlagha, K.; Espeli, M.; Balabanian, K.; et al. MDS/CMML with TET2 or IDH mutation Are Associated with Systemic Inflammatory and Autoimmune Diseases (SIAD) and T Cell Dysregulation. Blood 2020, 136, 31–32. [Google Scholar] [CrossRef]
  99. Smith, M.A.; Choudhary, G.S.; Pellagatti, A.; Choi, K.; Bolanos, L.C.; Bhagat, T.D.; Gordon-Mitchell, S.; Von Ahrens, D.; Pradhan, K.; Steeples, V.; et al. U2AF1 mutations induce oncogenic IRAK4 isoforms and activate innate immune pathways in myeloid malignancies. Nature 2019, 21, 640–650. [Google Scholar] [CrossRef] [PubMed]
  100. O’Connor, B.P.; Danhorn, T.; De Arras, L.; Flatley, B.R.; Marcus, R.A.; Farias-Hesson, E.; Leach, S.M.; Alper, S. Regulation of Toll-like Receptor Signaling by the SF3a mRNA Splicing Complex. PLoS Genet. 2015, 11, e1004932. [Google Scholar] [CrossRef] [Green Version]
  101. Pollyea, D.A.; Harris, C.; Rabe, J.L.; Hedin, B.R.; De Arras, L.; Katz, S.; Wheeler, E.; Bejar, R.; Walter, M.J.; Jordan, C.T.; et al. Myelodysplastic syndrome-associated spliceosome gene mutations enhance innate immune signaling. Haematologica 2019, 104, e388–e392. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Beck, D.B.; Ferrada, M.A.; Sikora, K.A.; Ombrello, A.K.; Collins, J.C.; Pei, W.; Balanda, N.; Ross, D.L.; Cardona, D.O.; Wu, Z.; et al. Somatic Mutations in UBA1 and Severe Adult-Onset Autoinflammatory Disease. N. Engl. J. Med. 2020, 383, 2628–2638. [Google Scholar] [CrossRef] [PubMed]
  103. Georgin-Lavialle, S.; Terrier, B.; Guedon, A.; Heiblig, M.; Comont, T.; Lazaro, E.; Lacombe, V.; Terriou, L.; Ardois, S.; Bouaziz, J.; et al. Further characterization of clinical and laboratory features occurring in VEXAS syndrome in a large-scale analysis of multicenter case-series of 116 French patients. Br. J. Dermatol. 2021. [Google Scholar] [CrossRef] [PubMed]
  104. Poulter, J.A.; Collins, J.C.; Cargo, C.; De Tute, R.M.; Evans, P.; Cardona, D.O.; Bowen, D.T.; Cunnington, J.R.; Baguley, E.; Quinn, M.; et al. Novel somatic mutations in UBA1 as a cause of VEXAS syndrome. Blood 2021, 137, 3676–3681. [Google Scholar] [CrossRef] [PubMed]
  105. Bourbon, E.; Heiblig, M.; Valentin, M.G.; Barba, T.; Durel, C.-A.; Lega, J.C.; Barraco, F.; Sève, P.; Jamilloux, Y.; Sujobert, P. Therapeutic options in VEXAS syndrome: Insights from a retrospective series. Blood 2021, 137, 3682–3684. [Google Scholar] [CrossRef] [PubMed]
  106. Obiorah, I.E.; Patel, B.A.; Groarke, E.M.; Wang, W.; Trick, M.; Ombrello, A.K.; Ferrada, M.A.; Wu, Z.; Gutierrez-Rodrigues, F.; Lotter, J.; et al. Benign and malignant hematologic manifestations in patients with VEXAS syndrome due to somatic mutations in UBA1. Blood Adv. 2021, 5, 3203–3215. [Google Scholar] [CrossRef]
  107. Comont, T.; Heiblig, M.; Rivière, E.; Terriou, L.; Rossignol, J.; Bouscary, D.; Rieu, V.; Le Guenno, G.; Mathian, A.; Aouba, A.; et al. Azacitidine for patients with Vacuoles, E1 Enzyme, X-linked, Autoinflammatory, Somatic syndrome (VEXAS) and myelodysplastic syndrome: Data from the French VEXAS registry. Br. J. Haematol. 2021. [Google Scholar] [CrossRef]
  108. Thol, F.; Klesse, S.; Köhler, L.; Gabdoulline, R.; Kloos, A.; Liebich, A.; Wichmann, M.; Chaturvedi, A.; Fabisch, J.; I Gaidzik, V.; et al. Acute myeloid leukemia derived from lympho-myeloid clonal hematopoiesis. Leukemia 2016, 31, 1286–1295. [Google Scholar] [CrossRef]
  109. Dickinson, A.M.; Norden, J.; Li, S.; Hromadnikova, I.; Schmid, C.; Schmetzer, H.; Jochem-Kolb, H. Graft-versus-Leukemia Effect Following Hematopoietic Stem Cell Transplantation for Leukemia. Front. Immunol. 2017, 8, 496. [Google Scholar] [CrossRef] [PubMed]
  110. Chen, S.; Jiang, B.; Da, W.; Gong, M.; Guan, M. Treatment of Myelodysplastic Syndrome with Cyclosporin A. Int. J. Hematol. 2007, 85, 11–17. [Google Scholar] [CrossRef]
  111. Passweg, J.R.; Giagounidis, A.A.; Simcock, M.; Aul, C.; Dobbelstein, C.; Stadler, M.; Ossenkoppele, G.; Hofmann, W.-K.; Schilling, K.; Tichelli, A.; et al. Immunosuppressive Therapy for Patients With Myelodysplastic Syndrome: A Prospective Randomized Multicenter Phase III Trial Comparing Antithymocyte Globulin Plus Cyclosporine with Best Supportive Care—SAKK 33/99. J. Clin. Oncol. 2011, 29, 303–309. [Google Scholar] [CrossRef] [Green Version]
  112. Komrokji, R.S.; Mailloux, A.W.; Chen, D.-T.; Sekeres, M.A.; Paquette, R.; Fulp, W.J.; Sugimori, C.; Paleveda-Pena, J.; Maciejewski, J.P.; List, A.F.; et al. A phase II multicenter rabbit anti-thymocyte globulin trial in patients with myelodysplastic syndromes identifying a novel model for response prediction. Haematologica 2014, 99, 1176–1183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Kochenderfer, J.N.; Kobayashi, S.; Wieder, E.D.; Su, C.; Molldrem, J.J. Loss of T-lymphocyte clonal dominance in patients with myelodysplastic syndrome responsive to immunosuppression. Blood 2002, 100, 3639–3645. [Google Scholar] [CrossRef] [Green Version]
  114. Lim, Z.Y.; Killick, S.; Germing, U.; Cavenagh, J.; Culligan, D.; Bacigalupo, A.; Marsh, J.; Mufti, G.J. Low IPSS score and bone marrow hypocellularity in MDS patients predict hematological responses to antithymocyte globulin. Leukemia 2007, 21, 1436–1441. [Google Scholar] [CrossRef] [PubMed]
  115. Stahl, M.; Deveaux, M.; De Witte, T.; Neukirchen, J.; Sekeres, M.A.; Brunner, A.M.; Roboz, G.J.; Steensma, D.P.; Bhatt, V.R.; Platzbecker, U.; et al. The use of immunosuppressive therapy in MDS: Clinical outcomes and their predictors in a large international patient cohort. Blood Adv. 2018, 2, 1765–1772. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Fenaux, P.; Giagounidis, A.; Selleslag, D.; Beyne-Rauzy, O.; Mufti, G.; Mittelman, M.; Muus, P.; Boekhorst, P.T.; Sanz, G.; Del Cañizo, C.; et al. A randomized phase 3 study of lenalidomide versus placebo in RBC transfusion-dependent patients with Low-/Intermediate-1-risk myelodysplastic syndromes with del5q. Blood 2011, 118, 3765–3776. [Google Scholar] [CrossRef] [PubMed]
  117. Fozza, C.; Crobu, V.; Isoni, M.A.; Dore, F. The immune landscape of myelodysplastic syndromes. Crit. Rev. Oncol. 2016, 107, 90–99. [Google Scholar] [CrossRef] [PubMed]
  118. Krönke, J.; Fink, E.; Hollenbach, P.W.; Macbeth, K.J.; Hurst, S.N.; Udeshi, N.D.; Chamberlain, P.P.; Mani, D.R.; Man, H.W.; Gandhi, A.K.; et al. Lenalidomide induces ubiquitination and degradation of CK1α in del(5q) MDS. Nat. Cell Biol. 2015, 523, 183–188. [Google Scholar] [CrossRef] [PubMed]
  119. Giagounidis, A.; Mufti, G.J.; Mittelman, M.; Sanz, G.; Platzbecker, U.; Muus, P.; Selleslag, D.; Beyne-Rauzy, O.; Boekhorst, P.T.; Del Cañizo, C.; et al. Outcomes in RBC transfusion-dependent patients with L ow-/ I ntermediate-1-risk myelodysplastic syndromes with isolated deletion 5q treated with lenalidomide: A subset analysis from the MDS -004 study. Eur. J. Haematol. 2014, 93, 429–438. [Google Scholar] [CrossRef] [Green Version]
  120. Lai, C.; Ranpura, V.; Wu, C.; Olnes, M.J.; Parikh, A.R.; Shenoy, A.; Thompson, J.; Weinstein, B.; Scheinberg, P.; Barrett, A.J.; et al. Long-term outcomes in myelodysplastic syndrome patients treated with alemtuzumab. Blood Adv. 2019, 3, 980–983. [Google Scholar] [CrossRef] [PubMed]
  121. Baron, F.; Suciu, S.; Amadori, S.; Muus, P.; Zwierzina, H.; Denzlinger, C.; Delforge, M.; Thyss, A.; Selleslag, D.; Indrak, K.; et al. Value of infliximab (Remicade(R)) in patients with low-risk myelodysplastic syndrome: Final results of a randomized phase II trial (EORTC trial 06023) of the EORTC Leukemia Group. Haematologica 2011, 97, 529–533. [Google Scholar] [CrossRef]
  122. Scott, B.L.; Ramakrishnan, A.; Storer, B.; Becker, P.S.; Petersdorf, S.; Estey, E.H.; Deeg, H. Prolonged responses in patients with MDS and CMML treated with azacitidine and etanercept. Br. J. Haematol. 2010, 148, 944–947. [Google Scholar] [CrossRef] [Green Version]
  123. Mekinian, A.; Dervin, G.; Lapidus, N.; Kahn, J.E.; Terriou, L.; Liozon, E.; Grignano, E.; Piette, J.-C.; Rauzy, O.B.; Grobost, V.; et al. Biologics in myelodysplastic syndrome-related systemic inflammatory and autoimmune diseases: French multicenter retrospective study of 29 patients. Autoimmun. Rev. 2017, 16, 903–910. [Google Scholar] [CrossRef] [Green Version]
  124. Malik, J.; Kim, A.R.; Tyre, K.A.; Cherukuri, A.R.; Palis, J. Erythropoietin critically regulates the terminal maturation of murine and human primitive erythroblasts. Haematologica 2013, 98, 1778–1787. [Google Scholar] [CrossRef]
  125. Suragani, R.N.V.S.; Cadena, S.M.; Cawley, S.M.; Sako, D.; Mitchell, D.; Li, R.; Davies, M.V.; Alexander, M.J.; Devine, M.; Loveday, K.S.; et al. Transforming growth factor-β superfamily ligand trap ACE-536 corrects anemia by promoting late-stage erythropoiesis. Nat. Med. 2014, 20, 408–414. [Google Scholar] [CrossRef] [PubMed]
  126. Fenaux, P.; Platzbecker, U.; Mufti, G.J.; Garcia-Manero, G.; Buckstein, R.; Santini, V.; Díez-Campelo, M.; Finelli, C.; Cazzola, M.; Ilhan, O.; et al. Luspatercept in Patients with Lower-Risk Myelodysplastic Syndromes. N. Engl. J. Med. 2020, 382, 140–151. [Google Scholar] [CrossRef]
  127. Kubasch, A.S.; Fenaux, P.; Platzbecker, U. Development of luspatercept to treat ineffective erythropoiesis. Blood Adv. 2021, 5, 1565–1575. [Google Scholar] [CrossRef] [PubMed]
  128. Garcia-Manero, G.; Platzbecker, U.; Tarantolo, S.R.; Gropper, S.; Talati, C.; Götze, K.S.; Dugan, J.; Winer, E.S.; Martinez, D.E.; Lieberman, C.; et al. A Phase 1, Open Label Dose Escalation Trial Evaluating the Safety, Pharmacokinetics, Pharmacodynamics, and Clinical Activity of Orally Administered CA-4948 in Patients with Acute Myelogenous Leukemia or Myelodysplastic Syndrome. Blood 2020, 136, 16. [Google Scholar] [CrossRef]
  129. Jonas, B.A.; Curtin, P.T.; Schiller, G.J.; Jeyakumar, D.; Wieduwilt, M.J.; Abedi, M.; Bejar, R.; Chow, H.; Oesterich, L.G.; Logan, A.C. A Phase 1 Trial of Ibrutinib (IBR) and Azacitidine (AZA) for the Treatment of Higher-Risk Myelodysplastic Syndromes (HR-MDS): Updated Results of University of California Hematologic Malignancies Consortium (UCHMC) Study 1503. Blood 2018, 132, 3088. [Google Scholar] [CrossRef]
  130. Attar, E.C.; Amrein, P.C.; Fraser, J.W.; Fathi, A.T.; McAfee, S.; Wadleigh, M.; DeAngelo, D.J.; Steensma, D.; Stone, R.M.; Foster, J.; et al. Phase I dose escalation study of bortezomib in combination with lenalidomide in patients with myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). Leuk. Res. 2013, 37, 1016–1020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Huselton, E.; Cashen, A.F.; DiPersio, J.F.; Jacoby, M.; Pusic, I.; Romee, R.; Schroeder, M.A.; Uy, G.L.; Westervelt, P. Updated Study Results of CX-01, an Inhibitor of CXCL12/CXCR4, and Azacitidine for the Treatment of Hypomethylating Agent Refractory AML and MDS. Blood 2019, 134, 3915. [Google Scholar] [CrossRef]
  132. Garcia-Manero, G.; Jabbour, E.J.; Konopleva, M.Y.; Daver, N.G.; Borthakur, G.; Dinardo, C.D.; Bose, P.; Patel, P.; Komrokji, R.S.; Shastri, A.; et al. A Clinical Study of Tomaralimab (OPN-305), a Toll-like Receptor 2 (TLR-2) Antibody, in Heavily Pre-Treated Transfusion Dependent Patients with Lower Risk Myelodysplastic Syndromes (MDS) That Have Received and Failed on Prior Hypomethylating Agent (HMA) Therapy. Blood 2018, 132, 798. [Google Scholar] [CrossRef]
  133. Linder, K.; Lulla, P. Myelodysplastic syndrome and immunotherapy novel to next in-line treatments. Hum. Vaccines Immunother. 2021, 17, 2602–2616. [Google Scholar] [CrossRef] [PubMed]
  134. Zeidan, A.M.; Knaus, H.A.; Robinson, T.M.; Towlerton, A.M.; Warren, E.; Zeidner, J.; Blackford, A.L.; Duffield, A.S.; Rizzieri, D.; Frattini, M.G.; et al. A Multi-center Phase I Trial of Ipilimumab in Patients with Myelodysplastic Syndromes following Hypomethylating Agent Failure. Clin. Cancer Res. 2018, 24, 3519–3527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Morita, K.; Kantarjian, H.M.; Bravo, G.M.; Sasaki, K.; Daver, N.; Jabbour, E.; Alvarado, Y.; Chien, K.S.; Dinardo, C.D.; Ravandi, F.; et al. A Phase II Study of Double Immune Checkpoint Inhibitor Blockade with Nivolumab and Ipilimumab with or without Azacitidine in Patients with Myelodysplastic Syndrome (MDS). Blood 2020, 136, 7–9. [Google Scholar] [CrossRef]
  136. Boddu, P.; Kantarjian, H.; Garcia-Manero, G.; Allison, J.; Sharma, P.; Daver, N. The emerging role of immune checkpoint based approaches in AML and MDS. Leuk. Lymphoma 2017, 59, 790–802. [Google Scholar] [CrossRef] [PubMed]
  137. Ravandi, F.; Assi, R.; Daver, N.; Benton, C.B.; Kadia, T.; A Thompson, P.; Borthakur, G.; Alvarado, Y.; Jabbour, E.J.; Konopleva, M.; et al. Idarubicin, cytarabine, and nivolumab in patients with newly diagnosed acute myeloid leukaemia or high-risk myelodysplastic syndrome: A single-arm, phase 2 study. Lancet Haematol. 2019, 6, e480–e488. [Google Scholar] [CrossRef]
  138. Garcia-Manero, G.; Sasaki, K.; Montalban-Bravo, G.; Daver, N.G.; Jabbour, E.J.; Alvarado, Y.; Dinardo, C.D.; Ravandi, F.; Borthakur, G.; Bose, P.; et al. A Phase II Study of Nivolumab or Ipilimumab with or without Azacitidine for Patients with Myelodysplastic Syndrome (MDS). Blood 2018, 132, 465. [Google Scholar] [CrossRef]
  139. Chien, K.S.; Kim, K.; Nogueras-Gonzalez, G.M.; Borthakur, G.; Naqvi, K.; Daver, N.G.; Montalban-Bravo, G.; Cortes, J.E.; DiNardo, C.D.; Jabbour, E.; et al. Phase II study of azacitidine with pembrolizumab in patients with intermediate-1 or higher-risk myelodysplastic syndrome. Br. J. Haematol. 2021. [Google Scholar] [CrossRef] [PubMed]
  140. Garcia-Manero, G.; Tallman, M.S.; Martinelli, G.; Ribrag, V.; Yang, H.; Balakumaran, P.S.A.; Chlosta, S.; Zhang, Y.; Smith, B.D. Pembrolizumab, a PD-1 Inhibitor, in Patients with Myelodysplastic Syndrome (MDS) after Failure of Hypomethylating Agent Treatment. Blood 2016, 128, 345. [Google Scholar] [CrossRef]
  141. Zeidan, A.M.; Cavenagh, J.; Voso, M.T.; Taussig, D.; Tormo, M.; Boss, I.; Copeland, W.B.; Gray, V.E.; Previtali, A.; O’Connor, T.; et al. Efficacy and Safety of Azacitidine (AZA) in Combination with the Anti-PD-L1 Durvalumab (durva) for the Front-Line Treatment of Older Patients (pts) with Acute Myeloid Leukemia (AML) Who Are Unfit for Intensive Chemotherapy (IC) and Pts with Higher-Risk Myelodysplastic Syndromes (HR-MDS): Results from a Large, International, Randomized Phase 2 Study. Blood 2019, 134, 829. [Google Scholar] [CrossRef]
  142. Gerds, M.A.T.; Scott, B.L.; Greenberg, P.L.; Khaled, S.K.; Lin, T.L.; A Pollyea, D.; Verma, A.; Dail, M.; Green, C.; Ma, C.; et al. PD-L1 Blockade with Atezolizumab in Higher-Risk Myelodysplastic Syndrome: An Initial Safety and Efficacy Analysis. Blood 2018, 132, 466. [Google Scholar] [CrossRef]
  143. O’Connell, C.L.; Kropf, P.L.; Punwani, N.; Rogers, D.; Sposto, R.; Grønbæk, K. Phase I Results of a Multicenter Clinical Trial Combining Guadecitabine, a DNA Methyltransferase Inhibitor, with Atezolizumab, an Immune Checkpoint Inhibitor, in Patients with Relapsed or Refractory Myelodysplastic Syndrome or Chronic Myelomonocytic Leukemia. Blood 2018, 132, 1811. [Google Scholar] [CrossRef]
  144. Brunner, A.M.; Esteve, J.; Porkka, K.; Knapper, S.; Vey, N.; Scholl, S.; Garcia-Manero, G.; Wermke, M.; Janssen, J.; Traer, E.; et al. Efficacy and Safety of Sabatolimab (MBG453) in Combination with Hypomethylating Agents (HMAs) in Patients with Acute Myeloid Leukemia (AML) and High-Risk Myelodysplastic Syndrome (HR-MDS): Updated Results from a Phase 1b Study. Blood 2020, 136, 1–2. [Google Scholar] [CrossRef]
  145. Zeidan, M.A.M.; Esteve, J.; Giagounidis, A.; Kim, H.-J.; Miyazaki, Y.; Platzbecker, U.; Schuh, A.C.; Sekeres, M.A.; Westermann, J.; Xiao, Z.; et al. The STIMULUS Program: Clinical Trials Evaluating Sabatolimab (MBG453) Combination Therapy in Patients (Pts) with Higher-Risk Myelodysplastic Syndromes (HR-MDS) or Acute Myeloid Leukemia (AML). Blood 2020, 136, 45–46. [Google Scholar] [CrossRef]
  146. Riether, C.; Pabst, T.; Höpner, S.; Bacher, U.; Hinterbrandner, M.; Banz, Y.; Müller, R.; Manz, M.G.; Gharib, W.H.; Francisco, D.; et al. Targeting CD70 with cusatuzumab eliminates acute myeloid leukemia stem cells in patients treated with hypomethylating agents. Nat. Med. 2020, 26, 1459–1467. [Google Scholar] [CrossRef] [PubMed]
  147. Sick, E.; Jeanne, A.; Schneider, C.; Dedieu, S.; Takeda, K.; Martiny, L. CD47 update: A multifaceted actor in the tumour microenvironment of potential therapeutic interest. Br. J. Pharmacol. 2012, 167, 1415–1430. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Chao, M.P.; Takimoto, C.H.; Feng, D.D.; McKenna, K.; Gip, P.; Liu, J.; Volkmer, J.-P.; Weissman, I.L.; Majeti, R. Therapeutic Targeting of the Macrophage Immune Checkpoint CD47 in Myeloid Malignancies. Front. Oncol. 2020, 9, 1380. [Google Scholar] [CrossRef] [PubMed]
  149. Matlung, H.L.; Szilagyi, K.; Barclay, N.A.; Berg, T.K.V.D. The CD47-SIRPα signaling axis as an innate immune checkpoint in cancer. Immunol. Rev. 2017, 276, 145–164. [Google Scholar] [CrossRef] [PubMed]
  150. Majeti, R.; Chao, M.P.; Alizadeh, A.A.; Pang, W.W.; Jaiswal, S.; Gibbs, K.; Van Rooijen, N.; Weissman, I.L. CD47 Is an Adverse Prognostic Factor and Therapeutic Antibody Target on Human Acute Myeloid Leukemia Stem Cells. Cell 2009, 138, 286–299. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Zeidan, M.A.M.; DeAngelo, D.J.; Palmer, J.M.; Seet, C.S.; Tallman, M.S.; Wei, X.; Li, Y.F.; Hock, R.N.; Burgess, M.R.; Hege, K.; et al. A Phase I Study of CC-90002, a Monoclonal Antibody Targeting CD47, in Patients with Relapsed and/or Refractory (R/R) Acute Myeloid Leukemia (AML) and High-Risk Myelodysplastic Syndromes (MDS): Final Results. Blood 2019, 134, 1320. [Google Scholar] [CrossRef]
  152. A Sallman, D.; Asch, A.S.; Al Malki, M.M.; Lee, D.J.; Donnellan, W.B.; Marcucci, G.; Kambhampati, S.; Daver, N.G.; Garcia-Manero, G.; Komrokji, R.S.; et al. The First-in-Class Anti-CD47 Antibody Magrolimab (5F9) in Combination with Azacitidine Is Effective in MDS and AML Patients: Ongoing Phase 1b Results. Blood 2019, 134, 569. [Google Scholar] [CrossRef]
  153. Aggarwal, N.; Swerdlow, S.H.; TenEyck, S.P.; Boyiadzis, M.; Felgar, R.E. Natural killer cell (NK) subsets and NK-like T-cell populations in acute myeloid leukemias and myelodysplastic syndromes. Cytom. Part B Clin. Cytom. 2016, 90, 349–357. [Google Scholar] [CrossRef]
  154. Cianga, V.A.; Catafal, L.C.; Cianga, P.; Tanasa, M.P.; Cherry, M.; Collet, P.; Tavernier, E.; Guyotat, D.; Rusu, C.; Aanei, C.M. Natural Killer Cell Subpopulations and Inhibitory Receptor Dynamics in Myelodysplastic Syndromes and Acute Myeloid Leukemia. Front. Immunol. 2021, 12, 665541. [Google Scholar] [CrossRef]
  155. 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]
  156. Warlick, E.D.; Weisdorf, D.J.; Vallera, D.A.; Wangen, R.; Lewis, R.D.; Knox, J.; Schroeder, M.; Felices, M.; Miller, J.S. GTB-3550 TriKE™ for the Treatment of High-Risk Myelodysplastic Syndromes (MDS) and Refractory/Relapsed Acute Myeloid Leukemia (AML) Safely Drives Natural Killer (NK) Cell Proliferation At Initial Dose Cohorts. Blood 2020, 136, 7–8. [Google Scholar] [CrossRef]
  157. Yalniz, F.; Daver, N.; Rezvani, K.; Kornblau, S.; Ohanian, M.; Borthakur, G.; Dinardo, C.D.; Konopleva, M.; Burger, J.; Gasior, Y.; et al. A Pilot Trial of Lirilumab With or Without Azacitidine for Patients With Myelodysplastic Syndrome. Clin. Lymphoma Myeloma Leuk. 2018, 18, 658–663.e2. [Google Scholar] [CrossRef]
  158. Miyakoshi, S.; Usuki, K.; Matsumura, I.; Ueda, Y.; Iwasaki, H.; Miyamoto, T.; Origuchi, M.; Tagashira, S.; Naoi, I.; Naoe, T.; et al. Preliminary Results from a Phase 1/2 Study of DSP-7888, a Novel WT1 Peptide-Based Vaccine, in Patients with Myelodysplastic Syndrome (MDS). Blood 2016, 128, 4335. [Google Scholar] [CrossRef]
  159. Brayer, J.; Lancet, J.E.; Powers, J.; List, A.; Balducci, L.; Komrokji, R.; Pinilla-Ibarz, J. WT1 vaccination in AML and MDS: A pilot trial with synthetic analog peptides. Am. J. Hematol. 2015, 90, 602–607. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Qazilbash, M.H.; Wieder, E.; Thall, P.F.; Wang, X.; Rios, R.; Lu, S.; Kanodia, S.; Ruisaard, K.E.; Giralt, S.A.; Estey, E.H.; et al. PR1 peptide vaccine induces specific immunity with clinical responses in myeloid malignancies. Leukemia 2016, 31, 697–704. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  161. Bergmann, L.; Maurer, U.; Weidmann, E. Wilms Tumor Gene Expression in Acute Myeloid Leukemias. Leuk. Lymphoma 1997, 25, 435–443. [Google Scholar] [CrossRef]
  162. Cilloni, D.; Gottardi, E.; Messa, F.; Fava, M.; Scaravaglio, P.; Bertini, M.; Girotto, M.; Marinone, C.; Ferrero, D.; Gallamini, A.; et al. Significant Correlation Between the Degree of WT1 Expression and the International Prognostic Scoring System Score in Patients With Myelodysplastic Syndromes. J. Clin. Oncol. 2003, 21, 1988–1995. [Google Scholar] [CrossRef]
  163. Rautenberg, C.; Germing, U.; Pechtel, S.; Lamers, M.; Fischermanns, C.; Jäger, P.; Geyh, S.; Haas, R.; Kobbe, G.; Schroeder, T. Prognostic impact of peripheral blood WT1-mRNA expression in patients with MDS. Blood Cancer J. 2019, 9, 1–8. [Google Scholar] [CrossRef] [Green Version]
  164. Jo, T.; Sakai, K.; Muranushi, H.; Okamoto, Y.; Tsukamoto, T.; Sugiura, H.; Matsui, H.; Ueda, T.; Okada, K.; Maeda, T.; et al. Pre-Treatment WT1 mRNA Expression Level In Peripheral Blood Predicts Response and Overall Survival Of Myelodysplastic Syndrome Patients In The Azacitidine Era. Blood 2013, 122, 1528. [Google Scholar] [CrossRef]
  165. Rezvani, K.; Yong, A.S.; Mielke, S.; Jafarpour, B.; Savani, B.N.; Le, R.Q.; Eniafe, R.; Musse, L.; Boss, C.; Kurlander, R.; et al. Repeated PR1 and WT1 peptide vaccination in Montanide-adjuvant fails to induce sustained high-avidity, epitope-specific CD8+ T cells in myeloid malignancies. Haematologica 2010, 96, 432–440. [Google Scholar] [CrossRef]
  166. Rezvani, K.; Yong, A.S.M.; Mielke, S.; Savani, B.N.; Musse, L.; Superata, J.; Jafarpour, B.; Boss, C.; Barrett, A.J. Leukemia-associated antigen-specific T-cell responses following combined PR1 and WT1 peptide vaccination in patients with myeloid malignancies. Blood 2008, 111, 236–242. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Suzuki, T.; Ueda, Y.; Ogura, M.; Uchida, T.; Ozawa, K.; Miyakoshi, M.S.; Naoe, T.; Murata, M.; Kizaki, M.; Uike, N.; et al. A Phase 1/2 Study of WT1 Peptide Cancer Vaccine WT4869 in Patients with Myelodysplastic Syndromes (MDS). Blood 2015, 126, 2868. [Google Scholar] [CrossRef]
  168. Keilholz, U.; Letsch, A.; Busse, A.; Asemissen, A.M.; Bauer, S.; Blau, I.W.; Hofmann, W.-K.; Uharek, L.; Thiel, E.; Scheibenbogen, C. A clinical and immunologic phase 2 trial of Wilms tumor gene product 1 (WT1) peptide vaccination in patients with AML and MDS. Blood 2009, 113, 6541–6548. [Google Scholar] [CrossRef] [Green Version]
  169. Thomas, R.; Al-Khadairi, G.; Roelands, J.; Hendrickx, W.; Dermime, S.; Bedognetti, D.; Decock, J. NY-ESO-1 Based Immunotherapy of Cancer: Current Perspectives. Front. Immunol. 2018, 9, 947. [Google Scholar] [CrossRef]
  170. Atanackovic, D.; Luetkens, T.; Kloth, B.; Fuchs, G.; Cao, Y.; Hildebrandt, Y.; Meyer, S.; Bartels, K.; Reinhard, H.; Lajmi, N.; et al. Cancer-testis antigen expression and its epigenetic modulation in acute myeloid leukemia. Am. J. Hematol. 2011, 86, 918–922. [Google Scholar] [CrossRef]
  171. Srivastava, P.; Matsuzaki, J.; Paluch, B.E.; Kaufman, S.; Karpf, A.R.; Odunsi, K.; Miller, A.; Kocent, J.; Wang, E.S.; Nemeth, M.J.; et al. Vaccination with NY-ESO-1 in Combination with Decitabine for Patients with MDS. Blood 2016, 128, 4326. [Google Scholar] [CrossRef]
  172. Griffiths, E.A.; Srivastava, P.; Matsuzaki, J.; Brumberger, Z.; Wang, E.S.; Kocent, J.; Miller, A.; Roloff, G.; Wong, H.Y.; Paluch, B.E.; et al. NY-ESO-1 Vaccination in Combination with Decitabine Induces Antigen-Specific T-lymphocyte Responses in Patients with Myelodysplastic Syndrome. Clin. Cancer Res. 2017, 24, 1019–1029. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Holmberg-Thydén, S.; Dufva, M.I.H.; Gang, M.A.O.; Breinholt, M.F.; Schejbel, L.; Andersen, D.M.K.; Kadivar, M.; Svane, I.M.; Grønbæk, K.; Hadrup, S.R.; et al. Therapeutic Cancer Vaccination Targeting Shared Tumor Associated Antigens in Combination with Azacitidine for High Risk Myelodysplastic Syndrome—A Phase I Clinical Trial. Blood 2020, 136, 23–24. [Google Scholar] [CrossRef]
  174. Anguille, S.; Van de Velde, A.L.; Smits, E.L.; Van Tendeloo, V.F.; Juliusson, G.; Cools, N.; Nijs, G.; Stein, B.; Lion, E.; Van Driessche, A.; et al. Dendritic cell vaccination as postremission treatment to prevent or delay relapse in acute myeloid leukemia. Blood 2017, 130, 1713–1721. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Powers, J.; Padron, E.; Dubovsky, J.; Berchmans, E.; Farnum, T.; Bai, F.; Sahakian, E.; Ahmed, J.; Komrokji, R.S.; Lancet, J.E.; et al. Enhanced Immunological Responses Following K562/GM-CSF/CD40L Vaccine Plus Lenalidomide in High-Risk Myelodysplastic Syndrome. Blood 2011, 118, 1725. [Google Scholar] [CrossRef]
  176. Spear, P.; Wu, M.-R.; Sentman, M.-L.; Sentman, C.L. NKG2D ligands as therapeutic targets. Cancer Immun. 2013, 13, 8. [Google Scholar] [PubMed]
  177. Le Bert, N.; Gasser, S. Advances in NKG2D ligand recognition and responses by NK cells. Immunol. Cell Biol. 2014, 92, 230–236. [Google Scholar] [CrossRef]
  178. Sallman, D.A.; Al-Homsi, A.S.; Davila, M.L.; Kerre, T.; Moors, I.; Poire, X.; Havelange, V.; Lewalle, P.; Pollyea, D.A.; Wang, E.S.; et al. Results from the Phase I Clinical Studies Evaluating Cyad-01, a First-Generation NKG2D CAR T-Cell Product in Relapsed or Refractory Acute Myeloid Leukemia and Myelodysplastic Syndrome Patients. Blood 2020, 136, 40–41. [Google Scholar] [CrossRef]
  179. Bachier, C.; Borthakur, G.; Hosing, C.; Blum, W.; Rotta, M.; Ojeras, P.; Barnett, B.; Rajangam, K.; Majhail, M.N.S.; Nikiforow, S. A Phase 1 Study of NKX101, an Allogeneic CAR Natural Killer (NK) Cell Therapy, in Subjects with Relapsed/Refractory (R/R) Acute Myeloid Leukemia (AML) or Higher-Risk Myelodysplastic Syndrome (MDS). Blood 2020, 136, 42–43. [Google Scholar] [CrossRef]
  180. Sallman, D.A.; Elmariah, M.H.; Sweet, M.K.L.; Talati, C.; Mishra, A.; Kelley, L.L.; Lankford, A.; Chan, T.; Shah, R.R.; Padron, E.; et al. A Phase 1/1b Safety Study of Prgn-3006 Ultracar-T™ in Patients with Relapsed or Refractory CD33-Positive Acute Myeloid Leukemia and Higher Risk Myelodysplastic Syndrome. Blood 2020, 136, 17. [Google Scholar] [CrossRef]
  181. Stevens, B.M.; Zhang, W.; Pollyea, D.A.; Winters, A.; Gutman, J.; Smith, C.; Budde, E.; Forman, S.J.; Jordan, C.T.; Purev, E. CD123 CAR T cells for the treatment of myelodysplastic syndrome. Exp. Hematol. 2019, 74, 52–63.e3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Claiborne, J.; Bandyopathyay, D.; Roberts, C.; Hawks, K.; Aziz, M.; Simmons, G.; Wiedl, C.; Chung, H.; Clark, W.; Mccarty, J.; et al. Managing post allograft relapse of myeloid neoplasms: Azacitidine and donor lymphocyte infusions as salvage therapy. Leuk. Lymphoma 2019, 60, 2733–2743. [Google Scholar] [CrossRef] [PubMed]
  183. Krishnamurthy, P.; Potter, V.T.; Barber, L.D.; Kulasekararaj, A.G.; Lim, Z.Y.; Pearce, R.M.; De Lavallade, H.; Kenyon, M.; Ireland, R.M.; Marsh, J.C.; et al. Outcome of Donor Lymphocyte Infusion after T Cell–depleted Allogeneic Hematopoietic Stem Cell Transplantation for Acute Myelogenous Leukemia and Myelodysplastic Syndromes. Biol. Blood Marrow Transplant. 2013, 19, 562–568. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. From immune dysregulations to therapeutic perspectives in myelodysplastic syndromes. (A) Immune key hubs involved in early stages, low-risk MDS, and high-risk MDS. (B) Available and potential drugs targeting immune system in MDS. CD: Cluster of Differentiation; CTLA-4: Cytotoxic T-Lymphocyte Antigen 4; DAMPs: Damage Associated Molecular Patterns; Del5q: Deletion 5q; DNMT3A: DNA methyl-transferase 3A; Gal9: Galectin 9; HR-MDS: High-risk myelodysplastic syndrome; IL: Interleukin; LR-MDS: Low-risk myelodysplastic syndrome; M1/M2: Macrophages type 1 and 2; MDSC: Myeloid-Derived Suppressor Cells; NKG2D: Natural killer group 2 member; NLRP3: NOD-like receptor family, pyrin domain containing 3; PD-1: Program cell death-1; PD-L1: Program cell death-Ligand 1; RANTES: Regulated upon Activation, Normal T Cell Expressed and Presumably Secreted; SF3B1: Splicing Factor 3b Subunit 1; SRSF2: Serine and Arginine Rich Splicing Factor 2; TET2: Ten-eleven-translocation 2; TIGIT: T cell immunoreceptor with Ig and ITIM domains; TIM3: T-cell immunoglobulin and mucin containing protein-3; TGF-β: Transforming Growth Factor-β; TLR: Toll Like Receptor; TNF-α: Tumor Necrosis Factor-α; TNFR: Tumor Necrosis Factor Receptor; TP53: Tumor Protein 53; Treg: Lymphocytes T regulators; WT1: Wilms Tumor 1.
Figure 1. From immune dysregulations to therapeutic perspectives in myelodysplastic syndromes. (A) Immune key hubs involved in early stages, low-risk MDS, and high-risk MDS. (B) Available and potential drugs targeting immune system in MDS. CD: Cluster of Differentiation; CTLA-4: Cytotoxic T-Lymphocyte Antigen 4; DAMPs: Damage Associated Molecular Patterns; Del5q: Deletion 5q; DNMT3A: DNA methyl-transferase 3A; Gal9: Galectin 9; HR-MDS: High-risk myelodysplastic syndrome; IL: Interleukin; LR-MDS: Low-risk myelodysplastic syndrome; M1/M2: Macrophages type 1 and 2; MDSC: Myeloid-Derived Suppressor Cells; NKG2D: Natural killer group 2 member; NLRP3: NOD-like receptor family, pyrin domain containing 3; PD-1: Program cell death-1; PD-L1: Program cell death-Ligand 1; RANTES: Regulated upon Activation, Normal T Cell Expressed and Presumably Secreted; SF3B1: Splicing Factor 3b Subunit 1; SRSF2: Serine and Arginine Rich Splicing Factor 2; TET2: Ten-eleven-translocation 2; TIGIT: T cell immunoreceptor with Ig and ITIM domains; TIM3: T-cell immunoglobulin and mucin containing protein-3; TGF-β: Transforming Growth Factor-β; TLR: Toll Like Receptor; TNF-α: Tumor Necrosis Factor-α; TNFR: Tumor Necrosis Factor Receptor; TP53: Tumor Protein 53; Treg: Lymphocytes T regulators; WT1: Wilms Tumor 1.
Diagnostics 11 01982 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Comont, T.; Treiner, E.; Vergez, F. From Immune Dysregulations to Therapeutic Perspectives in Myelodysplastic Syndromes: A Review. Diagnostics 2021, 11, 1982. https://doi.org/10.3390/diagnostics11111982

AMA Style

Comont T, Treiner E, Vergez F. From Immune Dysregulations to Therapeutic Perspectives in Myelodysplastic Syndromes: A Review. Diagnostics. 2021; 11(11):1982. https://doi.org/10.3390/diagnostics11111982

Chicago/Turabian Style

Comont, Thibault, Emmanuel Treiner, and François Vergez. 2021. "From Immune Dysregulations to Therapeutic Perspectives in Myelodysplastic Syndromes: A Review" Diagnostics 11, no. 11: 1982. https://doi.org/10.3390/diagnostics11111982

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