Skip to main content
Cancer Medicine logoLink to Cancer Medicine
. 2024 Sep 10;13(17):e70152. doi: 10.1002/cam4.70152

Immune‐dysregulation harnessing in myeloid neoplasms

Mohammad Jafar Sharifi 1, Ling Xu 2, Nahid Nasiri 1, Mehnoosh Ashja‐Arvan 3, Hadis Soleimanzadeh 1, Mazdak Ganjalikhani‐Hakemi 3,4,
PMCID: PMC11386321  PMID: 39254117

Abstract

Myeloid malignancies arise in bone marrow microenvironments and shape these microenvironments in favor of malignant development. Immune suppression is one of the most important stages in myeloid leukemia progression. Leukemic clone expansion and immune dysregulation occur simultaneously in bone marrow microenvironments. Complex interactions emerge between normal immune system elements and leukemic clones in the bone marrow. In recent years, researchers have identified several of these pathological interactions. For instance, recent works shows that the secretion of inflammatory cytokines such as tumor necrosis factor‐α (TNF‐α), from bone marrow stromal cells contributes to immune dysregulation and the selective proliferation of JAK2V617F+ clones in myeloproliferative neoplasms. Moreover, inflammasome activation and sterile inflammation result in inflamed microenvironments and the development of myelodysplastic syndromes. Additional immune dysregulations, such as exhaustion of T and NK cells, an increase in regulatory T cells, and impairments in antigen presentation are common findings in myeloid malignancies. In this review, we discuss the role of altered bone marrow microenvironments in the induction of immune dysregulations that accompany myeloid malignancies. We also consider both current and novel therapeutic strategies to restore normal immune system function in the context of myeloid malignancies.

Keywords: acute myeloid leukemia, myelodysplastic syndrome, myeloproliferative disorders, tumor‐infiltrating immune cells

1. INTRODUCTION

The bone marrow microenvironment plays a crucial role in normal hematopoiesis and the development and progression of myeloid malignancies. Normal bone marrow niches are regulated by the interaction of various factors, such as stromal mesenchymal cells and their progeny, the vascular network, nerve endings, mature blood cells, extracellular matrix proteins, and bone cells. The main function of the bone marrow in a healthy person is to provide a nurturing environment and to help produce blood cells. 1 In neoplastic myeloid diseases including acute myeloid leukemia (AML), myelodysplastic syndromes (MDS), and myeloproliferative neoplasms (MPNs), the functions of the immune system become dysregulated. Disruptions in the bone marrow microenvironment lead to uncontrolled clonal expansion of malignant cells. The molecular mechanisms underlying changes in the bone marrow environment and their effects on therapeutic approaches are not yet fully understood. 1 , 2 Interactions between stromal cells, immune cells, and neoplastic myeloid cells are dynamic and complex. Stromal cells such as fibroblasts and mesenchymal stromal cells secrete cytokines and other molecules to inhibit the anti‐tumor activity of the immune system, while antigen‐presenting cells, like dendritic cells (DCs) and macrophages promote malignant cells' growth and survival. 1 Recently, several studies have focused on the link between chronic inflammation, aging, clonal hematopoiesis, and the development of hematological malignancies in the bone marrow microenvironment. 3 , 4 , 5 , 6 There is also significant mutational and phenotypic variability in myeloid malignancies. 4 Therefore, it is difficult to make correct decisions regarding immunotherapy for myeloid malignancies. Despite these limitations, therapies targeting the immune‐dysregulated bone marrow microenvironment have been explored in recent years as promising methods for treating myeloid malignancies. Some immune‐targeting therapies, such as immune checkpoint inhibitors (CIs) and cytokines, may help restore immune system function to recognize and eliminate cancer cells. Targeting specific molecules involved in the development and progression of leukemic cells, such as FLT3 and epigenetic machinery, is effective in the treatment of myeloid malignancies due to their anti‐neoplastic and immunomodulatory effects. 7 Potential passive immunotherapies for myeloid malignancies include tumor‐specific antibodies, cytokines, and adaptive cell transfer, while active immunotherapies include dendritic cell vaccines, allogeneic whole‐cell vaccines, checkpoint inhibitors, and oncolytic viruses, among others. 5 This review aims to discuss and highlight the current information regarding the molecular mechanisms of alterations in the immunological bone marrow microenvironment in myeloid malignancies, as well as to provide insights into potential immunotherapies. A summary of main immune dysregulations and potential therapeutics is depicted in Figure 1.

FIGURE 1.

FIGURE 1

Myeloid malignancies are associated with key changes in the bone marrow immune microenvironment. By identifying more of these abnormalities, special treatments can be used to correct them and ultimately treat myeloid malignancies. The figure was made with the support of Dienst Medical Art (https://smart.servier.com). Ag, Antigen; HMAs, Hypomethylating agents; IDO; Indoleamine‐pyrrole 2,3‐dioxygenase.

2. IMMUNOLOGICAL TUMOR MICROENVIRONMENT IN MYELOID MALIGNANCIES, AN OVERVIEW

Although tumor immunology primarily focuses on the local immune responses within the tumor microenvironment (TME), it is important to acknowledge that these responses rely on continuous communication with peripheral organs. The bone marrow, blood, spleen, and draining lymph nodes together form an integrated immunological network that is in constant communication during the development of a myeloid malignancy. 5 The progression of leukemia is influenced not only by the neoplastic cell compartment but also by the immunological milieu present within the TME. 8 Interactions between bone marrow microenvironment and leukemia cells through a combination of adhesion molecules and soluble factors, play a crucial role in the development of myeloid malignancies. The immunological tumor microenvironment is well characterized for solid tumors but has recently gained attention for myeloid neoplasms. One of the major obstacles in conducting such studies in myeloid malignancies is that the immune cells themselves are part of the hematopoietic tissue and normal microenvironmental components of the bone marrow. However, due to the availability of powerful techniques such as multiplex immunohistochemistry, extensive studies have recently been conducted in this area. 9 , 10 The anticancer immune landscape consists of various elements. Innate immune cells, such as T cell subsets expressing γδ‐TCR, NK cells, myeloid suppressor cells, and phagocytes, are involved in the defense against leukemia. A consistent immune response to myeloid malignancies may be lacking due to factors like a low burden of malignant cell antigens, defects in antigen presentation, an increase in the number and activity of myeloid derived suppressor cells (MDSC), suppressive macrophages, an imbalance effector T cells, and regulatory T cell (Treg) (Treg cell dominance), T cell depletion caused by chronic inflammation, upregulation of immune checkpoint ligands and receptors, and production of immunosuppressive agents. 4 , 5 , 8 , 11 , 12 Interestingly, in myeloid neoplasms there is a decrease in the cytolytic activity of lymphocytes of the innate immune system compared to lymphoproliferative disorders. 11 Another important aspect of immune landscape in myeloid malignancies is the lowest tumor mutational burden among other neoplasms, which makes them unresponsive to PD‐L1 blockade immunotherapy. 13 Advances in understanding the complex immune interactions associated with AML will support the development and application of personalized immunotherapeutic approaches. 14 Recently, it has become clear that inflammation plays a major role in the development of myeloid malignancies, especially chronic types. Because of its vital role in the development of cancers, inflammation is now recognized as a hallmark of cancer. 15 A clear example of the association between auto‐inflammation and myeloid neoplasia was recently described with the diagnosis of VEXAS syndrome. 16 Each driver mutation in myeloid malignancies creates a specific inflammatory profile. 17 Published studies have shown that certain inflammatory cytokines, such as TNFα and interferon‐α, play a selective role in the clonal expansion of myeloid neoplasms. 8 These inflammatory cytokines activate positive feedback loops and transforming normal bone marrow niches into inflammatory microenvironments. 17 The pathophysiological roles of the pro‐inflammatory bone marrow microenvironment in the development of myeloid malignancies are summarized in Figure 2. The role of a defective immune system and an abnormal immune microenvironment in the development and progression of myeloid malignancies is discussed in more detail in the following sections.

FIGURE 2.

FIGURE 2

Current scheme of the role of the bone marrow inflammatory environment in the progression of myeloid malignancies. In MDS syndromes, inflammatory signals lead to a type of specific cell death called pyroptosis. Pyroptotic cells showed characteristic dysplastic changes associated with MDS neoplasia. In the presence of somatic mutations such as JAK2V617F, MPL, and CALR, an inflammatory microenvironment favors the development of myeloproliferative neoplasms. Inflammatory niches also lead to exhausted T and NK cells. In such situations, leukemic stem cells escaped immune surveillance and evolved into more aggressive AMLs. The figure was made with the support of Dienst Medical Art (https://smart.servier.com).

3. ACUTE MYELOID LEUKEMIA (AML)

Acute myeloid leukemia (AML) is a diverse group of acute hematologic malignancies derived from the myeloid lineage. The pathogenesis of AML involves various genetic alterations, including recurrent chromosomal structural variations and gene mutations, which typically determine the risk stratification of AML. Additionally, in recent years, epigenetic regulation and immune suppression have also been identified as playing roles in the pathogenesis of AML. 7 , 18 , 19 , 20 , 21 Understanding the pathogenesis of AML is crucial for developing new treatment approaches and improving the accuracy of risk classification. Here, we focus on summarizing the immune suppression alterations that exist in the bone marrow microenvironment of AML, particularly focusing on alterations of T cells. We also review the immunotherapy approaches that have been utilized or show promise for future use.

3.1. Immune suppression and alterations in AMLs

Abundant studies have proven that the immune‐suppressive microenvironment has a significant impact on the development and prognosis of AML. CD8 T cells are at the center of anti‐tumor immunity, while γδ T cells, natural killer cells (NK), and other invariant NK T cells also play important roles in killing tumor cells. Numerical and functional abnormalities of these immune effector cells have been reported in the AML microenvironment. Furthermore, other abnormalities in regulatory immune cells such as regulatory Tregs, MDSCs, regulatory macrophage cells (Mφs), DCs, and MSCs also contribute to the construction of the immune suppressive microenvironment. 21

The anti‐tumor function of NK cells is regulated by a balance of inhibitory and activating cell surface receptors. Multiple studies have found that AML patients often have low expression of activating receptors, like NK group 2D (NKG2D), natural cytotoxicity receptors (NCRs), and DNAX accessory molecule‐1 (DNAM‐1), while showing overexpression of inhibitory receptors, such as KIR2DL2/L3 and natural killer group 2A (NKG2A). 22 , 23 , 24 In addition to the classic MHC‐I specific inhibitory receptors, other non‐MHC specific inhibitory receptors, like T cell immunoglobulin and ITIM domain (TIGIT), killer cell lectin‐like receptor (KLRG1), and programmed cell death protein 1 (PD‐1) are also over‐expressed on NK cells from AML patients. 25 , 26 , 27 Additionally, the bone marrow microenvironment could hinder the cytotoxicity and proliferation of NK cells in AML through the release of immune‐suppressing cytokines, such as IL‐10, TGF‐β, IDO, and prostaglandin‐E2 (PGE2). These cytokines are secreted by leukemia blasts, MDSCs, and other cells. 22 , 28 , 29 , 30

MDSCs can suppress effector T cells through the release of arginase‐1 (Arg1), ROS, nitric oxide synthase 2 (NOS2), cyclooxygenase 2 (COX2), IL10, transforming growth factor beta (TGF‐β), and other cytokines. They can also mediate immunosuppression by upregulating Tregs. There is evidence showing that MDSCs accumulate in the peripheral blood (PB) and bone marrow of de novo AML patients. 29 , 30 , 31 , 32 , 33 According to the literature, possible mechanisms related to MDSC‐mediated immune suppression in AML include high expression of Arg1, IDO, and V‐domain Ig suppressor of T cell activation (VISTA). 30 , 34

Macrophages (Mφs) are derived directly from monocytes under normal or inflammatory conditions. 35 They can be broadly classified into two phenotypes: M1 (inflammatory) and M2 (regulatory). In blood cancers, leukemia associated macrophages (LAMs) are educated by the neoplastic bone marrow microenvironment to obtain leukemia‐supporting phenotype and mediate the progression of disease. 21 , 36 , 37 The percentage of CD206+ M2‐like macrophages in the bone marrow was significantly elevated, and the level of infiltration of M2‐like macrophages positively correlated with poor outcomes for AML patients. 38 , 39 The molecular mechanisms related to the leukemia supporting phenotype polarization of Mφs in AML include low expression of the monocytic leukemia zinc finger (MOZ) and expression of Growth factor independence 1 (Gfi1). 39 , 40

3.2. T‐cell alterations in AML

Both αβ and γδ T cells can eliminate tumor cells, and their accumulation in the tumor microenvironment is usually correlated with better overall survival. 41 One research that recruited 66 AML patients reported that high T cell percentages (>78.5% of total lymphocytes) in the bone marrow led to increased overall survival and leukemia‐free survival. 42 Additionally, as reported by Ravandi et al., 43 a higher prevalence of CD3‐positive T cells in the bone marrow before the administration of therapy appeared to predict response to nivolumab in combination with induction treatment. In addition to changes in cell counts, various types of T cell dysregulation also occur in AML. These include impaired synapse formation, exhaustion, senescence, metabolic disturbances, and epigenetic changes. Immune synapse formation is the first step in T cell activation. However, in AML, the ability to form immune synapses and recruit phosphotyrosine signaling molecules is significantly impaired. 44 In addition, the upregulation of immune check‐points (ICs), such as PD‐1, TIM‐3, CTLA‐4, LAG‐3, and TIGIT on CD8 T cells has been identified in de novo AML and relapsed and refractory AML (R/R AML). 45 , 46 , 47 , 48 , 49 In relation to the cytotoxic cytokine secretion, Schnorfeil et al. 50 found there were no functional defects in AML T cells. Meanwhile, Knaus et al. 48 found that granzyme‐B expressing CD8 T cells were higher in de novo AML patients compared to the healthy control group. Additionally, Tang et al. 12 have reported that the production of IFN‐γ by CD8 T cells from AML patients is impaired, but there was no reduction in IL‐2 and TNF‐α secretion. These different results may reflect the use of different stimulating agents for each experiment and the heterogeneous patient cohorts. Recently, several other T cell suppressive receptors expressed by AML blasts that inhibit T cell proliferation and function have been identified. These include leukocyte immunoglobulin‐like receptor B4 (LILRB4) and CD200. LILRB4 is an inhibitory immune checkpoint receptor that is selectively expressed on monocytic leukemic cells. It mediates the release of arginase‐1 (Arg1), which directly suppresses T cell proliferation and cytotoxicity. 51 , 52 CD200 is a type‐1a transmembrane cell‐surface glycoprotein. Coles et al. 53 , 54 , 55 discovered that CD200 may inhibit T cell function by binding with CD200R expressed by T cells or by promoting Treg formation.

T cell senescence is a dysfunctional state characterized by the downregulation of costimulatory molecules CD27 and CD28, expression of senescence‐associated surface markers B3GAT1 (CD57) and KLRG1, an active metabolic state, and continuous secretion of pro‐inflammatory cytokines. Sergio Rutella et al. 56 found that pre‐existing and chemotherapy‐induced senescent‐like T cells were correlated with a poor outcome in AML patients.

Tregs have been found to suppress the anti‐tumor function of T cells through direct cell‐to‐cell contact and secretion of inhibitory cytokines. Several studies have reported an increased number of Tregs in the peripheral blood and bone marrow of AML patients. 57 , 58 , 59 , 60 , 61 Shenghui et al. 58 also found that the accumulation of Tregs in the bone marrow is more significant than in the peripheral blood. These findings suggest that immune therapies targeting Tregs removal may enhance the remission rates of chemotherapy.

3.3. Immunotherapy Approaches in AML

Cytotoxic T cell plays a central role in the immune response against leukemia blast. Therefore, the targeted restoration of the T cell activity and adoptive transfer of gene‐manipulated T cells are generally considered for developing immunotherapeutic approaches.

Blocking CTLA‐4, TIM‐3, and PD‐1, or their ligands PD‐L1/PD‐L2, either alone or in combination with hypomethylating agents (HMAs) or chemotherapy agents, has been tested or is currently being tested in refractory/relapsed AML patients and those who have entered the remission phase but are at high risk of relapse. Some clinical trials have shown encouraging results, but more data from clinical trials are still needed to further explore which patient groups might benefit from ICIs. 62 , 63 , 64 , 65 , 66

The only approved monoclonal antibody for treatment of AML is anti‐CD33 (gemtuzumab ozogamicin). Refractory/relapsed AML has a poor outcome. Gemtuzumab could induce a remission rate of 33% in these patients. 67 New results demonstrate a significant decrease in the risk of relapse with gemtuzumab therapy in NPM1‐mutated AMLs compared to standard treatment (HR 0.65; 0.49–0.86; p = 0.0028). 68 The primary results of anti‐CD70 monoclonal antibody therapy in elderly patients with AML are also encouraging. A single dose of cusatuzumab monotherapy followed by a combination therapy with HMA resulted in 10 out of 12 patients achieving complete remission, while 2 had partial remission. 69

In vitro expansion of leukemia antigen‐specific T cells may have great potential for eliminating leukemia cells. However, difficulties in identifying and isolating of leukemia‐specific T cells have led to the development of antigen‐specific T cell receptor‐engineered T cells (TCR‐T) and chimeric antigen receptor T cells (CAR‐T). Unlike B‐cell lymphoma, AML has fewer surface markers on leukemia blasts that can be safely used. CD33 is expressed in up to 90% of AML blast cells but not in early pluripotent CD34+ hematopoietic stem cells, making it an attractive target. Several clinical trials using anti‐CD33 CAR‐T cells are currently ongoing. However, to date, only one study has reported on a patient with refractory AML who received autologous CAR‐T cell infusion. This patient experienced transient reduction in bone marrow blast cells along with cytokine release syndrome. 70 CD123 is an ideal CAR‐T target for AML. One clinical trial involving six patients with refractory AML following allogeneic hematopoietic stem cell transplantation (HSCT) showed that after treatment with anti‐CD123 CAR‐T cells, one patient achieved a morphologic leukemic‐free state that lasted 2 months, two patients achieved complete remission (CR), and then proceeded to a second allo‐HSCT. 71 In 2019, Yao et al. 72 reported the achievement of CR with incomplete hematologic recovery in a patient who received a single infusion of donor‐derived CD123 CAR‐T cells as part of conditioning for haploidentical HSCT. While encouraging results have been shown, the myelotoxicity of anti‐CD33 and anti‐CD123 CAR‐T cells should be avoided due to their expression on healthy myeloid and myeloid progenitor cells. Other promising targets for AML CAR‐T cell development include Lewis Y (LeY), FLT3, CLL1, CD44v6, Folate Receptor β (FRβ), CD38, and CD7. 63 , 73 , 74 TCR‐T is an approach to lyse tumor cells by genetically engineering T cells with a TCR that specifically recognize a leukemia antigen and MHC complex. Wilms “tumor 1” (WT1) is an overexpressed antigen in AML and MDS, with limited expression on normal CD34+ HSCs. Two clinical studies recruited patients with R/R AML to assess the efficacy of WT1‐TCR‐T. They found that the transferred T cells were well tolerated with minimal on‐target, off‐tumor toxicity. There were signs of anti‐leukemia activity, but no survival advantage was demonstrated. 75 , 76 Except for WT1, alternative intracellular antigens preferentially expressed by AML, such as PRAME, 77 telomerase, 78 HMMR/Rhamm, 79 and the mutated form of NPM1 80 have shown promise in clinical trials or preclinical research with engineered TCR‐T cells. Another approach is the use of bispecific antibodies to enhance the function of the patient's own T cells and overcome spatial barriers between effector T lymphocytes and leukemic blasts in the bone marrow. Bilantumomab is the first bispecific T cell engager (BiTE) immunotherapy that activates endogenous T cells by binding CD19 on acute B Lymphoblastic Leukemia (B‐ALL) cells and CD3 on T‐cells. This interaction leads to the formation of synapses between T cells and tumor cells, and ultimately resulting in the elimination of CD19+ cells. The success of bilantumomab spurred the development of various bispecific antibody constructs that link T cells to myeloid antigens in order to treat AML. CD33, which is widely expressed in AML leukemic stem and blast cells, has been particularly popular for constructing bispecific antibodies with CD3. There have been five different CD33‐CD3 bispecific antibodies that have been produced so far: AMG 330, AMG 673, AMV564, JNJ‐67571244, and GEM333. Each antibody has a unique construct aimed at extending its half‐life and increasing antigen affinity. In addition to CD33, other target antigens of interest in AML include CD123, CD47, CD70, FLT3, CLEC12A and CLL‐1. 81 , 82 Some of these targets are still under clinical trial investigation, while others have only been approved in preclinical studies. New results are expected in the future.

Several preclinical studies and clinical trials have provided insights into NK cell‐based immunotherapies, including adoptively transferred NK cells (CAR‐modified and cytokine‐induced), ICIs, and BiKE/tri‐specific killer cell engager (TriKE). GTB‐3550 TriKE is a cytokine immune engager that binds to CD16 on NK cells, CD33 on myeloid blasts and Interleukin 15 (IL‐15) between the two engager components. IL‐15 provides a self‐sustaining signal that activates NK cells and enhances their killing ability. In April 2021, Biopharma, Inc. launched the first‐in‐human GTB‐3550 TriKE Phase I/II clinical trial (NCT03214666) for the treatment of high‐risk myelodysplastic syndromes (MDS) and R/R AML. They reported a up to 63.7% reduction in bone marrow blast levels, resulting in clinical benefit, with no signs of cytokine release syndrome (CRS) or other observed toxicities. 83 More detailed discussion of NK‐based immune therapy for AML has been reviewed by Xu et al. 84

4. MYELODYSPLASTIC SYNDROMES (MDS)

4.1. MDS is an inflammatory disorder

Myelodysplastic syndromes are a diverse group of clonal stem cell malignancies characterized by increased apoptosis, ineffective hematopoiesis, resulting peripheral cytopenia, and an increased risk of transformation to AML. 85 A thorough understanding of the pathogenesis of the hallmark characteristics of MDS has emerged from recognition of the role of reciprocal interactions of innate immune, cell‐intrinsic genetic alterations, and pro‐inflammatory signaling. The damage‐associated molecular pattern (DAMP) proteins S100A8 and S100A9 dimerize to create calprotectin, which directs a sterile inflammatory cell death called pyroptosis through autocrine and paracrine interactions. 86 S100A9 leads to the expansion of mesenchymal niche cells and MDSCs. Additionally, cell‐intrinsic events directly stimulate S100A9 overexpression, providing a mechanism to induce pyroptotic cell death. 87 Indeed, the extraordinary medullary expansion of MDSCs and pyroptosis are fundamental players in the disease that could be exploited therapeutically against MDS.

4.2. Pyroptotic cell death and innate immune system activation in MDS

Inflammasome‐directed pyroptosis is the basis for MDS phenotypes like macrocytosis, proliferation, and ineffective hematopoiesis. 88 Multiple molecular mechanisms are involved in the precise regulation of inflammasome activation. Unlike apoptosis, noninflammatory caspase‐3 is responsible for mediating cell death. This process is initiated by multi‐portion inflammasome networks triggered by S100A8/A9 (alarmins or cytosolic DAMPS). Activation of TLR4 by key soluble mediators, leads to the increased expression of pro‐inflammatory cytokines like pro‐IL‐18, pro‐IL‐1β, and NLRP3. 88 The interaction between NLRP3 and apoptosis‐associated speck‐like protein containing a caspase recruitment domain (ASC) following activation stimulates ASC polymerization (ASC specks). ASC specks act as a platform for the recruitment of pro‐caspase‐1 (through its CARD domain), which then undergoes autocatalytic cleavage. Active caspase‐1 produces IL‐1β and IL‐18 from their precursors, as well as activates the pore‐forming protein gasdermin D (GSDMD). GSDMD is a nonselective membrane pore‐forming protein involved in the release of ROS, IL‐18, IL‐1β, cations, and initiation of pyroptotic cell death. 89 , 90 The NLRP3 inflammasome activates pyroptosis‐mediated lytic cell death in the bone marrow, leading to ineffective hematopoiesis, death of healthy hematopoietic stem and progenitor cells (HSPCs), cytopenias, and β‐catenin‐induced proliferation of leukemic cells. 88 This complex also contributes to hematopoiesis regulation. 91 , 92 The activation of pyroptosis by reciprocal interactions of cell‐extrinsic signals from alarmins and cell‐intrinsic genetic events, such as somatic gene mutations, is now recognized.

MDSCs are derived from bone marrow precursors usually as a result of perturbed myelopoiesis caused by various pathologies including a variety of cytokines and other molecules. In the BONE MARROW of patients with MDS, there is an accumulation of more MDSCs (Lin − HLA – DR – CD33+), which are the main effectors in the progression of cytopenia, accumulated. 93 The binding of S100A8/A9 to the CD33 receptor promotes MDSCs, inducing further secretion of S100A8/S100A9, mobilization of granzyme‐containing granules, and production of immunosuppressive cytokines such as TGF‐β and IL‐10 to reduce effector T‐cell proliferation. 93 MDSC‐derived S100A8/9 leads to autocrine and paracrine stimulation of pro‐inflammatory signals and further suppression of hematopoiesis and the innate immune system. 90 Additionally, MDSCs have gained attention due to their association with poor prognosis resistance to chemotherapy and immunotherapy. 94

High‐Mobility Group Box 1 (HMGB1), a ubiquitously expressed non‐histone DNA‐binding protein, shuttles between the nucleus and cytoplasm in nearly all eukaryotic cells. Upon cell activation or death, HMGB1 is released into the extracellular space, where it acts as an alarmin or damage‐associated molecular pattern (DAMP). Extracellular HMGB1 can activate various TLRs and is a confirmed activator of NLRP3, causing inflammation and pyroptosis. 95 Recently, its role in limiting erythropoiesis in inflammatory‐mediated and sepsis‐mediated anemia of chronic disease has been elucidated. 96 , 97

4.3. Somatic mutations and inflammatory responses in MDS

Studies on the pathogenesis of 5q deletion (del(5q)) MDS have shown that genetic modifications can directly activate signals from the innate immune system signals. Haploinsufficiency of MiR‐145 and MiR‐146 in del(5q) leads to several consequences, such as overexpression of TRAF6 and IRAK1 and haploinsufficiency of TRAF‐interacting protein with forkhead‐associated domain B (TIFA). 98

Somatic mutations affect both clonal selection and the immune microenvironment. Mutations in genes that modify the epigenetics (DNMT3A, TET2, ASXL1, and EZH2) or RNA splicing (SF3B1, SRSF2, and U2AF1) have been shown to increase pyroptotic fractions, pore formation, inflammation, and induction. 17 These mutations also activate β‐catenin signaling and its target genes, which are downregulated by suppression of the NLRP3 inflammasome or NADPH‐derived ROS. 90 As previously mentioned, somatic mutations in epigenetic modifiers create an inflammatory environment that enhances apoptosis. TET2 mutations reduce the need for histone deacetylase 2 (HDAC2), which is associated with high levels of IL‐6. 99 , 100 DNMT3A mutation is more strongly correlated with increased IFN‐γ or TNF‐α through induction of HDAC9 expression. 101 ASXL1 mutations lead to increased NADPH oxidase, ROS, TLR4, and pyroptosis. 102

Somatic mutations in the spliceosome compartments have been associated with innate immune dysregulation and inflammasome activation. Mutations in both SF3B1 and SRSF2 are linked to the hyperactivation of NF‐kB through downregulation of MAP3K7 and generation of a caspase‐8 isoform, respectively. Additionally, SRSF2 mutations result in elevated S100A8 and S100A9 alarmins. 103 , 104 , 105 , 106 Other somatic mutations, not involved in clonal hematopoiesis and driver mutations, lead to direct inflammatory manifestations and MDS. 90 VEXAS syndrome (Vacuoles, E1 enzyme, X‐linked, Autoinflammatory, Somatic) is an adult‐onset systemic auto‐inflammatory disease associated with thrombosis that typically occurs in fifth to seventh decade of life. 16 This syndrome results from somatic mutations in the gene UBA1, an X‐chromosome gene that provides instructions for making the ubiquitin‐like modifier‐activating enzyme 1. An increased risk of MDS has been reported in patients with VEXAS. 16 , 107 These data indicate that patients have an elevated risk for the developing myeloid and plasma cell neoplasms as well as symptoms of auto‐inflammatory diseases, and require monitoring for disease progression. The exact role of UBA1 somatic mutations in the induction of inflammatory flares should be clarified.

4.4. Therapeutic implications of targeting dysregulated immune pathways in MDS

To date, approved therapeutic interventions for MDS include lenalidomide for patients with transfusion‐dependent anemia due to lower‐risk MDS associated with del(5q) and hypomethylating agents (azacitidine and decitabine) for higher‐risk MDS. 90 , 108 The use of valid immune signatures for routine clinical research is expected to improve disease classification and patient outcomes. 109 Given the importance of innate immune signaling and subsequent NLRP3 inflammasome activation in disease pathophysiology, the players involved in these pathways can be used as excellent pharmacological targets for future clinical practice. 108 Novel therapeutic targeting of inflammatory/innate immune pathways in MDS including TLR signaling inhibitors, MDSC elimination, NLRP3 inflammasome inhibitors, IL‐1β inhibitors, Cas inhibitors, Wnt/β‐catenin antibodies, and PD‐1/PD‐L1 ICIs has been developed and has shown preliminary efficiency. 110 Results of the phase Ib trial of the anti‐Tim3 (sabatolimab) combination with HMAs in 51 patients with high‐risk/very high‐risk MDS show an OS rate of 56.9%. 111 This treatment protocol revealed a longer duration of clinical response (17.1 months) compared with HMAs alone treatments (10–15 months). Some clinical trials targeting inflammatory pathways are summarized in Table 1. Diagnosis of MDS could be challenging due to lack of diagnostic biomarkers in all cases. Pyroptosis and abnormal inflammatory responses can be harnessed as diagnostic biomarkers for MDS. 112 These issues have been addressed by our published and ongoing works. 113 , 114

TABLE 1.

Ongoing trials on innate inflammation pathway inhibitors in MDS.

Target biomolecules/ Therapeutic agent Mechanism of action Clinical trial phase/identifier Disease state
TLR2/Tomaralimab Anti‐TLR2 monoclonal antibody Phase 2 completed/NCT02363491 Low‐risk MDS
TLR4/CX‐01 Disrupting the HMGB1 interaction with TLR4 Phase 1 completed/NCT02995655 High‐risk MDS
IRAK4/Emavusertib Novel oral inhibitor of IRAK4 and FLT3 Phase 2 recruiting/NCT04278768 High‐risk MDS
NF‐kB/Bortezomib Inhibition of NF‐κB activity by blocking proteasomal degradation of inhibitor of κBα (IκBα) Phase 1 completed/NCT00580242 High‐risk MDS
NLRP3/Ibrutinib Indirect inhibition of ASC Phase 1 recruiting/NCT0255394, NCT03359460 High‐risk MDS
IL‐1/Canakinumab Anti‐IL‐1 monoclonal antibody Phase 2 recruiting/NCT0423915 Low‐risk MDS

Abbreviation: HMGB1, High‐mobility group box protein 1.

Immunosuppressive therapies, especially hourse anti‐thymocyte glubolin, have shown promising results in half of low‐risk MDS patients in a large international retrospective cohort study (involving 207 patients). 115 The effectiveness of such treatments has already been proven in a small prospective study and a case report. 116 , 117

5. MYELOPROLIFERATIVE NEOPLASMS (MPNs)

5.1. Abnormal signaling leads to clonal myeloproliferation

According to the latest update from WHO classification, the main categories of MPNs are chronic myeloid leukemia (CML), polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF). 118 Common driver mutations that cause BCR‐ABL1‐negative MPNs are JAK2V617F, CALR, and MPL. 118 The Janus kinase 2 gene (JAK2) is located on chromosome 9p24, and the JAK2V617F mutation occurs within its JH2 pseudokinase domain. Under normal circumstances, this domain has an inhibitory function that keeps the JAK2 protein in an inactive state. Ligand binding to cytokine receptors results in tyrosine phosphorylation of intracellular domains and auto‐phosphorylation of the JAK2 protein. This pathway leads to the activation of signaling mediators such as STATs (STAT5, STAT3, STAT1), ERK/MAP kinase and the PI3K/AKT/mTOR axis. 119 The aberrant activity of signaling pathways following the JAK2V617F mutation disrupts the regulation of multiple transcriptional targets and normal cellular functions. These include the stimulation of proliferation and disruption of apoptosis in malignant clones by PIM kinases, impaired apoptosis, cytokine‐independent maturation, erythropoietin‐independent colony formation by BCL‐XL, and increased intracellular reactive oxygen species (ROS) following the PI3K‐AKT‐FOXO3a pathway signaling. 119 ROS initiate inflammatory pathways that are crucial for the onset and progression of MPNs. 6 Gain‐of‐function mutations in MPL and INDELs in CALR genes can promote JAK/STAT signaling, proliferation, and selective differentiation into the megakaryocyte lineage in a manner similar to the JAK2V617F mutation. 119

5.2. Immune dysregulations in MPNs

Recent research has shown that MPN driver mutations are acquired many years before symptoms of disease appear. 120 These mutations alone cannot cause malignant diseases and require additional factors, such as a weakened immune system. Since the bone marrow is the primary site for the development of myeloid malignancies, other related events also occur at this site. A variety of events, including pro‐inflammatory conditions, defects in antigen processing, and dysfunction of T lymphocyte are involved. 11 In MPNs, due to the paracrine activity of malignant clones, healthy cells in the bone marrow environment exhibit abnormal functions that create optimal conditions for the growth of malignant cells. It has been shown that mutant CALR protein can trigger strong inflammatory responses and reduce the ability of normal immune cells to carry out phagocytosis and process antigen. 121 , 122 Recent studies indicate that, inflammatory cytokines play a crucial role in the pathogenesis of myeloproliferative malignancies. 17 , 123 They are produced by both neoplastic and stromal cells in the bone marrow. The inflammatory state in MPN bone marrow and overstimulation of mesenchymal stem cells (MSC) result in impaired communication between osteoclasts and osteoblasts in the niche. 124 Defective activity of osteoclasts can lead to bone marrow fibrosis. Osteoclasts derived from mutant clone monocytes have both functional and morphological defects, showing abnormalities in size and resorption capacity. 125 Neuropathy damage to sympathetic nerve fibers is another alteration that occurs in the MPN bone marrow microenvironment. This damage is due to the overproduction of IL‐1 beta from the JAK2V617F mutant clone. After such injuries, the Schwann cell and nestin + MSC content of the bone marrow is reduced. 35 Hyper activated JAK–STAT signaling in neoplastic MPNs leads to the production of a variety of inflammatory cytokines. 126 All three types of driver mutations in MPNs result in activation of the JAK–STAT pathway. Some inflammatory cytokines such as tumor necrosis factor‐α (TNF‐α) and interleukin‐1 beta (IL‐1β), have selective growth advantages for JAK2V617F+ clones. 127 , 128 TNF‐α is produced in myeloid cells following activation of the Toll‐like receptors (TLR). Negative feedback in TLR signaling is a critical step in regulating cytokine production. IL‐10 is a known inhibitor of TLR signaling. A study by Lai et al. 129 showed that monocytes from MPN patients do not respond adequately to the anti‐inflammatory cytokine IL‐10. Therefore, aberrant TLR signaling and TNF‐α production result in a steady state of inflammation in patients with JAK2V617F+ MPNs. In addition to known factors that exacerbate inflammation in the bone marrow of patients with MPNs, it has recently been shown that the NLRP3 inflammasome may be part of this phenomenon. 130 The NLRP3 inflammasome is a multiprotein part of the innate immune system that activates the generation of pathogen‐associated molecular patterns (PAMPs) and DAMPs in pathological conditions. Upon activation, pro‐caspase‐1 spontaneously cleaves to its functional form, converting pro‐IL‐1 and proIL‐18 to their mature forms. 131 The NLRP inflammasome‐associated genes NLRP3, NF‐κB1, CARDS, IL‐1, and IL‐18 were expressed at elevated levels in MPN bone marrow samples, and their levels correlated with splenomegaly, JAK2V617F mutation, and leukocyte counts. 132 Immune dysregulation and driver mutations have a two‐way correlation in MPNs. The JAK2V617F mutation can result in a reduction of T helper17 (Th17) cells, myeloid dendritic cells, and effector Tregs. While CALR mutations are associated with increases in innate lymphoid cell 3 (ILC3) and decreases in T helper 1 (Th1) cells. 121 Neutrophil extracellular traps (NETs) are part of the innate immune system, resulting in an increase in cytoplasmic ROS levels of neutrophils and activation of myeloperoxidase, elastase, and protein arginine deiminase type 4 to promote chromatin decondensation. 133 Wolach O et al. recently showed that neutrophil stimulation by ionomycin significantly increases NET formation in JAK2V617F+ MPNs. They also found that advanced NET formation correlates with increased thrombosis in JAK2V617F+ mice. 134 It has previously been shown that MDSCs from MPN patients overexpress arginase‐1. 135 Arginase‐1 converts l‐arginine to l‐ornithine and urea; therefore, an increase in arginase‐1 levels leads to l‐arginine depletion. l‐arginine deficiency can impair T‐cell differentiation and activity. 94

5.3. Targeting immune system to improve defense against MPN malignancies

Ruxolitinib, an inhibitor of the tyrosine kinases JAK1 and JAK2, is the first drug approved for patients with moderate‐ and high‐risk PMF and PV who are unresponsive to hydroxyurea. 136 Reducing spleen size and improving overall survival (OS) are the main effects of this inhibitor. These significant achievements have contributed to the anti‐proliferative and anti‐inflammatory effects of ruxolitinib. 137 Treatment with ruxolitinib has a strong immunosuppressive effect and leads to a high risk of infection. More specific JAK2 inhibitors are expected to demonstrate a lower incidence of infection. Ruxoltinib only improves splenomegaly in 30–40% of PMF patients and does not have significantly impact on OS. Therefore, in recent years, efforts have been made to explore other compounds like BCL‐2 inhibitors, HMA agents, and other JAK inhibitors in conjunction with roxulitinib (NCT04562389, NCT03222609) and. 138 In addition to being used as the first‐line treatment, roxulitinib has recently been used to control the progression of the blastic and accelerated phase of MPNs, and promising results have been obtained. One notable clinical features of these phases of MPNs is resistance to common treatments. 138

Interferon‐alpha (IFN‐α) is a cytokine produced by various cells as part of the innate and cellular immune response and used as an immunomodulatory agent. Studies conducted over the years have shown that IFN‐α has diverse functions and could be effective in modulating disease symptoms. It has long been used to treat MPNs and other malignancies. 139 , 140 IFN‐α induces the expression of pro‐apoptotic genes such as caspase 4, caspase 8, tumor necrosis‐associated apoptosis‐inducing ligand (TRAIL), Fas/CD95 and the X‐linked inhibitor of apoptosis (XIAP). 139 Other anti‐leukemia and immunomodulatory effects of IFN‐α include induction of cell differentiation, enhancement of macrophage, T‐lymphocyte and NK cell function, and enhancement of tumor antigen presentation. 141 , 142

6. DISCUSSION

Unlike other malignancies, normal innate immune cells are generated from the neoplastic microenvironment in myeloid neoplasms and exposed to dozens of known and unknown immune modulators. The story of the immune microenvironment is different for myeloid malignancies than for other neoplasms because the normal bone marrow microenvironment is the breeding ground for many cells of the immune system. In myeloid malignancies, the initial seeds of immunomodulatory events are sown by malignant cells. Such immunosuppressive effects occur through direct cell connections, production and secretion of cytokines and specific enzymes, and changes in the expression of antigen‐presenting molecules. Targeting each of these axes can form the basis of new treatments, but they have their own complexities.

Among myeloid malignancies, AML deserves special attention due to its poor prognosis. It is difficult to harness the abnormal immune environment in AML. Biologically, AML is a complex disease with intricate communications between blast cells and the bone marrow microenvironment. Impressive and innovative new methods are being tested to address these challenges. Conventional chemotherapy regimens or targeted therapies alone can have profound immunomodulatory effects. 48 , 143 Hypomethylating agents (HMAs) show promise in reactivating of immune anti‐leukemic responses. 144 The therapeutic synergies of combining conventional chemotherapy (HMAs) with new immunotherapies in AML and high‐risk MDS are currently under investigation. Results are conflicting and further adjustments should be made considering biological complexities. 145 One barrier to the interaction of CD3+ effector T lymphocytes with AML leukemic blasts is the spatial distance within the bone marrow microenvironment. An ingenious way to resolve this problem is to use BiTEs antibodies (NCT02152956, NCT02730312, NCT02520427). The application of CIs is a promising strategy to overcome the exhaustion of normal T cells in refractory/relapsed AMLs (NCT02996474, NCT02845297, NCT02775903, NCT03066648). Dysregulation of innate immune system cells can lead to disease progression, and future treatments should address these abnormalities. For instance, several studies have shown low expression of NKG2D, NCRs, and DNAM‐1as activating receptors and KIR2DL2/L3 and NKG2A overexpression as inhibitory receptors in AML patients. 146 Adaptive NK cell transfer, in‐vitro expansion and monoclonal engineered antibodies that activate NK cell‐mediated antibody‐dependent cellular cytotoxicity (ADCC) are therapeutic strategies for harnessing NK cell immunity in AML.

Recently, it has been elucidated that inflammation plays a vital role in the development of MDS and MPNs. Therapeutic agents like ruxolitinib for JAK2V617F+ MPNs, exert their effects mainly by suppressing the JAK–STAT pathway and subsequently alleviating of inflammatory niches. Upregulation of inflammatory cytokines through the NF‐κB pathway is involved in the pathogenesis of MDS. The demonstration of pyroptosis is a brilliant finding in the pathogenesis of MDS. Activation of the NLRP3 inflammasome in hematopoietic stem/progenitor cells leads to pro‐inflammatory pyroptotic cell death through caspase‐1. The result of inflammasome activation in the bone marrow is lytic cell death followed by ineffective hematopoiesis and sterile inflammation in BONE MARROW microenvironment. 88 Modulation of the inflammasome activation pathway is considered a therapeutic targeting for MDS (see Figure 3). These inhibitors hold promise a better future for MDS, which currently have limited therapeutic options. The TLR‐IRAK‐NF‐κB axis is also involved in the pathogenesis of BCR‐ABL1 negative MPNs, and its therapeutic options may be useful for these entities. 123 Immune cells such as MDSCs, NK cells, and dendritic cells, can also create an immune‐suppressive environment in MDS, and manipulating them therapeutically may be associated with improvements in clinical outcomes. 147

FIGURE 3.

FIGURE 3

Potential clinical implications of inflammatory cascade inhibition in myelodysplastic syndromes. Pyroptosis is a cell death mechanism that is induced by inflammation and itself triggers an inflammatory milieu. At the onset of pyroptosis, surface TLR receptors are activated by S100A8/A9. The IRAK/MYD88 axis transmits TLR signaling to the nucleus through NF‐kB and phosphorylated STAT3. Eventually, this cascade led to gene expression of the inflammasome elements Gasdermin‐D, IL‐1 and IL‐18. By inflammasome formation and activation, pro‐gasdermin‐D, IL‐1β and IL‐18 will be cleaved and matured. Gasdermin‐D subunits formed a membrane pore. Mature IL1β and IL18 large cytokines could be released from the pyroptotic cells by these pores. As indicated by the red dashed boxes in the image, certain inhibitors of different stages of pyroptosis have potential clinical implications for MDS treatment. Some of these active ingredients are in clinical trials. 148 , 149 , 150 , 151 , 152 The figure was produced with the assistance of Servier Medical Art (https://smart.servier.com).

7. CONCLUSION

Despite the central role of the bone marrow microenvironment in providing nourishment and immune modulation in myeloid malignancies, the specific players involved in the reciprocal interaction between leukemic cells and the immune system need to be identified. Recent advancements in understanding the pathologic mechanisms of immune system alterations in myeloid malignancies have enhanced our knowledge. A prime example of these advancements is the recognition of the inflammatory cytokines in immune modulation and the progression of MDS and MPNs. A better understanding of the key interactions within the bone marrow microenvironment requires the use of robust laboratory techniques. By elucidating the critical interactions that disturb immune system integrity in the bone marrow microenvironment, we can anticipate the development of more targeted treatments for myeloid malignancies. This review aims to summarize the current pathophysiologic findings and potential treatments for immune dysregulations in myeloid malignancies.

AUTHOR CONTRIBUTIONS

Mohammad Jafar Sharifi: Conceptualization (equal); project administration (equal); visualization (lead); writing – original draft (equal); writing – review and editing (lead). Ling Xu: Writing – original draft (equal); writing – review and editing (equal). Nahid Nasiri: Writing – original draft (equal); writing – review and editing (equal). Mehnoosh Ashja‐Arvan: Writing – original draft (equal); writing – review and editing (equal). Hadis Soleimanzadeh: Writing – original draft (equal); writing – review and editing (equal). Mazdak Ganjalikhani‐Hakemi: Conceptualization (lead); funding acquisition (lead); project administration (equal); resources (lead); supervision (equal); writing – review and editing (equal).

FUNDING INFORMATION

Thanks to TÜBİTAK for supporting the APC of the current work.

CONFLICT OF INTEREST STATEMENT

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Sharifi MJ, Xu L, Nasiri N, Ashja‐Arvan M, Soleimanzadeh H, Ganjalikhani‐Hakemi M. Immune‐dysregulation harnessing in myeloid neoplasms. Cancer Med. 2024;13:e70152. doi: 10.1002/cam4.70152

DATA AVAILABILITY STATEMENT

Not applicable.

REFERENCES

  • 1. Kumar R, Godavarthy PS, Krause DS. The bone marrow microenvironment in health and disease at a glance. J Cell Sci. 2018;131(4):jcs201707. [DOI] [PubMed] [Google Scholar]
  • 2. Agarwal P, Bhatia R. Influence of bone marrow microenvironment on leukemic stem cells: breaking up an intimate relationship. Adv Cancer Res. 2015;127:227‐252. [DOI] [PubMed] [Google Scholar]
  • 3. Nyamondo K, Wheadon H. Micro‐environment alterations through time leading to myeloid malignancies. Br J Pharmacol. 2022;181:283‐294. [DOI] [PubMed] [Google Scholar]
  • 4. Lamble AJ, Lind EF. Targeting the immune microenvironment in acute myeloid leukemia: a focus on T cell immunity. Front Oncol. 2018;8:213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Papaioannou NE, Beniata OV, Vitsos P, Tsitsilonis O, Samara P. Harnessing the immune system to improve cancer therapy. Ann Transl Med. 2016;4(14):261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Allegra A, Pioggia G, Tonacci A, Casciaro M, Musolino C, Gangemi S. Synergic crosstalk between inflammation, oxidative stress, and genomic alterations in BCR‐ABL‐negative myeloproliferative neoplasm. Antioxidants (Basel). 2020;9(11):1037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Kayser S, Levis MJ. Updates on targeted therapies for acute myeloid leukaemia. Br J Haematol. 2022;196(2):316‐328. [DOI] [PubMed] [Google Scholar]
  • 8. Binnewies M, Roberts EW, Kersten K, et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med. 2018;24(5):541‐550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Bruck O, Blom S, Dufva O, et al. Immune cell contexture in the bone marrow tumor microenvironment impacts therapy response in CML. Leukemia. 2018;32(7):1643‐1656. [DOI] [PubMed] [Google Scholar]
  • 10. Bruck O, Dufva O, Hohtari H, et al. Immune profiles in acute myeloid leukemia bone marrow associate with patient age, T‐cell receptor clonality, and survival. Blood Adv. 2020;4(2):274‐286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Dufva O, Polonen P, Bruck O, et al. Immunogenomic landscape of hematological malignancies. Cancer Cell. 2020;38(3):380‐399.e13. [DOI] [PubMed] [Google Scholar]
  • 12. Tang L, Wu J, Li CG, et al. Characterization of immune dysfunction and identification of prognostic immune‐related risk factors in acute myeloid leukemia. Clin Cancer Res. 2020;26(7):1763‐1772. [DOI] [PubMed] [Google Scholar]
  • 13. Chalmers ZR, Connelly CF, Fabrizio D, et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 2017;9(1):34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Vadakekolathu J, Minden MD, Hood T, et al. Immune landscapes predict chemotherapy resistance and immunotherapy response in acute myeloid leukemia. Sci Transl Med. 2020;12(546):eaaz0463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Hanahan D. Hallmarks of cancer: new dimensions. Cancer Discov. 2022;12(1):31‐46. [DOI] [PubMed] [Google Scholar]
  • 16. Beck DB, Ferrada MA, Sikora KA, et al. Somatic mutations in UBA1 and severe adult‐onset autoinflammatory disease. N Engl J Med. 2020;383(27):2628‐2638. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Balandran JC, Lasry A, Aifantis I. The role of inflammation in the initiation and progression of myeloid neoplasms. Blood Cancer Discov. 2023;4:254‐266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Licht JD, Sternberg DW. The molecular pathology of acute myeloid leukemia. Hematology. 2005;2005(1):137‐142. [DOI] [PubMed] [Google Scholar]
  • 19. Padmakumar D, Chandraprabha VR, Gopinath P, et al. A concise review on the molecular genetics of acute myeloid leukemia. Leuk Res. 2021;111:106727. [DOI] [PubMed] [Google Scholar]
  • 20. Thol F, Ganser A. Molecular pathogenesis of acute myeloid leukemia: a diverse disease with new perspectives. Front Med China. 2010;4(4):356‐362. [DOI] [PubMed] [Google Scholar]
  • 21. Hino C, Pham B, Park D, et al. Targeting the tumor microenvironment in acute myeloid leukemia: the future of immunotherapy and natural products. Biomedicine. 2022;10(6):1410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Stringaris K, Sekine T, Khoder A, et al. Leukemia‐induced phenotypic and functional defects in natural killer cells predict failure to achieve remission in acute myeloid leukemia. Haematologica. 2014;99(5):836‐847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Sandoval‐Borrego D, Moreno‐Lafont MC, Vazquez‐Sanchez EA, et al. Overexpression of CD158 and NKG2A inhibitory receptors and underexpression of NKG2D and NKp46 activating receptors on NK cells in acute myeloid leukemia. Arch Med Res. 2016;47(1):55‐64. [DOI] [PubMed] [Google Scholar]
  • 24. Sanchez‐Correa B, Morgado S, Gayoso I, et al. Human NK cells in acute myeloid leukaemia patients: analysis of NK cell‐activating receptors and their ligands. Cancer Immunol Immunother. 2011;60(8):1195‐1205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Zeng X, Yao D, Liu L, et al. Terminal differentiation of bone marrow NK cells and increased circulation of TIGIT(+) NK cells may be related to poor outcome in acute myeloid leukemia. Asia Pac J Clin Oncol. 2022;18(4):456‐464. [DOI] [PubMed] [Google Scholar]
  • 26. Yang L, Feng Y, Wang S, et al. Siglec‐7 is an indicator of natural killer cell function in acute myeloid leukemia. Int Immunopharmacol. 2021;99:107965. [DOI] [PubMed] [Google Scholar]
  • 27. Liu G, Zhang Q, Yang J, et al. Increased TIGIT expressing NK cells with dysfunctional phenotype in AML patients correlated with poor prognosis. Cancer Immunol Immunother. 2022;71(2):277‐287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Tumino N, Besi F, Di Pace AL, et al. PMN‐MDSC are a new target to rescue graft‐versus‐leukemia activity of NK cells in haplo‐HSC transplantation. Leukemia. 2020;34(3):932‐937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Szczepanski MJ, Szajnik M, Welsh A, Whiteside TL, Boyiadzis M. Blast‐derived microvesicles in sera from patients with acute myeloid leukemia suppress natural killer cell function via membrane‐associated transforming growth factor‐beta1. Haematologica. 2011;96(9):1302‐1309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Hyun SY, Na EJ, Jang JE, et al. Immunosuppressive role of CD11b(+) CD33(+) HLA‐DR(−) myeloid‐derived suppressor cells‐like blast subpopulation in acute myeloid leukemia. Cancer Med. 2020;9(19):7007‐7017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Pyzer AR, Stroopinsky D, Rajabi H, et al. MUC1‐mediated induction of myeloid‐derived suppressor cells in patients with acute myeloid leukemia. Blood. 2017;129(13):1791‐1801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Tohumeken S, Baur R, Bottcher M, et al. Palmitoylated proteins on AML‐derived extracellular vesicles promote myeloid‐derived suppressor cell differentiation via TLR2/Akt/mTOR signaling. Cancer Res. 2020;80(17):3663‐3676. [DOI] [PubMed] [Google Scholar]
  • 33. Peterlin P, Debord C, Eveillard M, et al. Peripheral levels of monocytic myeloid‐derived suppressive cells before and after first induction predict relapse and survivals in AML patients. J Cell Mol Med. 2022;26(21):5486‐5492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Wang L, Jia B, Claxton DF, et al. VISTA is highly expressed on MDSCs and mediates an inhibition of T cell response in patients with AML. Onco Targets Ther. 2018;7(9):e1469594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Arranz L, Sanchez‐Aguilera A, Martin‐Perez D, et al. Neuropathy of haematopoietic stem cell niche is essential for myeloproliferative neoplasms. Nature. 2014;512(7512):78‐81. [DOI] [PubMed] [Google Scholar]
  • 36. Wang L, Zheng G. Macrophages in leukemia microenvironment. Blood Sci. 2019;1(1):29‐33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Miari KE, Guzman ML, Wheadon H, Williams MTS. Macrophages in acute myeloid Leukaemia: significant players in therapy resistance and patient outcomes. Front Cell Dev Biol. 2021;9:692800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Xu ZJ, Gu Y, Wang CZ, et al. The M2 macrophage marker CD206: a novel prognostic indicator for acute myeloid leukemia. Onco Targets Ther. 2020;9(1):1683347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Al‐Matary YS, Botezatu L, Opalka B, et al. Acute myeloid leukemia cells polarize macrophages towards a leukemia supporting state in a growth factor independence 1 dependent manner. Haematologica. 2016;101(10):1216‐1227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Jiang M, Zhang J, Qian L, et al. MOZ forms an autoregulatory feedback loop with miR‐223 in AML and monocyte/macrophage development. iScience. 2019;11:189‐204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Park JH, Lee HK. Function of gammadelta T cells in tumor immunology and their application to cancer therapy. Exp Mol Med. 2021;53(3):318‐327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Ismail MM, Abdulateef NAB. Bone marrow T‐cell percentage: A novel prognostic indicator in acute myeloid leukemia. Int J Hematol. 2017;105(4):453‐464. [DOI] [PubMed] [Google Scholar]
  • 43. Ravandi F, Assi R, Daver N, 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(9):e480‐e488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Le Dieu R, Taussig DC, Ramsay AG, et al. Peripheral blood T cells in acute myeloid leukemia (AML) patients at diagnosis have abnormal phenotype and genotype and form defective immune synapses with AML blasts. Blood. 2009;114(18):3909‐3916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Kong Y, Zhu L, Schell TD, et al. T‐cell immunoglobulin and ITIM domain (TIGIT) associates with CD8+ T‐cell exhaustion and poor clinical outcome in AML patients. Clin Cancer Res. 2016;22(12):3057‐3066. [DOI] [PubMed] [Google Scholar]
  • 46. Zhu L, Kong Y, Zhang J, et al. Blimp‐1 impairs T cell function via upregulation of TIGIT and PD‐1 in patients with acute myeloid leukemia. J Hematol Oncol. 2017;10(1):124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Jia B, Wang L, Claxton DF, et al. Bone marrow CD8 T cells express high frequency of PD‐1 and exhibit reduced anti‐leukemia response in newly diagnosed AML patients. Blood Cancer J. 2018;8(3):34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Knaus HA, Berglund S, Hackl H, et al. Signatures of CD8+ T cell dysfunction in AML patients and their reversibility with response to chemotherapy. JCI Insight. 2018;3(21):e120974. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Wang M, Bu J, Zhou M, et al. CD8(+)T cells expressing both PD‐1 and TIGIT but not CD226 are dysfunctional in acute myeloid leukemia (AML) patients. Clin Immunol. 2018;190:64‐73. [DOI] [PubMed] [Google Scholar]
  • 50. Schnorfeil FM, Lichtenegger FS, Emmerig K, et al. T cells are functionally not impaired in AML: increased PD‐1 expression is only seen at time of relapse and correlates with a shift towards the memory T cell compartment. J Hematol Oncol. 2015;8:93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Deng M, Gui X, Kim J, et al. LILRB4 signalling in leukaemia cells mediates T cell suppression and tumour infiltration. Nature. 2018;562(7728):605‐609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Li Z, Deng M, Huang F, et al. LILRB4 ITIMs mediate the T cell suppression and infiltration of acute myeloid leukemia cells. Cell Mol Immunol. 2020;17(3):272‐282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Coles SJ, Gilmour MN, Reid R, et al. The immunosuppressive ligands PD‐L1 and CD200 are linked in AML T‐cell immunosuppression: identification of a new immunotherapeutic synapse. Leukemia. 2015;29(9):1952‐1954. [DOI] [PubMed] [Google Scholar]
  • 54. Coles SJ, Hills RK, Wang EC, et al. Increased CD200 expression in acute myeloid leukemia is linked with an increased frequency of FoxP3+ regulatory T cells. Leukemia. 2012;26(9):2146‐2148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Oda SK, Daman AW, Garcia NM, et al. A CD200R‐CD28 fusion protein appropriates an inhibitory signal to enhance T‐cell function and therapy of murine leukemia. Blood. 2017;130(22):2410‐2419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Rutella S, Vadakekolathu J, Mazziotta F, et al. Immune dysfunction signatures predict outcomes and define checkpoint blockade‐unresponsive microenvironments in acute myeloid leukemia. J Clin Invest. 2022;132(21):e159579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Williams P, Basu S, Garcia‐Manero G, 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. 2019;125(9):1470‐1481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Shenghui Z, Yixiang H, Jianbo W, et al. Elevated frequencies of CD4(+) CD25(+) CD127lo regulatory T cells is associated to poor prognosis in patients with acute myeloid leukemia. Int J Cancer. 2011;129(6):1373‐1381. [DOI] [PubMed] [Google Scholar]
  • 59. Wang M, Zhang C, Tian T, et al. Increased regulatory T cells in peripheral blood of acute myeloid leukemia patients rely on tumor necrosis factor (TNF)‐alpha‐TNF Receptor‐2 pathway. Front Immunol. 2018;9:1274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Delia M, Carluccio P, Mestice A, et al. After treatment decrease of bone marrow Tregs and outcome in younger patients with newly diagnosed acute myeloid leukemia. J Immunol Res. 2020;2020:2134647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Wan Y, Zhang C, Xu Y, et al. Hyperfunction of CD4 CD25 regulatory T cells in de novo acute myeloid leukemia. BMC Cancer. 2020;20(1):472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Thummalapalli R, Knaus HA, Gojo I, Zeidner JF. Immune checkpoint inhibitors in AML‐A new frontier. Curr Cancer Drug Targets. 2020;20(7):545‐557. [DOI] [PubMed] [Google Scholar]
  • 63. Vago L, Gojo I. Immune escape and immunotherapy of acute myeloid leukemia. J Clin Invest. 2020;130(4):1552‐1564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Daver N. Immune checkpoint inhibitors in acute myeloid leukemia. Best Pract Res Clin Haematol. 2021;34(1):101247. [DOI] [PubMed] [Google Scholar]
  • 65. Hao F, Sholy C, Wang C, Cao M, Kang X. The role of T cell immunotherapy in acute myeloid leukemia. Cells. 2021;10(12):3376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Borate U, Esteve J, Porkka K, et al. Phase Ib study of the anti‐TIM‐3 antibody MBG453 in combination with decitabine in patients with high‐risk myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). Blood. 2019;134(Supplement_1):570. [Google Scholar]
  • 67. Castaigne S, Pautas C, Terre C, et al. Effect of gemtuzumab ozogamicin on survival of adult patients with de‐novo acute myeloid leukaemia (ALFA‐0701): a randomised, open‐label, phase 3 study. Lancet. 2012;379(9825):1508‐1516. [DOI] [PubMed] [Google Scholar]
  • 68. Dohner H, Weber D, Krzykalla J, et al. Intensive chemotherapy with or without gemtuzumab ozogamicin in patients with NPM1‐mutated acute myeloid leukaemia (AMLSG 09‐09): a randomised, open‐label, multicentre, phase 3 trial. Lancet Haematol. 2023;10(7):e495‐e509. [DOI] [PubMed] [Google Scholar]
  • 69. Riether C, Pabst T, Hopner S, et al. Targeting CD70 with cusatuzumab eliminates acute myeloid leukemia stem cells in patients treated with hypomethylating agents. Nat Med. 2020;26(9):1459‐1467. [DOI] [PubMed] [Google Scholar]
  • 70. Wang QS, Wang Y, Lv HY, et al. Treatment of CD33‐directed chimeric antigen receptor‐modified T cells in one patient with relapsed and refractory acute myeloid leukemia. Mol Ther. 2015;23(1):184‐191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Budde L, Song JY, Kim Y, et al. Remissions of acute myeloid leukemia and Blastic plasmacytoid dendritic cell neoplasm following treatment with CD123‐specific CAR T cells: a first‐in‐human clinical trial. Blood. 2017;130(Supplement 1):811. [Google Scholar]
  • 72. Yao S, Jianlin C, Yarong L, et al. Donor‐derived CD123‐targeted CAR T cell serves as a RIC regimen for haploidentical transplantation in a patient with FUS‐ERG+ AML. Front Oncol. 2019;9:1358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Hofmann S, Schubert ML, Wang L, et al. Chimeric antigen receptor (CAR) T cell therapy in acute myeloid leukemia (AML). J Clin Med. 2019;8(2):200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Fiorenza S, Turtle CJ. CAR‐T cell therapy for acute myeloid leukemia: preclinical rationale, current clinical Progress, and barriers to success. BioDrugs. 2021;35(3):281‐302. [DOI] [PubMed] [Google Scholar]
  • 75. Keilholz U, Letsch A, Busse A, et al. 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(26):6541‐6548. [DOI] [PubMed] [Google Scholar]
  • 76. Maslak PG, Dao T, Bernal Y, et al. Phase 2 trial of a multivalent WT1 peptide vaccine (galinpepimut‐S) in acute myeloid leukemia. Blood Adv. 2018;2(3):224‐234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Qin Y, Zhu H, Jiang B, et al. Expression patterns of WT1 and PRAME in acute myeloid leukemia patients and their usefulness for monitoring minimal residual disease. Leuk Res. 2009;33(3):384‐390. [DOI] [PubMed] [Google Scholar]
  • 78. Sandri S, De Sanctis F, Lamolinara A, et al. Effective control of acute myeloid leukaemia and acute lymphoblastic leukaemia progression by telomerase specific adoptive T‐cell therapy. Oncotarget. 2017;8(50):86987‐87001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Spranger S, Jeremias I, Wilde S, et al. TCR‐transgenic lymphocytes specific for HMMR/Rhamm limit tumor outgrowth in vivo. Blood. 2012;119(15):3440‐3449. [DOI] [PubMed] [Google Scholar]
  • 80. van der Lee DI, Reijmers RM, Honders MW, et al. Mutated nucleophosmin 1 as immunotherapy target in acute myeloid leukemia. J Clin Invest. 2019;129(2):774‐785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Venugopal S, Daver N, Ravandi F. An update on the clinical evaluation of antibody‐based therapeutics in acute myeloid leukemia. Curr Hematol Malig Rep. 2021;16(1):89‐96. [DOI] [PubMed] [Google Scholar]
  • 82. Abaza Y, Fathi AT. Monoclonal antibodies in acute myeloid leukemia‐are we there yet? Cancer J. 2022;28(1):37‐42. [DOI] [PubMed] [Google Scholar]
  • 83. GT Biopharma I . GT Biopharma Announces GTB‐3550 TRIKE™ Monotherapy Rescues and Restores NK cell immune surveillance in relapsed/refractory AML and MDS cancer patients. 2021. Available from: https://bit.ly/3aanflR
  • 84. Xu J, Niu T. Natural killer cell‐based immunotherapy for acute myeloid leukemia. J Hematol Oncol. 2020;13(1):167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Shallis RM, Ahmad R, Zeidan AM. The genetic and molecular pathogenesis of myelodysplastic syndromes. Eur J Haematol. 2018;101(3):260‐271. [DOI] [PubMed] [Google Scholar]
  • 86. Crowe LAN, McLean M, Kitson SM, et al. S100A8 & S100A9: alarmin mediated inflammation in tendinopathy. Sci Rep. 2019;9(1):1463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Sallman DA, List A. The role of innate immunity in MDS pathogenesis. HemaSphere. 2019;3(Suppl):135‐137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Sallman DA, Cluzeau T, Basiorka AA, List A. Unraveling the pathogenesis of MDS: the NLRP3 inflammasome and pyroptosis drive the MDS phenotype. Front Oncol. 2016;6:151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Lara‐Reyna S, Caseley EA, Topping J, et al. Inflammasome activation: from molecular mechanisms to autoinflammation. Clin Transl Immunol. 2022;11(7):e1404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Sallman DA, List A. The central role of inflammatory signaling in the pathogenesis of myelodysplastic syndromes. Blood. 2019;133(10):1039‐1048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Espinoza JL, Kamio K, Lam VQ, Takami A. The impact of NLRP3 activation on hematopoietic stem cell transplantation. Int J Mol Sci. 2021;22(21):11845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Dadkhah M, Sharifi M, Sharifi MJ, Tehrani RM. Effects of glucose on the proliferation of human umbilical cord blood hematopoietic stem cells. Cell Tissue Bank. 2022;24:485‐494. [DOI] [PubMed] [Google Scholar]
  • 93. Papafragkos I, Grigoriou M, Boon L, Kloetgen A, Hatzioannou A, Verginis P. Ablation of NLRP3 inflammasome rewires MDSC function and promotes tumor regression. Front Immunol. 2022;13:889075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Lv M, Wang K, Huang XJ. Myeloid‐derived suppressor cells in hematological malignancies: friends or foes. J Hematol Oncol. 2019;12(1):105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Zhang X, Yang X, Ma L, Zhang Y, Wei J. Immune dysregulation and potential targeted therapy in myelodysplastic syndrome. Ther Adv Hematol. 2023;14:20406207231183330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Valdes‐Ferrer SI, Papoin J, Dancho ME, et al. HMGB1 mediates anemia of inflammation in murine sepsis survivors. Mol Med. 2016;21(1):951‐958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Dulmovits BM, Tang Y, Papoin J, et al. HMGB1‐mediated restriction of EPO signaling contributes to anemia of inflammation. Blood. 2022;139(21):3181‐3193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Starczynowski DT, Kuchenbauer F, Argiropoulos B, et al. Identification of miR‐145 and miR‐146a as mediators of the 5q‐ syndrome phenotype. Nat Med. 2010;16(1):49‐58. [DOI] [PubMed] [Google Scholar]
  • 99. Neves‐Costa A, Moita LF. TET1 is a negative transcriptional regulator of IL‐1β in the THP‐1 cell line. Mol Immunol. 2013;54(3–4):264‐270. [DOI] [PubMed] [Google Scholar]
  • 100. Zhang Q, Zhao K, Shen Q, et al. Tet2 is required to resolve inflammation by recruiting Hdac2 to specifically repress IL‐6. Nature. 2015;525(7569):389‐393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Li X, Zhang Q, Ding Y, et al. Methyltransferase Dnmt3a upregulates HDAC9 to deacetylate the kinase TBK1 for activation of antiviral innate immunity. Nat Immunol. 2016;17(7):806‐815. [DOI] [PubMed] [Google Scholar]
  • 102. Abdel‐Wahab O, Gao J, Adli M, et al. Deletion of Asxl1 results in myelodysplasia and severe developmental defects in vivo. J Exp Med. 2013;210(12):2641‐2659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Chen L, Chen JY, Huang YJ, et al. The augmented R‐loop is a unifying mechanism for myelodysplastic syndromes induced by high‐risk splicing factor mutations. Mol Cell. 2018;69(3):412‐425.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Lee SC, Dvinge H, Kim E, et al. Modulation of splicing catalysis for therapeutic targeting of leukemia with mutations in genes encoding spliceosomal proteins. Nat Med. 2016;22(6):672‐678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Lee SC, North K, Kim E, et al. Synthetic lethal and convergent biological effects of cancer‐associated Spliceosomal gene mutations. Cancer Cell. 2018;34(2):225‐241.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Park SM, Ou J, Chamberlain L, et al. U2AF35(S34F) promotes transformation by directing aberrant ATG7 pre‐mRNA 3′ end formation. Mol Cell. 2016;62(4):479‐490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Bourbon E, Heiblig M, Gerfaud Valentin M, et al. Therapeutic options in VEXAS syndrome: insights from a retrospective series. Blood. 2021;137(26):3682‐3684. [DOI] [PubMed] [Google Scholar]
  • 108. Chakraborty S, Shapiro LC, de Oliveira S, Rivera‐Pena B, Verma A, Shastri A. Therapeutic targeting of the inflammasome in myeloid malignancies. Blood Cancer J. 2021;11(9):152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Winter S, Shoaie S, Kordasti S, Platzbecker U. Integrating the “immunome” in the stratification of myelodysplastic syndromes and future clinical trial design. J Clin Oncol. 2020;38(15):1723‐1735. [DOI] [PubMed] [Google Scholar]
  • 110. Comont T, Treiner E, Vergez F. From immune dysregulations to therapeutic perspectives in myelodysplastic syndromes: a review. Diagnostics (Basel). 2021;11(11):1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Brunner AM, Esteve J, Porkka K, et al. Phase Ib study of sabatolimab (MBG453), a novel immunotherapy targeting TIM‐3 antibody, in combination with decitabine or azacitidine in high‐ or very high‐risk myelodysplastic syndromes. Am J Hematol. 2024;99(2):E32‐E36. [DOI] [PubMed] [Google Scholar]
  • 112. Basiorka AA, McGraw KL, Abbas‐Aghababazadeh F, et al. Assessment of ASC specks as a putative biomarker of pyroptosis in myelodysplastic syndromes: an observational cohort study. Lancet Haematol. 2018;5(9):e393‐e402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Khalilian P, Eskandari N, Sharifi MJ, Soltani M, Nematollahi P. Toll‐like receptor 4, 2, and interleukin 1 receptor associated Kinase4: possible diagnostic biomarkers in myelodysplastic syndrome patients. Adv Biomed Res. 2024;13:17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Soltani M, Sharifi MJ, Khalilian P, et al. Potential diagnostic value of abnormal Pyroptosis genes expression in myelodysplastic syndromes (MDS): a primary observational cohort study. Int J Hematol Oncol Stem Cell Res. 2024;18(2):156‐164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Stahl M, DeVeaux M, de Witte T, et al. The use of immunosuppressive therapy in MDS: clinical outcomes and their predictors in a large international patient cohort. Blood Adv. 2018;2(14):1765‐1772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Passweg JR, Giagounidis AA, Simcock M, 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(3):303‐309. [DOI] [PubMed] [Google Scholar]
  • 117. Olnes MJ, Sloand EM. Targeting immune dysregulation in myelodysplastic syndromes. JAMA. 2011;305(8):814‐819. [DOI] [PubMed] [Google Scholar]
  • 118. Khoury JD, Solary E, Abla O, et al. The 5th edition of the World Health Organization classification of Haematolymphoid Tumours: myeloid and histiocytic/dendritic neoplasms. Leukemia. 2022;36(7):1703‐1719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Grabek J, Straube J, Bywater M, Lane SW. MPN: the molecular drivers of disease initiation, progression and transformation and their effect on treatment. Cells. 2020;9(8):1901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Cordua S, Kjaer L, Skov V, Pallisgaard N, Hasselbalch HC, Ellervik C. Prevalence and phenotypes of JAK2 V617F and calreticulin mutations in a Danish general population. Blood. 2019;134(5):469‐479. [DOI] [PubMed] [Google Scholar]
  • 121. Romano M, Sollazzo D, Trabanelli S, et al. Mutations in JAK2 and calreticulin genes are associated with specific alterations of the immune system in myelofibrosis. Onco Targets Ther. 2017;6(10):e1345402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Garbati MR, Welgan CA, Landefeld SH, et al. Mutant calreticulin‐expressing cells induce monocyte hyperreactivity through a paracrine mechanism. Am J Hematol. 2016;91(2):211‐219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Mascarenhas J, Gleitz HFE, Chifotides HT, et al. Biological drivers of clinical phenotype in myelofibrosis. Leukemia. 2023;37(2):255‐264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Zupan J, Jeras M, Marc J. Osteoimmunology and the influence of pro‐inflammatory cytokines on osteoclasts. Biochem Med (Zagreb). 2013;23(1):43‐63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Karagianni A, Ravid K. Myeloproliferative disorders and their effects on bone homeostasis: the role of megakaryocytes. Blood. 2022;139(21):3127‐3137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Vainchenker W, Kralovics R. Genetic basis and molecular pathophysiology of classical myeloproliferative neoplasms. Blood. 2017;129(6):667‐679. [DOI] [PubMed] [Google Scholar]
  • 127. Fleischman AG, Aichberger KJ, Luty SB, et al. TNFalpha facilitates clonal expansion of JAK2V617F positive cells in myeloproliferative neoplasms. Blood. 2011;118(24):6392‐6398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Rahman MF, Yang Y, Le BT, et al. Interleukin‐1 contributes to clonal expansion and progression of bone marrow fibrosis in JAK2V617F‐induced myeloproliferative neoplasm. Nat Commun. 2022;13(1):5347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Lai HY, Brooks SA, Craver BM, et al. Defective negative regulation of toll‐like receptor signaling leads to excessive TNF‐alpha in myeloproliferative neoplasm. Blood Adv. 2019;3(2):122‐131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Di Battista V, Bochicchio MT, Giordano G, Napolitano M, Lucchesi A. Genetics and pathogenetic role of inflammasomes in Philadelphia negative chronic myeloproliferative neoplasms: a narrative review. Int J Mol Sci. 2021;22(2):561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Blevins HM, Xu Y, Biby S, Zhang S. The NLRP3 inflammasome pathway: a review of mechanisms and inhibitors for the treatment of inflammatory diseases. Front Aging Neurosci. 2022;14:879021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Zhou Y, Yan S, Liu N, et al. Genetic polymorphisms and expression of NLRP3 inflammasome‐related genes are associated with Philadelphia chromosome‐negative myeloproliferative neoplasms. Hum Immunol. 2020;81(10–11):606‐613. [DOI] [PubMed] [Google Scholar]
  • 133. Papayannopoulos V. Neutrophil extracellular traps in immunity and disease. Nat Rev Immunol. 2018;18(2):134‐147. [DOI] [PubMed] [Google Scholar]
  • 134. Wolach O, Sellar RS, Martinod K, et al. Increased neutrophil extracellular trap formation promotes thrombosis in myeloproliferative neoplasms. Sci Transl Med. 2018;10(436):eaan8292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Wang JC, Kundra A, Andrei M, et al. Myeloid‐derived suppressor cells in patients with myeloproliferative neoplasm. Leuk Res. 2016;43:39‐43. [DOI] [PubMed] [Google Scholar]
  • 136. Leroy E, Constantinescu SN. Rethinking JAK2 inhibition: towards novel strategies of more specific and versatile Janus kinase inhibition. Leukemia. 2017;31(5):1023‐1038. [DOI] [PubMed] [Google Scholar]
  • 137. Elli EM, Barate C, Mendicino F, Palandri F, Palumbo GA. Mechanisms underlying the anti‐inflammatory and immunosuppressive activity of Ruxolitinib. Front Oncol. 2019;9:1186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Mascarenhas JO, Rampal RK, Kosiorek HE, et al. Phase 2 study of ruxolitinib and decitabine in patients with myeloproliferative neoplasm in accelerated and blast phase. Blood Adv. 2020;4(20):5246‐5256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Kiladjian JJ, Mesa RA, Hoffman R. The renaissance of interferon therapy for the treatment of myeloid malignancies. Blood. 2011;117(18):4706‐4715. [DOI] [PubMed] [Google Scholar]
  • 140. Bewersdorf JP, Giri S, Wang R, et al. Interferon alpha therapy in essential thrombocythemia and polycythemia vera‐a systematic review and meta‐analysis. Leukemia. 2021;35(6):1643‐1660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Mullally A, Bruedigam C, Poveromo L, et al. Depletion of Jak2V617F myeloproliferative neoplasm‐propagating stem cells by interferon‐alpha in a murine model of polycythemia vera. Blood. 2013;121(18):3692‐3702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Paquette RL, Hsu N, Said J, et al. Interferon‐alpha induces dendritic cell differentiation of CML mononuclear cells in vitro and in vivo. Leukemia. 2002;16(8):1484‐1489. [DOI] [PubMed] [Google Scholar]
  • 143. Mathew NR, Baumgartner F, Braun L, et al. Sorafenib promotes graft‐versus‐leukemia activity in mice and humans through IL‐15 production in FLT3‐ITD‐mutant leukemia cells. Nat Med. 2018;24(3):282‐291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Jones PA, Ohtani H, Chakravarthy A, De Carvalho DD. Epigenetic therapy in immune‐oncology. Nat Rev Cancer. 2019;19(3):151‐161. [DOI] [PubMed] [Google Scholar]
  • 145. Zavras PD, Shastri A, Goldfinger M, Verma AK, Saunthararajah Y. Clinical trials assessing hypomethylating agents combined with other therapies: causes for failure and potential solutions. Clin Cancer Res. 2021;27(24):6653‐6661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Carlsten M, Järås M. Natural killer cells in myeloid malignancies: immune surveillance, NK cell dysfunction, and pharmacological opportunities to bolster the endogenous NK cells. Front Immunol. 2019;10:2357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. D'Silva S, Rajadhyaksha SB, Singh M. Immune dysregulation in MDS: the role of cytokines and immune cells. Recent Developments in Myelodysplastic Syndromes. IntchOpen; 2019:45. [Google Scholar]
  • 148. Garcia‐Manero G, Jabbour EJ, Konopleva MY, 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(Supplement 1):798. [Google Scholar]
  • 149. Randhawa JK, Jabbar KJ, Kadia T, et al. Phase II study of targeted subcutaneous (SC) bortezomib for patients with low‐ or Intermediate‐1 (Int‐1)‐risk myelodysplastic syndrome (MDS) with evidence of NF‐κB activation. Blood. 2014;124(21):1930. [Google Scholar]
  • 150. Winkler A, Sun W, De S, et al. The Interleukin‐1 receptor‐associated kinase 4 inhibitor PF‐06650833 blocks inflammation in preclinical models of rheumatic disease and in humans enrolled in a randomized clinical trial. Arthritis Rheumatol (Hoboken, NJ). 2021;73(12):2206‐2218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Tapia‐Abellán A, Angosto‐Bazarra D, Martínez‐Banaclocha H, et al. MCC950 closes the active conformation of NLRP3 to an inactive state. Nat Chem Biol. 2019;15(6):560‐564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Jonas BA, Logan AC, Jeyakumar D, et al. A phase 1 trial of the combination of ibrutinib and Azacitidine for the treatment of higher risk myelodysplastic syndromes: University of California Hematologic Malignancies Consortium (UCHMC) study 1503. Blood. 2017;130(Supplement 1):2973‐3088. [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Not applicable.


Articles from Cancer Medicine are provided here courtesy of Wiley

RESOURCES