Abstract
Myeloid‐derived suppressor cells (MDSCs) are an immature group of myeloid‐derived cells generated from myeloid cell precursors in the bone marrow. MDSCs appear almost exclusively in pathological conditions, such as tumor progression and various inflammatory diseases. The leading function of MDSCs is their immunosuppressive ability, which plays a crucial role in tumor progression and metastasis through their immunosuppressive effects. Since MDSCs have specific molecular features, and only a tiny amount exists in physiological conditions, MDSC‐targeted therapy has become a promising research direction for tumor treatment with minimal side effects. In this review, we briefly introduce the classification, generation and maturation process, and features of MDSCs, and detail their functions under various circumstances. The present review specifically demonstrates the environmental specificity of MDSCs, highlighting the differences between MDSCs from cancer and healthy individuals, as well as tumor‐infiltrating MDSCs and circulating MDSCs. Then, we further describe recent advances in MDSC‐targeted therapies. The existing and potential targeted drugs are divided into three categories, monoclonal antibodies, small‐molecular inhibitors, and peptides. Their targeting mechanisms and characteristics have been summarized respectively. We believe that a comprehensive in‐depth understanding of MDSC‐targeted therapy could provide more possibilities for the treatment of cancer.
Keywords: inflammation, MDSC‐targeted drugs, myeloid‐derived suppressor cells, targeted therapy, tumor microenvironment
Myeloid‐derived suppressor cells play a crucial role in tumor progression and metastasis through their immunosuppressive effects. In different environments and conditions, these cells vary in mechanisms of immunosuppressive functions. Since only a small amount exists in physiological conditions, myeloid‐derived suppressor cell‐targeted therapy has become a promising research orientation for tumor treatment with minimal side effects. In recent years, various kinds of targeted drugs against myeloid‐derived suppressor cells have been developed, as a novel way of cancer treatment.

1. INTRODUCTION
The immune system is closely associated with the general health of humans who withstands exogenic and endogenic stimuli and damage via immune cells and related signaling pathways. Various diseases develop along with immune system dysfunctions. In steady‐state conditions, myeloid precursors in the bone marrow eventually differentiate into monocytes and neutrocytes, maintaining a normal and well‐balanced immune status. However, under some pathological processes, such as tumor progression, myeloid cells can abnormally differentiate into immature cells named myeloid‐derived suppressor cells (MDSCs). 1
MDSC, a kind of cell that mainly accumulates in a few situations, such as cancer and chronic inflammations, 2 plays important roles in immunosuppression, angiogenesis, tumor drug resistance, and tumor metastasis in the tumor microenvironment (TME). 3 MDSCs can be simply classified into two types based on their precursor cells’ features:granulocytic/polymorphonuclear MDSCs (PMN‐MDSCs) and monocytic MDSCs (M‐MDSCs). 4 While they share some molecular features and most functions, these two cell types also have their unique properties. For example, CD14 and CD15 expression differs between these types, which is a key feature for distinguishing them. 5
In tumor progression, MDSCs can be generated and activated via complex molecular mechanisms and then recruited into the TME to perform protumoral functions, which is modulated mainly by CXC motif chemokine receptor (CXCR) 2. 6 These cells primarily play immunosuppressive roles by affecting T cells, including suppressing T cell functions and promoting their apoptosis. 6 , 7 In addition, MDSCs also express immunosuppressive and trophic factors, including vascular endothelial growth factor (VEGF), interleukin (IL) 1/2/10, transforming growth factor‐β (TGF‐β), programmed death‐ligand (PD‐L) 1/2, indoleamine 2,3‐dioxygenase (IDO), inducible nitric oxide synthase (iNOS), and arginase‐1 (ARG‐1), to assist in immunosuppression, angiogenesis, and tumor progression. 6
Since MDSCs have multiple mechanisms promoting tumor progression, MDSC‐targeted therapy appears to be a promising treatment for malignant tumors. Another notable peculiarity of MDSCs is that they are present in small quantities under healthy and physiological conditions. Therefore, therapy targeting MDSCs may reduce damage to normal tissues, side effects are expected to be averted. 1 In recent, MDSC‐related targeted therapy has become an anticipated direction. Based on molecular structures, MDSC‐targeted drug therapies can be categorized into three groups: monoclonal antibodies (mAbs), small‐molecular inhibitors, and peptides. Some of these drugs have already been approved for clinical use in malignant tumors. For example, sunitinib, a receptor tyrosine kinase inhibitor (TKI), has been reported to deplete MDSCs in both peripheral blood and tumor sites. 8 , 9 mAbs with common target sites (e.g., programmed cell death 1 [PDCD1/PD‐1]/PD‐L1, cytotoxic T‐lymphocyte antigen 4 [CTLA‐4]) also negatively regulate MDSCs. 10 , 11 , 12 In addition, with their high biocompatibility and convenience, tumor‐targeting peptides have become one of the most advanced and promising areas in tumor‐targeted therapies. These peptides can either suppress tumors directly or function as accurate carriers for antitumor drugs by recognizing and combining specific receptors; some also show anti‐MDSC effects. 13
This review first presents the essential characteristics of MDSCs to briefly introduce them. Then, it describes the functional features of MDSCs in various conditions and classifies them based on their molecular mechanisms. This review mainly highlights the environmental specificity of MDSC, including the differences between tumor‐infiltrating and circulating MDSCs and between MDSCs from patients with cancer and healthy donors. The following section summarizes and emphasizes MDSC‐targeted drugs and therapies, hoping to provide researchers and clinical workers with guidance and insights into a new target in cancer therapy.
2. FEATURES OF MDSCs
2.1. Classification of MDSCs
In the bone marrow, myeloid cells can be briefly classified into two groups (granulocytes and mononuclear cells), which primarily contribute to the body's immune reaction. MDSCs are pathologically and immaturely differentiated from myeloid precursors (granulocytic and monocytic cell lines) and are divided into the two main populations as mentioned above (PMN‐MDSCs and M‐MDSCs). 4 In particular, the phenotypic characteristics of these two MDSC types are fundamentally analogous to those of neutrophils and monocytes, respectively. 14 Furthermore, a small proportion of myeloid precursors equipped with MDSC characteristics are considered early MDSCs. 15
2.2. Origination and maturation of MDSCs
Myeloid cells in the bone marrow differentiate into mature neutrophils and monocytes in response to acute inflammatory stimulations, such as pathogenic invasion. Those differentiating into monocytes subsequently generate macrophages and dendritic cells (DCs) to provide protection and manifest a proinflammatory activity. 16 However, these myeloid cells are observed to be activated immaturely in chronic pathological states, generating MDSCs. 17 , 18 Myeloid cell activation signals differ in the development of different pathogenic states. Persistent stimuli‐related myeloid cell activations are characterized by long‐lasting signals, comprising growth factors and inflammatory cytokines. 1
MDSC accumulation from myeloid progenitor cells in vivo comprises two stages: immature myeloid cell expansion and pathological activation. 19 The first stage is regulated by the granulocyte‐macrophage colony‐stimulating factor (GM‐CSF), granulocyte colony‐stimulating factor (G‐CSF), macrophage colony‐stimulating factor, stem cell factor, and some other chronic inflammatory or tumor‐related factors. These molecules induce expansion via signal transducer and activator of transcription (STAT) 3, interferon regulatory factor 8 (IRF8), CCAAT enhancer‐binding protein beta (CEBPB), retinoblastoma 1 (RB1), and Notch pathways. 20 Various studies have shown the essential role of STAT3—an inducer of immunosuppressive regulation during MDSC expansion. 21 However, STAT3 is only the regulatory factor of PMN‐MDSCs. While the population decline of PMN‐MDSCs is strongly correlated with the inhibition of STAT3 in the TME, the accumulation of M‐MDSCs is not relevant to the state of STAT3. 22
In the second stage, besides inflammation‐promoting factors, such as IL‐1β and tumor necrosis factor (TNF)‐α, and some other mediators, MDSCs can also be activated by a specific highly conserved mechanism—endoplasmic reticulum stress. 20 Most cells, including malignant cells, can barely survive under misfolded protein accumulating in the endoplasmic reticulum, mainly due to lower pH, higher free levels of radicals, and hypoxia in the TME. However, the unfolded protein response can be subsequently activated in MDSCs, as shown by the activation of inositol‐requiring enzyme 1, pancreatic endoplasmic reticulum kinase, and activating transcription factor 6. 23 , 24 The cascade of reactions induced by these molecules can promote MDSC differentiation, accumulation, and functioning, 23 partly reversing the inhibitory effect of the adverse environment on tumor progression.
The immature differentiation of granulocytes in the bone marrow usually leads to the generation of PMN‐MDSCs. However, Mastio et al. showed that a branch of cells from the monocytic lineage could differentiate into PMN‐MDSCs. 25 These precursors are named “monocyte‐like precursors of granulocytes” and can be detected only in the presence of a tumor. The downregulation of RB1 regulates the generation of this specific branch of myeloid cells. 25 IRF8, which promotes granulocyte‐derived PMN‐MDSC expansion, barely affects the expansion of monocyte‐like granulocyte precursors. 25 However, additional features of these monocyte‐derived PMN‐MDSCs must be elucidated.
2.3. Phenotypic characteristics of MDSCs
Different MDSC subtypes in mice share the same molecular features as CD11b+ Gr‐1+. Movahedi et al. 26 first distinguished and purified M‐MDSCs and PMN‐MDSCs using the marker of Ly6G maker. Further studies refined the features of MDSCs in detail, showing that they have entirely different characteristics in mice and humans, with the subpopulation of MDSCs showing much heterogeneity. PMN‐MDSCs are described as CD11b+ Ly6G+Ly6Clow and M‐MDSCs as CD11b+ Ly6G− Ly6Chigh 27 in mice. And CD49d expresses mainly in murine M‐MDSCs rather than murine PMN‐MDSCs. 28 The Gr‐1 molecule does not exist in humans. In humans, PMN‐MDSCs and M‐MDSCs are both CD11b+CD33+, but have discrepant CD14 and CD15: while PMN‐MDSCs and neutrophils are CD11b+CD14−CD15+ CD66b+ CD33+, M‐MDSCs and monocytes are CD11b+CD14+CD15− CD66b− CD33+ 5. However, density gradient is a distinguishing indicator of PMN‐MDSCs and neutrophils, 5 and M‐MDSCs and monocytes can be distinguished by the status of major histocompatibility complex (MHC) class II molecule expression (i.e. human leukocyte antigen [HLA]‐DR). 29
In the TME, more features of MDSCs with specificity have been identified. Lectin‐like oxidized low‐density lipoprotein receptor 1 (LOX1), a kind of transmembrane glycoprotein with a mass of 50 kDa, is defined as a specific marker of PMN‐MDSCs, 30 , 31 and another specific marker is fatty acid transport protein 2 (FATP2). 32 PD‐L1 is primarily expressed in M‐MDSCs, functioning as one primary approach for inducing immune regulation. 3 In addition, a single‐cell transcriptomics analysis showed that CD84+ cells in tumor tissues could be marked as MDSCs. 33 Highly specific molecules can function as markers of MDSCs and serve as promising targets for cancer immunotherapy (Figure 1). Several other literatures have also reviewed the surface markers of human and murine MDSCs in detail, 4 , 34 providing references of high‐quality for MDSC distinguishment.
FIGURE 1.

Main surface markers of monocytic myeloid‐derived suppressor cells (M‐MDSCs) and polymorphonuclear myeloid‐derived suppressor cells (PMN‐MDSCs) in mice and human. The green area represents mouse M‐MDSCs, the yellow area represents mouse PMN‐MDSCs, the blue area represents human M‐MDSCs, and the red area represents human PMN‐MDSCs. The gray surface markers represent the shared markers of both MDSC subtypes; the blue ones represent the marker only shown in M‐MDSCs; while the orange ones are indicated as the unique markers of PMN‐MDSCs.
Notably, the current definition and classification methods for MDSC subsets are based on the immune suppressive functions and surface markers. However, MDSCs have more complex heterogeneity and plasticity, and a more integrated definition and classification of MDSCs should focus on the differences in immunosuppressive mechanisms (e.g., stimulating factors and impacted pathways/cells). 35
3. ENVIRONMENTAL SPECIFICITY OF MDSCs
As previously mentioned, MDSCs rarely exist under healthy conditions in humans. Under tumor progression, MDSCs can form during tumor progression, leading to their expansion and discrepant quality. Therefore, it is essential to find out how MDSCs differ in different sites so that they can be targeted more accurately. Since the differences between mature myeloid cells and MDSCs and between MDSC subpopulations have already been detailed in other reviews, 4 , 35 our review focuses on the discrepancies in environmental factors that arise.
3.1. Differences between MDSCs from patients with cancer and healthy donors
The number of MDSCs was significantly larger in patients with cancer than in healthy donors. 36 , 37 The proportion of circulating PMN‐MDSCs is larger in most patients with cancer than that in healthy individuals, indicating that the largest expansion occurs in PMN‐MDSCs. 38 The expression of LOX1 in PMN‐MDSCs and PD‐L1 in M‐MDSCs is not significantly higher in patients with tumors than in healthy populations, 38 and the quantity of MDSCs is closely related to the clinical cancer stage and tumor metastasis in colorectal carcinoma. 37 These findings indicate that the number of MDSCs is associated with the strength of immunosuppressive activities. Regarding immunosuppression, Zhang et al. 37 found that MDSCs from healthy donors could not inhibit the division of autologous T cells, unlike those from patients with tumors. MDSCs from patients with cancer express higher ARG‐1, IDO, IL10, and iNOS expression was higher in MDSCs from patients with cancer than from healthy donors. 39 , 40 While M‐MDSCs produced more PD‐L1, PMN‐MDSCs produced more C‐C motif chemokine receptor 5 (CCR5). 41 Briefly, the transition from a steady state to cancerous state results in MDSC expansion and the enhancement of their immunosuppressive functions (Figure 2).
FIGURE 2.

The differences of myeloid‐derived suppressor cells (MDSCs) under various environments. The descriptions in the left frame demonstrate the features of tumor‐infiltrating MDSCs compared with circulating MDSCs. The descriptions in the right frame display the features of MDSCs from cancer patients compared with MDSCs from healthy donors. Cat2, cationic amino acid transporter 2; Arg‐1, arginase‐1; Nos2, nitric oxide synthase 2; PD‐L1, programmed death ligand 1; STAT3, signal transducer and activator of transcription 3; MAPK, mitogen‐activated protein kinase; CCL, C‐C motif chemokine ligand; IDO, indoleamine 2,3‐dioxygenase; IL‐10, interleukin‐10; iNOS, inducible nitric oxide synthase; CCR, C‐C motif chemokine receptor; PGE2, prostaglandin E2; AA‐PEox, arachidonic acid‐poly (ethylene oxide).
3.2. Differences between tumor‐infiltrating and circulating MDSCs
3.2.1. Differences in molecular phenotype
MDSCs primarily form in the bone marrow but can also be induced by tumor‐related cytokines in peripheral blood from peripheral blood myeloid cells. The phenotypic characteristics of MDSCs vary in the tumor site and peripheral blood in patients with different tumors. In patients with non‐small‐cell lung cancer (NSCLC), PMN‐MDSCs obtained from tumor tissue were CD11b+HLA−DR−/lowCD14−CD15+, 41 while ILT3high can be acquired from peripheral blood. 42 Similarly, M‐MDSCs in NSCLC sites are CD11b+ HLA‐DR−/low CD14+ CD15− 41 ; however, circulating M‐MDSCs can be featured as CD16low. 43 Discrepancies also occur in other tumors, 37 although many studies have found identical MDSC phenotypes in tumor tissue and peripheral blood. 44 , 45 Therefore, the phenotypes of MDSCs can vary in different tissues; however, resemblance among different tumors could barely be noted, partly because the phenotypes of MDSCs in different studies are diverse.
3.2.2. Differences in quantity and subtype proportion
In some cancer types, there are more tumor‐infiltrating MDSCs than MDSCs in peripheral organs. 41 , 46 Since M‐MDSCs have stronger immunosuppressive activities, the proportion of these cells in total MDSCs has gained increasing attention. In tumors, PMN‐MDSCs predominate the circulating MDSC population, while the M‐MDSC/PMN‐MDSC ratio increases in the TME (Figure 2). 47 , 48
3.2.3. Differences in molecular expression and function
Tumor‐infiltrating MDSCs have stronger immunosuppressive activities than circulating MDSCs. This difference can be attributed to the functional differences in the total number of MDSCs and the M‐MDSC/PMN‐MDSC ratio. Furthermore, on a per‐cell basis, tumor‐infiltrating MDSCs are more likely to be a suppressor. MDSCs in prostate cancer tissues can upregulate cationic amino acid transporter 2 (CAT2), ARG‐1, and nitric oxide synthase 2 (NOS2) expression, decreasing l‐arginine production, increasing nitric oxide (NO) production, and enhancing the detrimental effects on T cells. 49 Hypoxia‐inducible factor 1 subunit alpha (HIF1α) expression increases in the tumor environment due to hypoxia, enhancing PD‐L1 expression in MDSCs. 41 Kumar et al. 50 considered phosphorylated STAT3 the key factor inducing MDSCs to differentiate into tumor‐associated macrophages (TAMs), reporting that tumor‐infiltrating MDSCs had a lower phosphorylated STAT3 level to retain their immature state. Our previous study noted higher basal mRNA levels and protein levels of TNF‐alpha‐induced protein 3 (TNFAIP3/A20) increased in tumor‐infiltrating MDSCs than in MDSCs from the spleen. 51 We also observed an upregulation of the mitogen‐activated protein kinase (MAPK) pathway in tumor‐infiltrating MDSCs, especially the extracellular signal‐regulated protein kinase (ERK)‐related pathway. 47 Tumor‐infiltrating M‐MDSCs in lymphoma and melanoma can induce high C‐C motif chemokine ligand (CCL) 3, CCL4, and CCL5 expression to recruit regulatory T cells (Tregs) via chemokine–CCR5 interactions (Figure 2). 52
Tumor‐infiltrating PMN‐MDSCs but not M‐MDSCs or circulating PMN‐MDSCs were recently found to induce spontaneous ferroptosis, one basis of their immunosuppressive ability. Ferroptosis is the dominating cell death pattern in tumor‐infiltrating PMN‐MDSCs, causing tumor progression. 53 Unlike circulating PMN‐MDSCs, tumor‐infiltrating PMN‐MDSCs induced the upregulation of ferroptosis‐related genes and the accumulation of oxidized phosphatidylethanolamine containing arachidonic acid, one of the main characteristics of ferroptosis, and prostaglandin E2 (PGE2). The induction of ferroptosis, which may partly underlie the downregulation of hypoxia‐related glutathione peroxidase 4 (GPX4) in the TME, gives polymorphonuclear cells immunosuppressive activity, restraining the proliferation of CD8+ T cells. Consequently, ferroptosis convert polymorphonuclear cells into PMN‐MDSCs. Conversely, suppressing ferroptosis in tumor‐infiltrating PMN‐MDSCs activates antitumor immunity, such as neutrophil‐related immunity and antigen processing/presentation, which turns PMN‐MDSCs back into classical polymorphonuclear cells. 53 Ferroptosis inhibition in tumor‐infiltrating PMN‐MDSCs downregulates ferroptosis‐associated lipid signals (FALIS) and PGE2 expression in TAMs, leading to their dysfunction. Substantial increases occur in memory and effector CD8+ T cells and natural killer (NK) cells due to upregulation of IL‐2‐ and TNF‐α‐associated inflammatory signaling and suppression of oxidative phosphorylation. 53 Eventually, ferroptosis inhibition results in the regeneration of antitumor immunity (Figure 3).
FIGURE 3.

The immunosuppressive effects of ferroptosis in tumor‐infiltrating polymorphonuclear myeloid‐derived suppressor cells (PMN‐MDSCs). Ferroptosis occurs spontaneously in tumor‐infiltrating PMN‐MDSCs. When classical PMNs are influenced by hypoxia, FATP2 upregulation, or ferroptosis inducers, they may transform to PMN‐MDSCs and obtain the ability of immunosuppression. Tumor‐infiltrating PMN‐MDSCs were observed upregulating ferroptosis feature genes, gathering AA‐PEox, PGE2, and FATP2. The downregulation of GPX4 is also observed, which restrains the proliferation and activation of CD8+ T cells. In addition, TAMs are activated with the upregulation of the expressions of FALIS and PGE2. PGE2, prostaglandin E2; FALIS, ferroptosis‐associated lipid signals; TAM, tumor‐associated macrophage; AA‐PEox, arachidonic acid‐poly (ethylene oxide); FATP2, fatty acid transport protein 2; GPX4, glutathione peroxidase 4.
3.3. Differences between artificially induced circulating MDSCs and noninduced circulating MDSCs
Some studies have artificially induced circulating MDSCs using cytokines, such as GM‐CSF, IL‐1, IL‐6, VEGF, TNF‐α, and PGE2, which differ from noninduced MDSCs. 54 Compared with noninduced circulating M‐MDSCs, cytokine‐induced M‐MDSCs have low expression of S100 calcium‐binding protein A9 (S100A9), a chemokine strategic for MDSC migration, 55 but demonstrate good performance against T cell proliferation. 56 Generated peripheral blood MDSCs have various immunosuppressive activities via different inducing factors; processing peripheral blood myeloid cells with the combination of GM‐CSF and IL‐6 is one of the most effective ways. In Roussel's study, the proportion of MDSCs increased more than fivefold after induction, 55 and in Lechner's study, the generated MDSCs inhibited T cell proliferation by 80%. 54 Their increased immunosuppressive activity was closely associated with the upregulation of iNOS, NADPH oxidase 2 (NOX2), VEGF, TGF‐β, and PD‐L1. 54
3.4. Differences between MDSCs from patients with tumors and premalignant conditions
Circulating MDSCs in patients with cancer and premalignant conditions share the same composition and function but with larger amounts in cancer environments. 57 This finding suggests that MDSCs in both cancerous and precancerous lesions can be targeted using similar drugs. T cell proliferation and interferon‐γ (IFN‐γ) production return to normal after MDSC elimination in precancerous, 57 , 58 but not in cancerous environments. 59 Therefore, targeting MDSCs in precancerous lesions is an effective way to reduce cancer risk.
4. FUNCTIONS OF MDSCs
MDSCs play a vital immunosuppressive role in various pathological processes, especially in all kinds of tumors and TMEs. Herein, we orderly introduce the specific functions of MDSCs in a physiological situation, inflammation, and tumor progression.
4.1. Functions of MDSCs in healthy conditions and inflammation
4.1.1. Functions of MDSCs in healthy conditions
Bone marrow‐derived myeloid cells usually differentiate into mononuclear cells and granulocytes. 60 With their small quantity and modest functions, MDSCs rarely show immunosuppressive effects under physiological homeostasis conditions. However, there are exceptions: pregnancy and neonates. Since a fetus is a type of allograft in a mother, preventing its rejection, or maternal‐fetal tolerance, is vital. 61 , 62 One study observed SR‐E1+ PMN‐MDSCs in all stages of pregnancy, which correlated negatively with intrauterine growth retardation. 63 In this process, MDSCs inhibit T cell, T helper cell, and NK cell function by producing reactive oxygen species (ROS), expressing ARG‐1, or functioning through cell contact. Moreover, MDSCs appear in neonates for a transient period. 64 These cells may originate from cord blood 65 or be transported via breast milk 66 and participate actively in inflammation regulation. 67
4.1.2. Functions of MDSCs in infections
The roles of MDSCs in chronic and acute infectious diseases have been elucidated in recent years, whose immunosuppressive ability promotes the progression of these infectious diseases, and the detailed mechanisms of MDSCs acting on individual diseases were summarized by several reviews. 68 , 69 , 70 In brief, to enhance the infectious effects, MDSCs commonly produce immunosuppressive metabolites via iNOS and ARG‐1, and increase the expressions of immunosuppressive molecules like IL‐10, ROS, STAT3, and so on, mainly to prevent immune cell proliferation and activation, especially T cells. 71 , 72 , 73 , 74 , 75 For instance, in the chronic infectious condition induced by Staphylococcus aureus (S. aureus), a large number of MDSCs were found in the surrounding bacterial biofilm, contributing to the persistence and low clearance of S. aureus, by secret iNOS, ARG‐1, as well as IL‐10 to inhibit the infiltration of immune cells 76 ; in turn, stimulated by MDSCs, S. aureus will upregulate formate acetyltransferase (pflB) to form a larger and more pervasive biofilm. 77 Apart from the common mediatory mechanisms, MDSC can induce high‐level ER stress to enhance its immunosuppressive activity, therefore leading to the promotion of leprosy and tuberculosis. 78 , 79 During sepsis or some parasite infections like Schistosoma japonicum, MDSCs are also able to increase the expression of PD‐L1, modulating the immunoactivity of T cells via the PD‐1/PD‐L1 pathway. 80 , 81
In virus infections, since the acquired immune deficiency syndrome is characterized by immune deficiency, the role of MDSCs after human immunodeficiency virus (HIV) invasion has been widely explored. For one thing, MDSCs will inhibit the proliferation of CD4+ and CD8+ T cells while promoting the expansion of Tregs and HIV infection. 82 , 83 For another, however, they are beneficial to inhibit the persistent immune activation and the accompanying chronic inflammations induced by HIVs. 84 Similarly, in severe COVID‐19 patients, IL‐8 will induce the transitory recruitment of PMN‐MDSCs in peripheral blood to help the patients’ recovery 85 to fight against hyperinflammation. Therefore, the immunosuppressive activity of MDSCs can also have positive effects during virus infections.
4.1.3. Functions of MDSCs in autoimmune diseases
Rheumatoid arthritis (RA) is a systemic, cartilage‐and‐bone‐destructive autoimmune disease of which immune cell infiltration and accumulation in the synovial joints have a vital role in the pathogenesis, 86 , 87 , 88 and MDSCs possess immunosuppressive functions to alleviate the worsening of RA. MDSC accumulations were found in synovial fluid, lymph nodes, peripheral blood, as well as spleen of RA patients. 89 , 90 , 91 They inhibit CD4+ T cell proliferation (via ROS, NO, IFN‐γ generation, etc.) and infiltration and the proinflammatory cytokine production by CD4+ T cells, restrict B cell proliferation and antibody production (via NO, PGE2 production), promote Treg cell proliferation (via IL‐10 generation), as well as stimulate IL‐10+ Breg cell generation (via exosomal PGE2 secretion). 90 , 92 , 93 , 94 , 95 Although MDSCs display obvious anti‐inflammatory capacities in autoimmune diseases, they also surprisingly play some proinflammatory roles. 90 , 91 In RA, with the progression of disease, the number of MDSCs also expands, which is negatively related to the number of T helper 17 cells in peripheral blood. 96 , 97 The research found that MDSCs could stimulate T helper 17 cell differentiation by producing IL‐1β, reprograming T helper 17 cells into pro‐osteoclastogenic phenotype, promoting osteoclast differentiation to aggravate RA severity. 97 , 98 , 99 This kind of uncharacteristic, proinflammatory effect of MDSCs has been also demonstrated in other autoimmune diseases. 100 The abnormal activation of Notch1 signaling plays an important role in the pathogenesis of systemic lupus erythematosus (SLE), which also promote the MDSC differentiation process. 101 In the peripheral blood of SLE patients, the frequency of MDSCs increase with the disease severity, and MDSCs promote T helper 17 cell differentiation via the activation of ARG‐1, contributing to SLE‐related renal injury. 100 The activation of ARG‐1/miR‐322‐5p/TGF‐β pathway can also change the T helper 17 cell/Treg cell ratio in SLE. 102 In experimental autoimmune encephalomyelitis (EAE), similarly, spleen‐derived MDSCs facilitate T helper 17 cell differentiation in an IL‐1β‐dependent manner, 103 while lung‐derived MDSCs generate IL‐6 to achieve the same purpose. 104 In brief, MDSC is a double‐edged sword during autoimmune disease progression, but its immunomodulatory mechanisms in different stages of disease still need further exploration. Apart from its function of affecting immune microenvironment, MDSC was discovered to promote oligodendrocyte precursor cell maturation and remyelination by secreting osteopontin in EAE models, 105 indicating that MDSC can not only be regarded as an immune cell but also has potential in neural system regeneration.
4.1.4. Functions of MDSCs in organ transplantation
As a kind of therapeutic tool, MDSCs can be used for immunosuppression in organ transplantation for beneficial utilization. Acute graft‐versus‐host disease (GVHD) is the most substantial threat among organ transplant recipients, and MDSCs play a positive role in transplantation immune tolerance, reducing the incidence of GVHD. 106 , 107 , 108 Tobias et al. 109 found that the level of PMN‐MDSCs increases in stable lung transplant recipients (52.1% when that of the control group reaches 9.4%) compared with that in patients with infection (28.2%) and chronic lung allograft dysfunction (33.0%). Okano et al. 110 added MDSCs in a coculture with donor‐reactive T cells and donor intestinal epithelium, achieving an increased organ survival rate. Another study among 38 kidney transplant patients concluded that the level of M‐MDSCs increased at 6 and 12 months after transplantation. 111
4.1.5. Functions of MDSCs in anti‐inflammatory therapy
Another possible direction is to apply MDSCs in anti‐inflammatory therapy. Various substances can induce MDSC production in vivo or in vitro (Table 1). Xu et al. 112 identified mitogen‐activated protein kinase kinase kinase 8 (MAP3K8/Tpl2), a protein kinase acting as a pivotal mediator of MDSC recruitment in fulminant hepatitis. When patients develop fulminant hepatitis, their IL‐25 level increases, activating the signal pathway mediated by IL‐17 receptors A/B (IL‐17RA/RB) and Tpl2, leading to the expression of CXC motif chemokine ligands (CXCLs) 1/2. Then, the increased CXCL1/2 expression stimulates the recruitment of MDSCs, promoting inflammation inhibition. Another study showed that artepillin C in green propolis attenuates allergic asthma by inducing M‐MDSCs. 113 Studies have found an augmented frequency of MDSCs in both in vitro and in vivo experiments. Moreover, artepillin C‐treated mice had a lower frequency of eosinophils in their bronchoalveolar lavage fluid, lower lung inflammation score, and lower mucus score. Propolis therapy may also positively affect PMN‐MDSC recruitment. 114 However, further research on the exact mechanism is needed. In heart failure models, MDSCs also participated in myocardial protection. In a recent study, pharmaceutical depletion of MDSCs significantly increased inflammation and pathological cardiac remodeling. 115
TABLE 1.
Different substance inducing myeloid‐derived suppressor cells (MDSCs) in various models.
| Study | Year | In vivo or in vitro | Substance inducing MDSC | Model | References |
|---|---|---|---|---|---|
| Lechner et al. | 2010 | In vitro | IL‐6 | Healthy donor peripheral blood mononuclear cell | 54 |
| 2010 | In vitro | GM‐CSF | Healthy donor peripheral blood mononuclear cell | ||
| Morales et al. | 2010 | In vitro | GM‐CSF |
Murine bone marrow CD11b‐Gr1‐ cells |
116 |
| Chen et al. | 2013 | In vitro | GM‐CSF, IL‐4, and CpG oligodeoxynucleotide | Murine bone marrow cell | 117 |
| Liu et al. | 2015 | In vitro | IL‐6, GM‐CSF, and norepinephrine | Healthy donor peripheral blood mononuclear cell | 118 |
| Yang et al. | 2019 | In vitro | M‐CSF and IFN‐γ | Murine bone marrow cell | 119 |
| Scheurer et al. | 2021 | In vitro | GM‐CSF | Murine bone marrow cell | 120 |
| Wang et al. | 2021 | In vitro | M‐CSF and phorbol 12‐myristate 13‐acetate | Murine bone marrow cell | 121 |
| Sarra et al. | 2013 | In vivo | IL‐25 | d‐Gal/lipopolysaccharide (LPS) induced fulminant hepatitis mice | 122 |
| Zhou et al. | 2018 | In vivo | Rapamycin | Heart failure patients; isoproterenol infusion/transverse aortic constriction mice; induced heart failure mice | 115 |
| Xu et al. | 2019 | In vivo | Tpl2 | Propionibacterium acnes/LPS induced fulminant hepatitis mice | 112 |
| Martins et al. | 2021 | In vivo and in vitro | Artepillin C |
In vitro M‐MDSC culture; Ovalbumin induced asthma mice |
113 |
As mentioned above, MDSCs can be used when treating inflammation‐related diseases in multiple organs in three ways: first, endogenous MDSCs are able to be increased by inducing their formation or inhibiting continued differentiation. For example, rapamycin can inhibit MDSC differentiation and improve heart failure. 115 Second, the immunosuppressive effects of MDSCs are expected to be improved. For example, cyclic helix B peptide can induce IL 7 receptor (ILR7/CD127)+ M‐MDSCs with a stronger immunosuppressive activity. 123 Third, exogenous MDSCs can be introduced via adoptive transfer, which has been proposed in organ transplantation; this method may also be applied in treating inflammatory diseases (Figure 4). 124 There are currently fewer studies in this area, and there may be a broad scope for development.
FIGURE 4.

Possible methods to enhance myeloid‐derived suppressor cell (MDSC)‐associated immunosuppression and the application of MDSC in anti‐inflammation therapy. ArtC, artepillin C; CHBP, cyclic helix B peptide; IL‐25, interleukin‐25; Tpl2, Tumor progression locus 2; CXCL, CXC motif chemokine ligand.
4.2. Tumor‐related immunosuppressive functions of MDSCs
4.2.1. Effects of MDSCs on T cells
The role of MDSCs in the TME mainly involves inhibiting of T cell activation. The direct effects of MDSCs are principally to reduce the expression of specific recognition receptors on the T cells’ surface and to promote T cell apoptosis. Their indirect effect is to regulate the T cells’ proliferation and differentiation by changing the microenvironment's physical and chemical components.
Direct effects on T cells
MDSCs can produce large amounts of ROS and reactive nitrogen species (RNS), which inhibit the activation of the Janus kinase (JAK) 3/STAT5 signaling pathway, downregulating the expression of MHC‐II molecules and inhibiting T cell apoptosis. 118 Meanwhile, ROS can also induce T cells to express Fas ligand (FASLG/FasL), which contributes to the activation of T cell apoptosis through a Fas cell surface death receptor (Fas)–FasL interaction. 125 PMN‐MDSCs mainly produce ROS and function by directly contacting T cells. 126 M‐MDSCs mainly produce RNS, impacting T‐cell apoptosis without directly contacting them since RNS are more stable than ROS. 127 Mionectic TME‐generated HIF‐1α can also promote PD‐L1 expression on the surface of PMN‐MDSCs, leading to T cell apoptosis. 128
Besides their impact on the death of proinflammatory T cells, MDSCs can also directly mediate T cells’ generation, differentiation, and migration of T cells. MDSCs inhibit the generation and proliferation of tumor antigen‐specific T helper 1 cells by producing IL‐6. 129 They also promote the differentiation of CD4+ T cells into Tregs by releasing of cytokines or cell‐to‐cell contact and Treg proliferation. 7 For example, through the TGF‐β1‐dependent pathways, MDSCs promote the generation of Tregs, and IDO overexpression by MDSCs causes Tregs migrate to tumor sites and lymph nodes. 130 Moreover, the NO produced by M‐MDSCs can react with superoxide to form peroxynitrite (ONOO–), which nitrifies T‐cell receptors and T‐cell‐specific chemokines to directly block T‐cell migration. 131 Zhao et al. 132 discovered that MDSCs could release the specific metabolite itaconate, derived from the tricarboxylic acid cycle, to repress the proliferation and the antitumor cytotoxicity of CD8+ T cells by decreasing aspartate, serine, and glycine synthesis and reducing the dominating aggressor granzyme and perforin. Itaconate can serve as a promising cancer therapy target and a tumor prognosis biomarker.
Indirect effects on T cells
M‐MDSCs can degrade arginine by secreting ARG‐1 and RNS. When arginine is competitively consumed in the microenvironment, the T‐cell‐receptor ξ chain and CD3 expression on T cells’ surface is downregulated, and the expression of cyclin D3 (CCND3) and cyclin‐dependent kinase 4 (CDK4) is inhibited, inducing T cell cycle arrest in the G0/G1 phase. 133 In addition, T cells cannot synthesize cysteine autonomously and only use cysteine released into the environment. PMN‐MDSCs use solute carrier family 7 member 11 (SLC7A11) to scramble for cysteine with T cells by transferring it into the cytoplasm, 118 inhibiting T cell activation, and promoting their apoptosis. Moreover, by producing IL‐1β, IL‐6, IL‐23, and PGE2, MDSCs can promote the differentiation and proliferation of T helper 17 cells. 134 The production of IL‐17 by T helper 17 cells inhibits the antitumor effect of T cells. 135 The specific immune checkpoint protein V‐domain immunoglobulin suppressor of T cell activation (VISTA) is crucial for regulating the effector functions of myeloid cells. By expressing VISTA, MDSCs can also participate in the negative regulation of T cell antitumor functions. 136
4.2.2. Effects of MDSCs on macrophages
The inhibitory effect of MDSCs on immune cells is also manifested in macrophages. MDSCs are involved in activating of TAMs and promoting the conversion of M1 to M2/TAM. 137 They transform macrophages in tumor‐bearing individuals primarily by increasing IL‐10 production and decreasing macrophage‐derived IL‐12 production. Sinha et al. 138 found that coculturing macrophages activated via lipopolysaccharide and IFN‐γ with MDSCs for 16 hours greatly downregulated their IL‐12 production. STAT3 has a unique function in controlling the differentiation of MDSCs into TAMs in the tumor environment. 50
While TAMs and MDSCs are regarded as independent entities, the boundary between them is unclearly demarcated, and they have many common features. 139 Tumor‐infiltrating MDSCs have the pleiotropic characteristics of M1 and M2 macrophages. 140 M‐MDSC‐derived macrophages retain the main characteristics of their precursors, including immunosuppressive activity. S100A9 promotes the M2 polarization of macrophages to a certain extent. 140 M‐MDSC‐derived macrophages retain S100A9 expression, but most monocyte‐derived macrophages or tissue‐resident macrophages do not. The difference in S100A9 expression indicates that the presence of S100A9 in macrophages is critical to its inhibitory activity.
4.2.3. Expression of immunosuppressive and trophic factors
MDSCs recruit suppressive immune cells by expressing TGF‐β, VEGF, fibroblast growth factor 2 (FGF2), MMP9, IL‐10, PD‐L1/2, IDO, iNOS, and ARG‐1 and establishing a microenvironment that suppresses host immunity by expressing suppressive cytokines (e.g., IL‐10 and TGF‐β) and surface molecules (e.g., PD‐L1 and PD‐L2). 141
VEGF and MMP‐9
The nonimmunosuppressive functions of MDSCs include tumor angiogenesis and metastasis. 142 MDSCs can express VEGF and basic FGF (bFGF) to help tumor cells build new vascular systems and deliver nutrients and oxygen to cancer cells. 143 VEGF is a key angiogenic factor in tumors and participates in the initial stage of tumor development, progression, and metastasis. 144 It stimulates vascular endothelial cells’ proliferation and survival, increases vascular permeability, and recruits vascular progenitor cells from the bone marrow, promoting tumor angiogenesis. 145
PMN‐MDSCs can secrete proangiogenic factors. 143 With their ability to activate the p38 MAPK and ERK1/ERK2 signaling pathways, PMN‐MDSCs can upregulate VEGF and CXCR expression in tumor cells, 146 inducing proangiogenic effects and facilitating angiogenesis. 147 Coinoculation of M‐MDSCs with tumor cells promoted TME microvessel formation and vascular leakage more than tumor cells alone. 148
Notably, unlike the formation of blood vessels under physiological conditions, tumor blood vessel formation has a disorganized structure and a discontinuous and incomplete vessel wall. 55 MDSCs secrete matrix metalloproteinases (MMPs), which degrade the extracellular matrix and thus promote angiogenesis. 149 MMP‐9 activity is required to promote angiogenesis. 143 It disrupts the basement membrane and alters the local microenvironment, 150 promoting tumor progression and enabling tumor cells to cross the extracellular matrix into the peripheral circulation and become circulating tumor cells (CTCs). VEGF upregulates the activity of NK cells and their natural immune response and inhibits DC differentiation and development, 151 suggesting its role in CTCs immune escape. While direct stimulatory effects of VEGFA have not been reported, the VEGFC–VEGF receptor (VEGFR) 3 axis has been reported to promote megakaryopoiesis and increase the platelet count, thereby further protecting CTCs. 144
In addition, VEGF secreted by MDSCs directly effects on cancer cells or tumor stem cells. It may induce cancer cell proliferation and promote tumor‐initiated cell self‐renewal by activating VEGFR1 signaling. 145 VEGF may also exert a proinflammatory cytokine release effect through VEGFR2/STAT3 signaling, inducing increased IL‐6, IL‐8/CXCL8, and growth‐regulated protein‐α/CXCL1 secretion by endothelial cells (nonleukocytes) and exerting an immunosuppressive effect.
TGF‐β
TGF‐β is secreted by various tumor cells and is closely associated with tumor growth and immunity. 152 Inhibition of TGF‐β, PD‐L1, and LAIR‐1 was recently reported to effectively control the growth of collagen‐rich mouse MC38 colon cancer and EMT6 breast cancer, leading to tumor cure and long‐term tumor‐specific protection. 153 In the TME, dysregulated TGF‐β signaling suppresses antitumor immunity and promotes cancer fibrosis, epithelial–mesenchymal transition (EMT), and angiogenesis. TGF‐β is an autocrine survival signal for myeloid precursor cells and drives their differentiation into highly immunosuppressive MDSCs at the expense of macrophages and DCs. 154
MDSCs induce EMT in tumor cells via the TGF‐β signaling pathway. They can secrete multifunctional proteoglycans to induce EMT in cancer cells via the TGF‐β signaling pathway, 155 promoting tumor invasion and metastasis. 156 PMN‐MDSCs preferentially accumulate in primary tumors where they induce cancer cells to undergo EMT, facilitating the rapid acquisition of a mobile phenotype by cancer cells and leading to multinodal growth, systemic spread, and distant metastasis of the primary tumor. 157 In the TME, dysregulated TGF‐β signaling suppresses antitumor immunity and promotes tumor fibrosis, EMT, and angiogenesis. 154
TGF‐β is also involved in downregulating the expression of the killer cell lectin‐like receptor K1 (KLRK1/NKG2D) on the NK cell surface, 148 inhibiting their production of cytokines, such as IFN‐γ, CCL5, and protein 1 alpha (MIP1α/CCL3).
IL‐10 and IL‐6
MDSCs induce the differentiation of regulatory B cells and regulatory DCs 158 by secreting IL‐10. In addition, IL‐10 inhibits the activation and proliferation of NK cells, NK T cells, and DCs and the production of various antitumor cytokines (e.g., IL‐12), 159 attenuating the immune response in the TME and facilitating tumor growth and spread of tumors. Tumor cell‐derived IL‐10 can promote autonomous cell growth and immune escape in diffuse large B cell lymphomas, and neutralizing IL‐10 signaling can reduce tumor growth. 160 MDSCs secrete IL‐6, IL‐10, and IL‐23, which also inhibit the activation and proliferation of NK cells and NK T cells. 161 Conversely, IL‐6 acts as a classical agonist to activate the JAK/STAT3 pathway and induce the expression of the MYC proto‐oncogene bHLH transcription factor (MYC/c‐Myc). 162 The upregulation of STAT3 by IL‐6 also promotes PD‐L1 and ARG‐1 expression and NO and ROS production, increasing MDSC function. 163 IL‐6 can also recruit circulating MDSCs into the TME. 164
S100A8 and S100A9
MDSCs also secrete chemokines, such as S100 calcium‐binding proteins A8 (S100A8) and S100A9, which are actively transported to the TME and distant tissues via exosomes secreted by tumor‐associated immune cells (e.g., MDSCs). 55 They further stimulate the release of inflammatory cytokines via Toll‐like receptor 4, 165 promote Ca2+ influx, immune cell recruitment, and inflammatory processes 166 and are involved in MDSC migration to tumor sites. After release, S100A8/A9 block the differentiation of MDSCs into DCs, leading to an increase in MDSCs, 150 further promoting MDSC accumulation and thus coordinating the formation of an immunosuppressive microenvironment conducive to tumor growth in premetastatic tissues. S100A9 acts as an endogenous natural ligand for CD33//SIGLEC3 and promotes Lin‐HLA‐DR‐CD33+ MDSCs to secrete cellular effectors, such as IL‐10, TGF‐β, NO, and ARG molecules that inhibit hematopoiesis and promote T cell tolerance as well as myelosuppression and inflammation. 167
CD39 and CD73
MDSCs also express ectonucleoside triphosphate diphosphate hydrolase 1 (ENTPD1/CD39) and 5’‐nucleotidase ecto (NT5E/CD73). CD39 catabolizes adenosine triphosphate to adenosine diphosphate/adenosine monophosphate (AMP), while CD73 mediates the subsequent catabolism of AMP to adenosine. Their expression and activity are elevated in tumor tissue and blood and are frequently associated with clinical signs of disease and poor prognosis in some cancers. 168 The accumulation of extracellular adenosine consequently activates the differentiation of M‐MDSCs into highly immunosuppressed TAMs and reduces MHC‐I and MHC‐II expression in tumor‐infiltrating DCs, 169 attenuating antigen‐specific T‐cell responses. Moreover, through an intracellular cascade reaction mediated by cyclic‐AMP, adenosine shifts the TME from cytotoxic T‐cell inflammation to immune tolerance, influencing the cytokines and cell morphology. 170
IL‐1
IL‐1 is an inflammatory cytokine that plays a key role in tumorigenesis and progression. It is a key mediator of natural and acquired immunities and is involved in multiple pathways related to microbial recognition, activation, and functional localization of lymphocytes. IL‐1 plays different roles in tumorigenesis and progression by influencing different components of the TME. 171 It mainly affects inflammatory processes but also has multiple immune, degradative, and progrowth properties. MDSCs secrete IL‐1 to enhance their immunosuppressive effects. 172 Immune cells can remove senescent cells from tumors or induce cancer cell senescence by secreting proinflammatory cytokines. MDSCs have been reported to interfere with the secretory phenotype associated with tumor senescence by secreting IL‐1 receptor antagonist (IL1RN/IL‐1RA) to fight senescence in a paracrine manner. Treatments that lower IL‐1RA levels can tilt IL‐1α toward IL‐1RA homeostasis, enhancing tumor senescence and improving the efficacy of chemotherapy. 173
IL‐1α/IL‐1 receptor signaling is located upstream of nuclear factor kappa B (NF‐κB), 174 enhancing NF‐κB transcriptional activity and thereby regulating PD‐1/ PD‐L1 expression. Malignant tumor cells suppress the immune function of T lymphocytes through pathways such as PD‐1/PD‐L1 pathway to weaken their recognition by the immune system and evade immune surveillance, achieving immune escape and prolonging the life span of tumor cells. NF‐κB also regulates TNF‐α and epidermal growth factor receptor (EGFR), forming a positive feedback regulatory loop between them to promote tumor cell proliferation. 175
Both IL‐1α and IL‐1β promote tumor angiogenesis and invasion, 172 but IL‐1β has a more prominent role in these processes. This difference may be attributed to the secretion of IL‐1β into the microenvironment, where it activates cells in the tumor mesenchyme, including malignant tumor cells. IL‐1β stimulates IL‐10 production by MDSCs 176 and induces the production of proangiogenic cytokines, such as IL‐8, by endothelial cells and surrounding stromal cells. 171
Altogether, MDSCs in the TME can induce immunosuppressive functions in various ways: affecting T cells’ generation, differentiation, migration, proliferation, and viability; inducing M2 polarization of macrophages; mediating the status of NK cells and DCs; recruiting other immunosuppressive cells; inhibiting self‐differentiation; and promoting tumor cells’ progression, EMT, and metastasis of tumor cells to construct a tumor‐promoting inflammatory environment (Figure 5).
FIGURE 5.

Immunosuppressive functions of myeloid‐derived suppressor cells (MDSCs). The effects of MDSCs on T cells include direct and indirect effects. The direct effects: MDSCs release ROS and RNS to act on T cells and inhibit JAK3/STAT5 signaling pathway and promote the overexpression of FasL or PD‐1 on the surface of T cells, resulting in apoptosis of T cells. MDSCs also release itaconate to repress the proliferation and antitumor cytotoxicity of T cells. The indirect effect is mainly to inhibit the development of T cells by consuming and competing for arginine and cysteine in the environment; releasing IL‐1β, IL‐6, IL‐23, PGE2, and other factors to generate Th17 and Treg cells to inhibit T cells’ immune function. At the same time, MDSCs are involved in the activation of TAMs, converting M1 to M2, mainly by increasing the release of IL‐10. VGEF may exert a proinflammatory cytokine release effect through VEGFR2/STAT3 signaling, inducing increased IL‐6, IL‐8/CXCL8, and GRO‐α/CXCL1 secretion in endothelial cells (nonleukocytes) and exerting an immunosuppressive function. VEGF upregulates the activity of NK cells and their natural immune response. In contrast, TGF‐β has involved in down‐regulating the expression of the NK‐activated receptor NKG2D on the NK cell surface, inhibiting NK cells from producing cytokines. In TME, dysregulated TGF‐β signaling suppresses antitumor immunity and promotes tumor fibrosis, epithelial‐to‐mesenchymal transition, and angiogenesis. MDSCs induce the differentiation of Bregs and DCregs through the secretion of IL‐10. In addition, IL‐10 also can inhibit the activation and proliferation of NK cells, NKT cells, and DCs and the production of various antitumor cytokines. EMT, epithelial–mesenchymal transition; Bregs, regulatory B cells; DCregs, regulatory dendritic cells; Tregs, regulatory T cells; TGF‐β, transforming growth factor‐β; VEGF, vascular endothelial growth factor; IL, interleukin; IDO, indoleamine 2,3‐dioxygenase; PGE2, prostaglandin E2; TAMs, tumor‐associated macrophages; CXCL, CXC motif chemokine ligand; GRO‐α, growth related oncogene‐α; Arg‐1, arginase‐1; ROS, reactive oxygen species; RNS, reactive nitrogen species; NO, nitric oxide; PD‐L1, programmed death ligand‐1; SLC7A11, cystine/glutamate antiporter solute carrier family 7 member 11; PD‐1, programmed death‐1; PNT, peroxynitrite; JAK3/STAT5, Janus kinase 3/signal transducer and activator of transcription 5; TCR, T‐cell receptor.
5. MDSC‐TARGETED THERAPY
MDSC‐targeted drugs in cancer therapies can be divided into mAbs, small‐molecular inhibitors, and peptides based on their chemical structure. In this section, we summarize studies of MDSC‐targeted drugs and introduce their applications and mechanisms. These targeted drugs can achieve anti‐MDSC effects by inhibiting MDSC functions, inhibiting MDSC recruitment into the TME, restraining myelopoiesis or MDSC development, inducing MDSC differentiation, inhibiting dedifferentiation into MDSCs, and mediating MDSC depletion (Figure 6).
FIGURE 6.

Mechanisms of myeloid‐derived suppressor cell (MDSC)‐targeted drugs in cancer therapy. MDSC‐targeted drugs can achieve anti‐MDSC effects in 5 ways: (1) inhibiting MDSC functions, (2) inhibiting MDSC recruitment into TME, (3) restraining myelopoiesis or MDSC development, (4) inducing MDSC differentiation or inhibiting dedifferentiation into MDSCs, and (5) mediating MDSC depletion. PI3K, phosphoinositide‐3‐kinase; DHODH, dihydroorotate dehydrogenase; PDE5, phosphodiesterase 5; DKK1, dickkopf‐1; HDAC, histone deacetylase; TKI, tyrosine kinase inhibitor; VEGF, vascular endothelial growth factor; CXCR2, CXC motif chemokine receptor 2; COX‐2, cyclooxygenase‐2; IMiD, immunomodulatory drug; PD‐1, programmed death‐1; CTLA‐4, cytotoxic T‐lymphocyte antigen 4; EGFR, epidermal growth factor receptor.
5.1. The applications of mAbs in MDSC‐targeted therapy
5.1.1. Anti‐PD‐1/PD‐L1
PD‐1 is a self‐protective protein receptor expressed on the surface of immune cells (mainly T cell). PD‐L1 is its corresponding ligand on the surface of multiple cells, including hematopoietic and tumor cells. 177 Receptor–ligand bonding can activate the T‐cell receptor and CD28 signaling pathways in T cells restraining of T cell proliferation, cytokine production, and immunity promotion; eventually, programmed T‐cell death occurs. 178 Some tumor cells overexpress PD‐L1 to achieve immune escape; therefore, PD‐1 and its ligand are important targets for antitumor therapy. PD‐L1 is also expressed on the surface of MDSCs and is upregulated in the TME due to hypoxia. 179 Therefore, anti‐PD‐1/PD‐L1 mAbs can directly act on the corresponding target in MDSCs to induce tumor suppression. The blockade of PD‐L1 in MDSCs decreases the expression of immunosuppressive cytokines, such as IL‐10, and activates T cells to resist MDSC‐induced immune suppression. 179
In the study conducted by Tzeng et al., 180 the mean reduction in peripheral M‐MDSC levels reached 4.70% per dose during the successive doses of anti‐PD‐L1 treatment (atezolizumab or avelumab) targeting metastatic urothelial carcinoma. In parallel, anti‐PD‐1 mAb (pembrolizumab) decreased PD‐1‐positive M‐ and immature‐MDSCs by 1.42% and 0.73% per dose, respectively. 180 This study did not show a correlation between MDSC PD‐1/PD‐L1 expression and prognosis after mAb treatment. However, the MDSC level before anti‐PD‐L1 mAb treatment could be regarded as a promising predictor of clinical status. 181 Similar results of MDSC frequency reduction induced by nivolumab, a type of PD‐1 blockade, were also reported. 10
Patients suffering from various cancers (such as melanoma and NSCLC) easily showed undesired or poor clinical effects after receiving anti‐PD‐1/PD‐L1 treatment due to drug resistance. 182 , 183 But fortunately, depleting ARG‐1‐expressing PMN‐MDSCs could reverse the anti‐PD‐1/PD‐L1 resistance of tumors. 184 Targeting some other MDSC‐related immune checkpoints has reduced drug resistance and enhanced the antitumor effects of PD‐1/PD‐L1 blockades. 185 M‐MDSCs expressing galectin 9 (LGALS9), the ligand of hepatitis A virus cellular receptor 2 (HAVCR2/TIM3), are correlated with reduced CD8+ T cell secretion. Pretreatment with anti‐TIM3 could be an effective approach for reducing resistance to nivolumab, a type of PD‐1 blockade, in patients with NSCLC. 186 Using cabozantinib to deplete ARG‐1‐expressing PMN‐MDSCs enhanced the efficacy of anti‐PD‐1 therapy. 184 Targeting HIF‐1α could regulate PD‐L1 expression in MDSCs, and combining anti‐VEGF/VEGFR mAbs with PD‐1 could also reduce MDSCs proliferation, improving the antitumor effects. 179 , 187 Given multiple PD‐1/PD‐L1 blockade resistance, personalized treatment is required, and accurate judgments of patients’ prognosis and disease progression after mAb treatment are required. Specific predictors are needed to achieve this, among which MDSCs are a promising option. 188 However, research is still at the phenomenon observation stage; a complete mechanical explanation of MDSC‐related clinical status remains to be elucidated.
5.1.2. Anti‐CTLA‐4
Whether MDSCs express CTLA‐4 remains unclear. 189 However, some studies have shown the effects of CTLA‐4 blockade on MDSCs, especially with ipilimumab. The effects on MDSC subsets differed during ipilimumab treatment in patients with melanoma. 12 In particular, the concentration of PMN‐MDSCs decreased in all patients undergoing CTLA‐4 blockade, while the concentration of M‐MDSCs significantly changed only in clinically benefitting patients. 12 , 190 Additionally, the MDSC level prior to ipilimumab treatment and its changes during therapy are highly relevant to patient survival. 191 , 192 , 193 CTLA‐4 blockades combined with some other targeted drugs can reduce the frequency of MDSCs, and combinational therapies can alter MDSC functions. 194 , 195 Anticolony stimulating factor 1 (CSF1)/CSF1 receptor (CSF1R) treatment combined with CTLA‐4 blockade caused regression in breast and colon cancers in vivo by reprogramming MDSCs into antineoplastic ones. 195 M‐MDSCs showed a reduced immunosuppressive ability after anti‐CTLA4 therapy combined with anti‐VEGFR therapy in mice with melanoma. 196
5.1.3. Anti‐VEGF
SET domain‐containing 2 histone lysine methyltransferase (SETD2/HIF‐1) and VEGF are abundantly activated in tumor sites, enhancing angiogenesis, which also induces MDSC migration to and differentiation at tumor sites. 197 , 198 , 199 VEGF can recruit MDSCs via the JAK/STAT3 pathway and VEGF–VEGFR2 interaction 21 and enhance their ability to generate other immunosuppressive cells. 145 In addition, MDSCs produce VEGF to facilitate angiogenesis. 199 Studies have observed the MDSC‐targeting effects induced by bevacizumab, an anti‐VEGF mAb. 200 , 201 The concentration of MDSC subsets was significantly associated with disease prognosis after treatment, 202 with CD15+ M‐MDSCs increasing in abundance in patients with tumor progression. Undesirable anti‐VEGF therapeutic effects may be attributed to increase GM‐CSF levels after treatment, which mediate MDSC migration and differentiation. 203 To reverse anti‐VEGF refractoriness in tumors, Iwai et al. 204 considered capecitabine as a promising agent since it reduced tumor drug resistance and pyrimidine nucleoside phosphorylase‐expressing MDSCs in tumor sites and peripheral blood in patients with Lewis lung carcinoma. Low‐dose capecitabine treatment with bevacizumab also had an inhibitory effect on circulating MDSCs in patients with glioblastoma. 201
5.1.4. Anti‐EGFR
While the modulating pathways between the EGFR and MDSCs are unclear, studies have observed changes in MDSCs after interfering with the EGFR. 205 , 206 In T helper 1 cell cytokine‐enriched environments in pancreatic cancer, EGFR‐targeted therapy induced by EGFRBi (EGFR bispecific antibodies) armed active T cells, restraining the prostaglandin‐endoperoxide synthase 2 (PTGS2/COX2)/PGE2/ARG‐1 axis and enhancing TNF‐α, IL‐12, CCL3, CCL4, CCL5, CXCL9, and CXCL10 to suppress MDSC differentiation and accumulation. 207 This effect may be partly attributed to EGFR interference but is more likely due to cytokine interactions between active T cells and the environment. Therefore, the T cell carrier may be further removed to determine the individual impact induced by EGFR. In clinical practice, Li et al. 206 reported that cetuximab, an EGFR‐specific mAb, significantly decreased PMN‐MDSCs only in responders among patients with head and neck squamous cell carcinoma (HNSCC); meanwhile, it can inhibit the polarization of monocytes into M2 macrophages and induce functioning by bonding Fc‐gamma receptors in myeloid cells. In nonresponders, the number of circulating M‐MDSCs significantly increased. Another clinical trial showed that a higher pretreatment M‐MDSC/PMN‐MDSC ratio might induce a higher pseudo F‐statistic in patients with HNSCC. 208 Therefore, M‐MDSCs could be a promising predictor of clinical prognosis after cetuximab treatment. However, combinational therapy with cetuximab and radiation markedly upregulated IL‐6–STAT3 signaling in circulating MDSCs and might lead to undesirable patient prognoses. 209
5.2. The applications of small‐molecular inhibitors in MDSC‐targeted therapy
5.2.1. Tyrosine kinase inhibitors
Sunitinib is a type of receptor protein‐TKI (RPTKI) with multiple targets, including VEGFR, ret proto‐oncogene (RET), and KIT proto‐oncogene receptor tyrosine kinase (KIT/c‐Kit), 210 that reduced the peripheral MDSC frequency 8 and part of the MDSC frequency in tumor sites. 9 Mechanistically, its inhibitory effects on MDSCs might inhibit the VEGFR/VEGF axis since it functions in the recruitment of immunosuppressive cells, such as MDSCs and Tregs. 145 Sunitinib‐induced MDSC inhibition is related to STAT3 pathway downregulation, constraining the development of MDSCs from myeloid progenitor cells 8 and improvement of angiogenesis in tumor sites. 211 Similarly, directly targeting STAT3 is also an effective approach for inhibiting the generation of M‐MDSCs which has been observed in patients with prostate cancer. 212 Interestingly, the anti‐MDSC effect of sunitinib is independent of its antitumor activities since significant effects on MDSCs have occurred in the absence of tumor progression inhibition. 13 c‐Kit is another promising target for anti‐MDSC therapies, which accounts for MDSC accumulation via enhanced myelopoiesis and inhibited myeloid cell differentiation, 213 and sunitinib has demonstrated a c‐kit‐dependent anti‐MDSC activity. 214 However, sunitinib resistance also occurs in tumors due to the recombinant GM‐CSF, which selectively activates STAT5 and suppresses STAT3 instead. 13 Additionally, GM‐CSF induces the transformation of two MDSC subsets, specifically 215 to gain higher immunosuppressive activities in the TME. 216 , 217
Other TKIs have also shown anti‐MDSC efficacies. Stiff et al. showed that ibrutinib, a type of Bruton's TKI, could weaken NO and IDO production in MDSCs and MDSC cytopoiesis and migration. 218 Another TKI, cabozantinib, significantly decreased the MDSC numbers in both the tumor site and spleen, restraining ARG‐1 expression and assisting the recruitment of CD8+ T cells into the TME. 219 Unlike sunitinib, some TKIs, including nilotinib, dasatinib, and sorafenib, have shown dose‐dependent inhibitory effects on the differentiation process of mature monocytes into M‐MDSCs but not on the maturation process of M‐MDSCs as well as their function, suggesting that these inhibitors are more applicable in the early stage of MDSC infiltration. 220 However, the MDSC‐modulating patterns of TKIs and the different regulatory pathways of sunitinib compared with other TKIs require further studies.
5.2.2. CXCR inhibitors
Chemokines are important mediators in tumor progression. The CXCR–CXCL system can induce immune cell infiltration, tumor proliferation, metastasis, and angiogenesis. 221 CXCL1, CXCL2, CXCL5, and CXCL8 can mediate MDSC recruitment and accumulation at the tumor site via CXCL–CXCR1/CXCR2 interactions. 222 SX‐682, a selective CXCR1/CXCR2 inhibitor, has shown antitumor capability via MDSCs: suppressing MDSC recruitment in the TME. 223 , 224 CXCR2 inhibition suppressed the metastasis of pancreatic ductal adenocarcinoma 225 and enhanced the efficacy of immune checkpoint inhibitors (ICIs) by increasing T cell infiltration. 225 , 226 However, Sun et al. 224 indicated that the antitumor activity of SX‐682 is separate from its effect on MDSCs, partly because it only inhibits the migration of PMN‐MDSCs merely without down‐regulating their function and tumor CXCL expression. Therefore, combining of CXCR blockades with ICIs is essential. CXCR2 is also a mediator of MDSC recruitment in regional lymph nodes, playing a role in preventing tumor metastasis, and CXCR‐antagonist SB225002 reversed MDSC trafficking. 227 Moreover, the CXCL12/CXCR4, CXCL13/CXCR3, and CXCL17/CXCR8 axes also induce MDSC migration to tumor sites. 222 CXCR4 inhibitors potentiated anti‐PD‐1 ability in pancreatic cancer, ovarian cancer, and glioblastoma. 228 , 229 , 230 In a phase IIa clinical trial, combining the CXCR4 inhibitor BL‐8040 with the PD‐1 mAb pembrolizumab decreased the PMN‐MDSC density in patients with pancreatic ductal adenocarcinoma; one stable patient showed reduced PMN‐MDSC levels (by approximately 25%) after two cycles of combined treatment. 229 No published studies support the inhibition of the CXCL13/CXCR3 and CXCL17/CXCR8 axes.
5.2.3. Immunomodulatory drugs
Immunomodulatory drugs (IMiDs), consisting of lenalidomide and pomalidomide, are a group of antitumor drugs derived from the prototype compound thalidomide that can be used for some hematologic tumors, especially multiple myeloma. 231 , 232 Lenalidomide reverses tumor progression mainly via cytotoxicity and angiogenesis resistance, 233 with its antitumor mechanisms attributable to MDSC inhibition. 234 , 235 Sakamaki et al. 233 found a significant 1.39% reduction in splenic MDSCs in tumor‐bearing but not in naïve mice. Since lenalidomide is a cereblon‐targeting (CRBN‐targeting) drug, comparing CRBN expression in MDSCs from naïve and tumor‐bearing individuals is necessary. Kuwahara‐Ota et al. 236 found that combining lenalidomide with pomalidomide, inhibited MDSCs by downregulating CCL5 and macrophage migration inhibitory factor in myeloma cells, enhancing IRF8 expression and downregulating CCR5 in peripheral blood mononuclear cells in vitro. Notably, the multiregulatory mechanism of lenalidomide could also increase CD14+CD15+ MDSCs, causing T cell suppressive effects. 237 Therefore, it is worth exploring the optimal concentration that provides good antitumor efficacy with minimal immunosuppressive activity.
5.2.4. IDO inhibitors
Tryptophan is an essential amino acid required that is taken in through diet, and tumor burden is associated with tryptophan catabolism. Initially, this kind of catabolism, which results in the depletion of tryptophan, is controlled by IDO and tryptophan dioxygenase. Tryptophan starvation suppresses T cell growth and activation, promoting tumor immune escape, metastasis, and angiogenesis. 238 , 239 Physiologically and initially, IDO1, which induces the establishment of self‐tolerance in the fetal period, facilitates immunosuppressive behaviors by promoting the generation of MDSCs and forkhead box P3 (FOXP3)+ Tregs. 240 , 241 In the TME, tumor IDO promotes MDSC recruitment, infiltration, and activation via Treg‐dependent pathways. 242 Consequently, MDSCs in breast cancer stimulated with the IL‐6 family showed increased IDO1 expression and assisted in tryptophan catabolism partly via the STAT3/NF‐κB/IDO pathway, which forms positive feedback. 243 Epacadostat is a selective IDO1 inhibitor, which has antineoplastic effects via facilitating T and NK cell proliferation and immune activity, increasing the number of IFN‐γ and CD86hi DCs, and reversing the accumulation of Treg‐like cells. 244 A phase II clinical trial reported that the mean MDSC level decreased by 1.9% from the baseline after epacadostat treatment. 245 Navoximod and indoximod are other IDO1 inhibitors with tumor growth suppressive activity but generally lack information about MDSC‐related changes. Only a few studies have reported the correlation between IDO1 inhibitors and MDSCs; thus, further research is needed to bridge this gap.
5.2.5. Phosphoinositide‐3‐kinase inhibitors
Once activated by upstream signals from receptor tyrosine kinases, phosphoinositide‐3‐kinases (PI3Ks) will lead to several key regulations in tumor‐associated immunity. 246 The PI3K signaling pathway has diverse protumoral abilities: promoting the production of immunosuppressive cytokines and enhancing PD‐L1 expression and MDSC and Treg infiltration. 247 Class I PI3K has four isoforms, namely α, β, δ, and γ. However, anti‐MDSC activity is only observed with PI3Kγ inhibitors (IPI‐549) both in vivo and in vitro. 248 , 249 In the in vivo murine study conducted by Zhang et al., 248 IPI‐549 decreased CD11b+ Gr‐1+ MDSCs in pancreatic cancer by almost 50% via two distinct delivery routes: oral administration and a targeted nanoparticle‐encapsulated approach. Nanoparticle usage doubled the half‐life of IPI‐549 and ensured drug accumulation in the tumor site, but the MDSC concentration did not significantly differ between the two distinct routes, suggesting that the anti‐MDSC effects of IPI‐549 may be dose‐independent. Additionally, the dual PI3K/mechanistic target of rapamycin kinase (mTOR) inhibitor dactolisib downregulated the oxidative phosphorylation of MDSCs. 250 The dual PI3K/mTOR inhibitor SF2523 has also been reported to block CD11b+ Gr‐1+ MDSC recruitment at tumor sites. 251
5.2.6. Phosphodiesterase 5 inhibitors
Phosphodiesterase (PDEs) are common metallohydrolases downstream of cyclic‐AMP and cyclic‐GMP that induce the inactivation of both secondary messengers. Among the 11 subtypes of PDEs, PDE5 has a cGMP‐specific hydrolytic function. 252 PDE5 inhibitors have been developed and used for various cancer types, including HNSCC, melanoma, myeloma, prostate cancer, gastrointestinal cancer, and neurologic tumors, 253 , 254 , 255 , 256 , 257 augmenting antitumor immune responses in cancer‐bearing hosts. The targeted roles of PDE5 inhibitors are observed in MDSCs by competently suppressing the function and recruitment of MDSCs and decreasing the concentration of MDSCs via two pivotal pathways: reduction of ARG‐1 and iNOS production, which induces T‐cell dysfunction. 258 ARG‐1 and iNOS expressions are mediated by STAT6/IL‐4 receptor alpha (IL‐4Rα), and PDE5 inhibitors can affect outcomes by reducing of IL‐4 Rα in the upstream pathway. 258 , 259 Moreover, the decrease in ROS induced by PDE5 inhibitors also benefits to tumor‐specific immunity restoration. 256 In both murine tumor models and clinical trials, tadalafil has been shown to have potential utility in MDSC‐targeted treatments. Noonan et al. 256 found that the number of CD14+ MDSCs decreased by 5.6% in patients with end‐stage multiple myeloma after 11 months of treatment with tadalafil. Given its influence on MDSC function, IL‐4Rα expression markedly decreased from 51.37 to 2.13% after treatment, and both IL‐4Rα and ROS levels were restored to baseline. 256 Similarly, a phase II clinical trial conducted on patients with HNSCC suggested that MDSC generation and function were significantly impaired after tadalafil therapy. 260 Notably, a high PDE5 inhibitor intake may result in contrasting effects on antitumor immunity modulation since they may cause off‐target effects on PDE11 at high doses, which mediates the increase in cyclic‐AMP. 261
5.2.7. Histone deacetylase inhibitors
Vorinostat, entinostat, and ricolinostat are histone deacetylase (HDAC) inhibitors that promote histone acetylation, transforming gene expressions via epigenetic reprogramming. 262 HDAC inhibitors are proven to have shown inhibitory functions in MDSCs of breast, renal, and lung cancers; neuroblastoma; and lymphoma. 262 , 263 , 264 , 265 The principal effect of entinostat is to inhibit the functions of PMN‐MDSCs, but its effect against M‐MDSCs remains unclear. 263 , 266 Entinostat can reduce ARG‐1, iNOS, and COX‐2 production and modulate NF‐κB‐ and STAT3‐related pathways. 264 , 266 The side effect is that entinostat alone may increase the number of PMN‐MDSCs, 265 , 266 therefore, combined treatment with other targeted drugs can make full use of MDSC inhibitory functions. Combined with 5‐azacytidine and DNA methyltransferase inhibitors, entinostat restrained the trafficking of MDSCs and promoted their differentiation. 267 In addition, entinostat‐ICI combinations altered the TME from an M‐MDSC‐ to PMN‐MDSC‐predominant milieu and improved tumor sensitivity to ICIs. 265 , 266
5.2.8. COX‐2 inhibitors
PGE2 is an inflammatory cytokine that tumors secrete to create a local proinflammatory microenvironment. 268 , 269 , 270 COX‐2 is the enzyme that induces PGE2 production. Its expression in the TME can promote tumor proliferation and angiogenesis and reduce tumor apoptotic and immunologic responses. 270 PGE2 can induce MDSC maturation from bone marrow stem cells 271 and promote MDSC accumulation via COX2/PGE2/prostaglandin E receptor 4 (PTGER4/EP4) signaling. 272 The COX2 inhibitor celecoxib reduced the number and immunosuppressive ability of MDSCs by reducing ROS and NO production and reversing MDSC‐related T‐cell tolerance. 273 , 274 Furthermore, GM‐CSF production is suppressed, which affects MDSC maturation and accumulation. 275 Another COX‐2 inhibitor, SC58236, inhibited MDSC differentiation in breast cancer. 271
5.2.9. Dihydroorotate dehydrogenase inhibitor
Colligan et al. 276 recently found that the dihydroorotate dehydrogenase (DHODH) inhibitor brequinar, once used to treat leukemia by restoring terminal differentiation of leukemic myeloid progenitors, could successfully target MDSCs in a breast cancer model. DHODH accelerates pyrimidine synthesis, 277 an important step leading to MDSC biogenesis and immunosuppression. DHODH inhibition ultimately promotes the terminal differentiation and maturation of myeloid cells, which is already achieved in vitro in human bone marrow, and the antitumor immunity of T cells. 276 Combining brequinar with an anti‐PD‐1 mAb overcame the drug resistance of breast cancer and restored CD8+ T cell proliferation and CD8+ T cell‐mediated direct tumoricidal effects. 276
5.3. The applications of peptides in MDSC‐targeted therapy
Tumor‐targeting peptide is a state‐of‐the‐art research direction for tumor‐related targeted treatments. Peptides are notable for their small molecule mass, synthetic convenience, and good biocompatibility compared with proteins. The peptides used to target MDSCs, or broadly the TME can be divided into two categories: (1) those without antitumor efficacies used only in targeted combinations of specific receptors, and (2) those with specific antitumor functions. The first group of peptides functions as carriers of antitumor drugs. The pharmacological actions of antitumor drugs can be achieved via specific binding to superficial, intracellular, or intranuclear receptors. Most studies have focused on delivering drugs to tumor cell surfaces or targeting vascular endothelial cells to block tumor nutrient sources. FAM‐A54, selected by phage display approaches, has shown binding specificity for diverse liver cancer cell lines with the potential binding motif of Pro‐Ser. A54‐covered doxorubicin has shown a high absorptivity in cancer cells. 278 The tripeptide NGR (Asn‐Gly‐Arg) was notably competent for anchoring tumor‐related angiogenic vessels, molecularly targeting aminopeptidase N (ANPEP/CD13) in endothelial cells. 279 Zheng et al. 280 synthesized an NGR‐apoprotein fusion protein and loaded active enediyne chromophore to realize lidamycin carrying. Lidamycin comprises two components, apoprotein and the active enediyne chromophore, and is considered a common chemotherapeutic drug that damages DNA. Tumor growth inhibition was reportedly elevated by 14.1% compared with tumor suppression after a single lidamycin treatment. The antitumor effects of doxorubicin can also be enhanced by NGR. 281 While minimal evidence has shown that NGR alone can affect MDSCs, the NGR–TNF complex (low dose: 0.8 μg/m2) does not induce MDSC mobilization in peripheral blood and recruitment to the tumor site. 282 Therefore, NGR–TNF antitumor treatment excludes the drug‐fast impact of MDSCs. RGD (Arg‐Gly‐Asp) is another tripeptide specifically recognizing integrin αvβ3, Xia et al. 283 synthesized a RGD‐contained, lactate dehydrogenase A (LDHA) small inhibitory RNA‐cored nanostructure assembly, this specific assembly could inhibit LDHA expression, which subsequently decreased G‐CSF and GM‐CSF production and suppressed MDSC recruitment. Another peptide AXT201, a 20‐amino polypeptide, achieves antitumor effect by the reduction of protumor growth factors, such as VEGF, HGF, and insulin‐like growth factor 1, inhibition of angiogenesis, remodeling of the TME, and promotion of CD8+ T cell activation. 284 AXT201 was shown to mainly alter the proportion of MDSC subtypes in triple‐negative breast cancer to realize an antitumor change in the TME. In particular, M‐MDSCs decreased by 28%, close to sunitinib's effects on the entire MDSC population by sunitinib (33%). 13 However, AXT201 had minimal impact on PMN‐MDSCs. 284 The mechanisms underlying M‐MDSC reduction mechanisms may be attributed to the coreceptor and the corresponding pathways of AXT201 since MDSCs can promote tumor angiogenesis by producing VEGF, MMP9, and bFGF. 199 , 285 Anti‐VEGF treatment also significantly affects MDSCs as mentioned above. While this biomimetic peptide might also modulate other growth factors, further research is required. MDSC‐targeted drugs and studies reporting their effectiveness are summarized in Table 2.
TABLE 2.
Drugs and studies to target myeloid‐derived suppressor cells (MDSCs) in cancers.
| Drug type | Drug name | Year | Cancer samples | Effects | Drug status | References |
|---|---|---|---|---|---|---|
| Monoclonal antibody | Atezolizumab/avelumab (anti‐PD‐L1 mAb) + pembrolizumab (anti‐PD‐1 mAb) | 2018 | Metastatic urothelial carcinoma blood samples (human) |
The percentage of PDL1+ monocytic myeloid‐derived suppressor cells (M‐MDSC) decreased after anti‐PDL1 treatment. The percentage of PD1+ M‐MDSC and immature‐myeloid‐derived suppressor cells (I‐MDSC) decreased after anti‐PD1 treatment. |
Preclinical trial | 180 |
| Atezolizumab (anti‐PD‐L1 mAb) | 2018 | Non‐small cell lung cancer (NSCLC) blood samples (human) | MDSC frequency was decreased . | Preclinical trial | 11 | |
| RMP1–14 (anti‐PD‐1 mAb) + peptide R (CXCR4 antagonist) | 2019 |
MC38 colon cancer cell line (mice); B16 melanoma transduced with human CXCR4 cell line (mice) |
Increase the CD8/MDSC ratio | Preclinical trial | 286 | |
| Nivolumab (anti‐PD‐1 mAb) + temozolomide (alkylating agents) | 2020 | Small cell lung cancer (SCLC) blood samples (human) | MDSC experienced an early decrease during therapy |
Phase II clinical trial |
10 | |
| Ipilimumab (anti‐ CTLA‐4 mAb) + ATRA (all‐trans retinoic acid) | 2018 | Melanoma blood samples (human) |
ATRA reduces PD‐L1, IL‐10, and IDO gene expression by MDSCs. ATRA‐ipilimumab combination significantly decreases the frequency of circulating MDSCs. |
Phase II clinical trial |
287 | |
| Cetuximab (anti‐EGFR mAb) | 2015 | Locally advanced head and neck squamous cell carcinoma (HNSCC) blood samples (human) | The M‐MDSCs of nonresponders increased and the polymorphonuclear myeloid‐derived suppressor cells (PMN‐MDSCs) of responders decreased significantly. |
Phase II clinical trial |
206 | |
| Bevacizumab (anti‐VEGF mAb) | 2016 | Non‐small cell lung cancer (NSCLC) blood samples (human) | The decrease of PMN‐MDSCs and the increase of CD15+ M‐MDSCs were shown in patients with disease progression. | Preclinical trial | 202 | |
| Bevacizumab (anti‐VEGF mAb) + EGFR TKI | 2018 | EGFR‐mutated lung adenocarcinoma blood samples (human) | The percentage of circulating MDSCs decreased after bevacizumab–TKI combined treatment while TKI alone had no significant changes. | Phase II clinical trial | 200 | |
| N‐809 (IL‐15 superagonist N‐803 with two αPD‐L1 domains) | 2020 |
4T1 triple negative breast carcinoma cell line (mice) Yac‐1 lymphoma cell line (mice) MC38 colon carcinoma cell line (mice) |
Decrease the M‐MDSC infiltration in primary tumor tissues, but increase the concentration of M‐MDSCs in the draining lymph nodes. | Preclinical trial | 288 | |
| MR16–1 (anti‐IL‐6R mAb) | 2012 | CMC‐1 methylcholanthrene‐induced skin squamous cell carcinoma cell line (mice) | Anti‐IL‐6R mAb reduced the number of MDSCs, subsequently improving antitumor T cell response and IFN‐γ expression. | Preclinical trial | 289 | |
| mDKN‐01 (anti‐DKK1 mAb) | 2021 |
B16F0 melanoma cell line (mice); 4T1 breast cancer cell line (mice) |
Reduction of Gr‐1+CD11b+ MDSCs in the tumor and spleen Upregulation of PD‐L1 expression on MDSCs |
Preclinical trial | 290 | |
| Small‐molecule inhibitor | Ibrutinib (inhibitor of Bruton's tyrosine kinase and IL2‐inducible T‐cell kinase) | 2016 |
4T1 breast cancer cell line (mice) B16F10 melanoma cell line (mice) metastatic melanoma blood samples (human) |
Ibrutinib reduced the NO production, MDSC migration, and the frequency of MDSCs with significance. It enhanced the antitumor effect of PD‐L1 blockade as well. | Preclinical trial | 218 |
| Ibrutinib (inhibitor of Bruton's tyrosine kinase and IL2‐inducible T‐cell kinase) | 2021 | NB9464 neuroblastoma cell line (mice) | The treatment of ibrutinib on MDSCs changed the production of NO and reduced Ido, Arg and Tgfβ mRNA expression. | Preclinical trial | 291 | |
| Ibrutinib (inhibitor of Bruton's tyrosine kinase and IL2‐inducible T‐cell kinase) | 2021 | Chronic lymphocytic leukemia blood samples (human) | The number of PMN‐MDSCs, not M‐MDSCs, decreased consistently after treatment, and the decrease in PMN‐MDSC number was related to CLL level. | Preclinical trial | 292 | |
| Eganelisib (IPI‐549) (Phosphoinositide‐3‐kinases inhibitor) | 2019 | KPC pancreatic cancer cell line (mice) | The decrease of MDSCs was shown after IPI‐549 oral treatment or polymeric nanoparticle treatment. | Preclinical trial | 248 | |
| SX‐682 (CXCR2 inhibitor) | 2020 |
MOC2 oral cancer (mice) Head and neck squamous cell carcinoma (HNSCC) tumor biospecimens and blood samples (human) |
SX‐682 can significantly decrease the trafficking of MDSCs into TME, thus causing the disinhibition of NK cells. | Preclinical trial | 223 | |
| SX‐682 (CXCR2 inhibitor) | 2019 |
MOC1 oral cancer cell line (mice) LLC Lewis lung carcinoma cell line (mice) |
SX‐682 induced the decrease of CXCR2‐positive PMN‐MDSCs without altering CXCR2 ligand expression. | Preclinical trial | 224 | |
| Lenalidomide (CC‐5013) (ligand of ubiquitin E3 ligase cereblon) | 2014 | A20 lymphoma cell line (mice) | Lenalidomide reduced the splenic MDSCs without reducing tumor burden. | Preclinical trial | 233 | |
| Lenalidomide (CC‐5013) (ligand of ubiquitin E3 ligase cereblon) | 2018 |
MM1.s multiple myeloma cell line (human) Multiple myeloma blood samples (human) |
The lenalidomide‐based treatment reduced both the number of PMN‐MDSCs and ARG‐1 expression. | Preclinical trial | 234 | |
| Tadalafil (PDE 5 inhibitor) | 2014 | Multiple myeloma tumor biospecimens and blood samples (human) | Tadalafil reduced the frequency of M‐MDSCs as well as ARG‐1, iNOS, and ROS production of MDSCs. Changes were more significant in tumor tissues than periphery. | Preclinical trial | 256 | |
| Sunitinib (receptor TKI) | 2009 |
786‐O and RCC4 renal cell carcinoma (RCC) cell lines (human) RENCA renal carcinoma cell line (mice) |
Sunitinib suppressed Stat3 expression in renal cell carcinoma‐related MDSCs. | Preclinical trial | 293 | |
| Sunitinib (receptor TKI) | 2009 |
MCA26 colon carcinoma cell line (mice) LLC1 Lewis lung carcinoma cell line (mice) |
Sunitinib reduced the frequency of MDSCs, suppressive activity of MDSCs as well as the PD‐L1 expression on MDSCs. | Preclinical trial | 214 | |
| Sunitinib (receptor TKI) | 2009 | renal cell carcinoma (RCC) blood samples (human) | Sunitinib reduced MDSC viability and function significantly, which were associated with the reversal of type 1 T‐cell inhibition and CD3+CD4+CD25hiFoxp3+ Treg activation. | Preclinical trial | 294 | |
| Sunitinib (receptor TKI) | 2010 |
4T1 mammary carcinoma (mice) CT26 colonic carcinoma (mice) RENCA renal carcinoma (mice) |
Both M‐MDSCs and PMN‐MDSCs were suppressed by sunitinib, and MDSC resistance was induced by GM‐CSFs generation. | Preclinical trial | 13 | |
| Sunitinib (receptor TKI) | 2011 |
Metastatic renal cell carcinoma (mRCC) blood samples (human) 4T1 mammary carcinoma cell line (mice) CT26 colonic carcinoma cell line (mice) RENCA renal carcinoma cell line (mice) |
Sunitinib reduced the tumor‐infiltrating MDSCs in RENCA and CT26, but not 4T1, bearing mice. Sunitinib can also reduce circulating MDSCs accumulation in mRCC patients significantly, but MDSC resistance may happen. |
Preclinical trial | 9 | |
| Sunitinib (receptor TKI) | 2015 | Blood samples of various cancer patients with oligometastases (human) | Sunitinib decreased the number of M‐MDSCs as well as Arg levels in M‐MDSCs. | Preclinical trial | 295 | |
| Sunitinib | 2015 | Renal cell carcinoma (RCC) tumor biospecimens (human) | The sunitinib‐related MDSC suppression improved tumor‐infiltrating lymphocyte expansion. | Preclinical trial | 296 | |
| Sunitinib (receptor TKI) | 2019 | Nonmuscle invasive bladder cancer (NMIBC) blood samples (human) | CD33+ MDSCs decreased significantly after cycle 2 and 3 of the sunitinib treatment. |
Phase II clinical trial |
297 | |
| Sunitinib (receptor TKI) | 2019 | TC‐1 cell line (mice) | Both MDSCs in tumor site and spleen decreased after treatment. | Preclinical trial | 8 | |
| Nilotinib, dasatinib, sorafenib, and sunitinib (TKIs, TKIs) | 2016 | LX2 hepatic stellate cell line (human) | Nilotinib, dasatinib, and sorafenib, but not sunitinib decreased the differentiation of monocytes into M‐MDSCs. both the MAPK and NF‐κB pathways were not affected by the applied TKIs. | Preclinical trial | 220 | |
| INCB024360 (indoleamine 2,3‐dioxygenase inhibitor, IDO inhibitor) | 2018 | Myelodysplastic syndromes bone marrow samples (human) | Most patients experienced a decline on bone marrow MDSCs. |
Phase II clinical trial |
245 | |
| Vorinostat (histone deacetylase inhibitor, HDAC inhibitor) | 2016 | 9464D neuroblastoma cell line (mice) | Vorinostat decreased the number and function of MDSCs. | Preclinical trial | 262 | |
| Entinostat (histone deacetylase inhibitor, HDAC inhibitor) | 2017 |
RENCA‐Luc renal cell carcinoma cell line (mice) LLC lung carcinoma cell line (mice) CT26 colon carcinoma cell line (mice) |
Entinostat suppressed MDSC functions by reducing ARG‐1, iNOS, and COX‐2 production of MDSCs. | Preclinical trial | 264 | |
| Entinostat (histone deacetylase inhibitor, HDAC inhibitor) + immune‐checkpoint inhibitors (ICI) | 2018 | NT2.5 mammary cancer cell line (mice) | The Entinostat‐ICI combined therapy altered the TME from M‐MDSC‐ to PMN‐MDSC‐predominant milieu, and reduced ARG‐1 production of MDSCs. | Preclinical trial | 265 | |
|
Entinostat (histone deacetylase inhibitor, HDAC inhibitor) Ricolinostat (histone deacetylase inhibitor, HDAC inhibitor) |
2020 |
EL4 lymphoma cell line (mice) LLC lung carcinoma cell line (mice) |
Entinostat can only inhibit the immunosuppressive functions of PMN‐MDSCs, while ricolinostat can inhibit the functions of M‐MDSCs, the combination of two HDACis is capable of targeting two major MDSC subsets. | Preclinical trial | 263 | |
| Entinostat (histone deacetylase inhibitor, HDAC inhibitor) + 5‐azacytidine + DNA methyltransferase | 2020 |
LLC1 Lewis lung carcinoma cell line (mice) 4T1 mammary cancer cell line (mice) HNM007 p53‐null esophageal squamous cell carcinoma cell line (mice) |
The combined therapy downregulated CCR2 and CXCR2 to inhibit the trafficking of MDSCs, and promoted MDSC differentiation into a more‐interstitial macrophage‐like phenotype. | Preclinical trial | 267 | |
| Axitinib (TKI of VEGFR‐1, ‐2, and ‐3) | 2015 | MO4 melanoma cell line (mice) | Splenic and tumor‐infiltrating M‐MDSCs had a decline on immunosuppressive capacity after treatment. And axitinib‐treated MDSCs can stimulate allogeneic T cells, differentiating toward an antigen‐presenting phenotype. | Preclinical trial | 298 | |
| Celecoxib (cyclooxygenase‐2 inhibitor, COX‐2 inhibitor) | 2010 | AB1 mesothelioma cell line (mice) | Celecoxib reduced the number of MDSCs, reduced ROS and NO production and reversed T cell tolerance of MDSCs. | Preclinical trial | 274 | |
|
Celecoxib (cyclooxygenase‐2 inhibitor, COX‐2 inhibitor) Sunitinib (receptor TKI) |
2017 |
RENCA renal carcinoma cell line (mice) 769‐p renal carcinoma cell line (human) |
GM‐CSF expression can be inhibited by both celecoxib and sunitinib. The combined therapy reduced the number of circulating MDSCs as well as STAT3 expression in MDSCs. |
Preclinical trial | 275 | |
| SC58236 (cyclooxygenase‐2 inhibitor, COX‐2 inhibitor) | 2007 | 4T1 mammary cancer (mice) | SC58236 retarded MDSC development from bone marrow stem cells. | Preclinical trial | 271 | |
| Brequinar (dihydroorotate dehydrogenase inhibitor, DHODH inhibitor) | 2022 |
4T1 breast cancer cell line (mice); bone marrow culture system (human) |
Brequinar retrained pyrimidine synthesis in myeloid cells in human bone marrow to inhibit MDSC biogenesis and suppress the function of MDSCs, enhancing T cell immunity. The combination of brequinar and anti‐PD‐1 can relieve the ICI resistance of breast cancer and promote CD8+T‐induced cytotoxicity against tumor cells. |
Preclinical trial | 276 | |
| Cabozantinib (TKI) + Anti‐HER2 mAb | 2022 | 4T1 breast cancer cell line (mice) | The number of MDSCs in TME, lymph nodes, and spleens decreased by 20, 0.8, and 35% after the combination of cabozantinib and anti‐HER2 mAb. the combined treatment also inhibited the Arg 1 expression on MDSCs and recruited CD8+T cells into tumor site, tumor‐draining lymph nodes, and splenic lymphocytes | Preclinical trial | 219 | |
| Dactolisib (Phosphoinositide‐3‐kinases inhibitor, PI3K inhibitor) + anti‐CTLA4 mAb + anti‐PD1 mAb | 2017 | Prostate cancer (mice) | Dactolisib suppressed the PI3K signaling of MDSCs, upregulated interleukin‐1 receptor antagonist, and retain the activity of CD4+ and CD8+ T cells, finally promoting the antitumor effect of anti CTLA4 and anti‐PD1 mAbs. | Preclinical trial | 299 | |
| Dactolisib (Phosphoinositide‐3‐kinases inhibitor, PI3K inhibitor) | 2020 | Prostate cancer (mice) | Dactolisib inhibited mTOR and oxidative phosphorylation on PMN‐MDSCs, and therefore downregulate fatty acid oxidation and cause MDSC immunosuppressive activity. | Preclinical trial | 250 | |
| Peptides | AXT201 (an integrin‐binding peptide) | 2020 | Triple‐negative breast cancer cell line (mice) | M‐MDSC showed a 28% reduction after AXT201 treatment, while PMN‐MDSC did not demonstrate significant change. | Preclinical trial | 284 |
| R‐mPDV/PDV/DOX/siL (redox‐responsive nanoassembly) | 2021 | 4T1 Breast cancer cell line (mice) | R‐mPDV/PDV/DOX/siL induced LDHA silencing, suppressing the production of G‐CSF and GM‐CSF and leading to the inhibition of MDSCs recruitment. | Preclinical trial | 283 |
5.4. Potential MDSC‐targeted therapeutic methods dependent on ferroptosis inhibition
Previous studies have revealed that drug‐resistant tumors are especially sensitive to ferroptosis, indicating the promising effect of ferroptosis in tumor therapy. 300 , 301 On the contrary, ferroptosis also release damage‐associated molecular patterns to disturb antitumor innate immunity. 302 Meanwhile, induction of ferroptosis can cause bone marrow injury as the main obstacle for antitumor usage of ferroptosis activators. 303 Therefore, the clinical application of ferroptosis activators still has left many problems to fix. As mentioned above, taking TME into consideration, ferroptosis promotes the immunosuppressive activities of PMN‐MDSCs. Specifically inhibiting the ferroptosis of MDSCs may enhance antitumor immunity and consolidate the efficacy of other antitumor drugs, and research has already revealed that inhibiting ROS, the critical effector of ferroptosis, in MDSCs by catalase can induce T cell rejuvenation. 304 So far, there are no treatments proposed to specifically inhibit ferroptosis of MDSC in tumor‐bearing patients, in the future, MDSC characteristic receptor binding protein (such as targeted peptides) and ferroptosis inhibitors can be combined to become an alternative to accurately target MDSC through drug carriers.
6. CONCLUSIONS
MDSC is a kind of immunosuppressive cell which plays a vital role in various inflammatory diseases and tumor pathogenesis, progression as well as metastasis. Several studies have described the characteristics of MDSCs, dividing them into subtypes and elaborating on their functions in pathological conditions. Under most inflammatory circumstances, their immunosuppressive capacity can worsen inflammation progression. But in some scenarios such as hyperinflammation and autoimmune diseases, inhibiting autologous immune response is beneficial to disease prognosis. Besides, MDSCs were surprisingly proven to have some proimmune functions as well as nerve system regeneration promoting capacities in autoimmune diseases, indicating that MDSC cannot be merely considered as an immunosuppressive cell, and further investigations of its potential functions are necessary. Applying the immunosuppressive property of MDSCs to treat inflammation through in vitro MDSC induction and amplification is another novel field that draws much attention. A number of studies have already achieved artificial MDSC induction by adding cytokines, and the process is accessible theoretically. However, how to systematically induce high‐quality anti‐inflammatory MDSCs at a low cost remains unclear.
MDSCs have a critical role in transforming TME into a protumoral environment, and the main mechanism is to modulate the proliferation and functions of multifarious immune cells. In the current literature, we have summarized the various immunosuppressive functions of MDSCs and the corresponding mechanisms, and we finally established an integrated MDSC interaction network based on previous research results. However, the explorations on MDSC generation, differentiation, and functional mechanisms are still insufficient. Future work is required and is vital to improve clinical antitumor treatment.
Anti‐MDSC effects have recently shown for many targeted antitumor drugs, including some widely used mAbs, small‐molecular inhibitors, and peptides. And we have generalized their functions into five issues: (1) inhibiting MDSC functions, (2) inhibiting MDSC recruitment into TME, (3) restraining myelopoiesis or MDSC development, (4) inducing MDSC differentiation or inhibiting dedifferentiation into MDSCs, and (5) mediating MDSC depletion. Our summary will provide comprehensive references and instructions for clinical drug applications. However, the detailed mechanisms behind the clinical benefits of MDSC‐targeted therapy are poorly understood for most drugs. Further studies should evaluate drug‐MDSC interaction modes, including direct/indirect targeting, or outcomes of secondary tumor‐targeted changes. Since only a small amount of MDSCs exist under steady‐state and normal conditions, MDSC‐targeted therapy can be much less harmful to healthy tissues and cells than traditional chemotherapies, which therefore prevents severe side effects during anticancer treatments.
In the future, after further investigation into MDSCs and their mechanisms in the immune microenvironment, MDSC‐targeted anticancer therapy and MDSC‐based anti‐inflammatory treatment may be finally applied.
AUTHOR CONTRIBUTIONS
B. S. and R. Y. R. designed the project. R. Y. R., C. Y. X., R. Y. M., and Y. X. W. drafted the manuscript. R. Y. R., C. Y. X., R. Y. M., Y. X. W., and T. Y. Y. finished the search of data for the article. B. S., R. Y. R., and J. Y. Y. revised and finalized the manuscript. All authors read and approved the final manuscript.
CONFLICT OF INTEREST STATEMENT
The authors declare that they have no competing interests.
ETHICS APPROVAL AND CONSENT TO PARTICIPATE
Not applicable.
ACKNOWLEDGMENTS
We sincerely acknowledge the Adobe Illustrator and Microsoft Power Point software for image drawing. All the images and elements used in this manuscript are original and there are no copyright issues. This study was funded by the National Natural Science Foundation of China (No. 81972193 and 82102897), the Full‐Time Postdoctoral Research and Development Fund of Sichuan University (No. 2021SCU12026), and Scientific Research Foundation for Recruited Talents, West China Hospital of Stomatology Sichuan University (No. QDIF2019‐1).
Ren R, Xiong C, Ma R, et al. The recent progress of myeloid‐derived suppressor cell and its targeted therapies in cancers. MedComm. 2023;4:e323. 10.1002/mco2.323
DATA AVAILABILITY STATEMENT
Not applicable.
REFERENCES
- 1. Veglia F, Perego M, Gabrilovich D. Myeloid‐derived suppressor cells coming of age. Nat Immunol. 2018;19(2):108‐119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Law AMK, Valdes‐Mora F, Gallego‐Ortega D. Myeloid‐derived suppressor cells as a therapeutic target for cancer. Cells. 2020;9(3): 561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Gabrilovich DI. Myeloid‐derived suppressor cells. Cancer Immunol Res. 2017;5(1):3‐8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Veglia F, Sanseviero E, Gabrilovich DI. Myeloid‐derived suppressor cells in the era of increasing myeloid cell diversity. Nat Rev Immunol. 2021;21(8):485‐498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Bronte V, Brandau S, Chen SH, et al. Recommendations for myeloid‐derived suppressor cell nomenclature and characterization standards. Nat Commun. 2016;7:12150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Stadtmann A, Zarbock A. CXCR2: from bench to bedside. Front Immunol. 2012;3:263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Huang B, Pan PY, Li Q, et al. Gr‐1+CD115+ immature myeloid suppressor cells mediate the development of tumor‐induced T regulatory cells and T‐cell anergy in tumor‐bearing host. Cancer Res. 2006;66(2):1123‐1131. [DOI] [PubMed] [Google Scholar]
- 8. Draghiciu O, Nijman HW, Hoogeboom BN, Meijerhof T, Daemen T. Sunitinib depletes myeloid‐derived suppressor cells and synergizes with a cancer vaccine to enhance antigen‐specific immune responses and tumor eradication. Oncoimmunology. 2015;4(3):e989764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Finke J, Ko J, Rini B, Rayman P, Ireland J, Cohen P. MDSC as a mechanism of tumor escape from sunitinib mediated anti‐angiogenic therapy. Int Immunopharmacol. 2011;11(7):856‐861. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Owen DH, Benner B, Pilcher C, et al. Deep and durable response to nivolumab and temozolomide in small‐cell lung cancer associated with an early decrease in myeloid‐derived suppressor cells. Clin Lung Cancer. 2021;22(4):e487‐e497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Zhuo M, Chen H, Zhang T, et al. The potential predictive value of circulating immune cell ratio and tumor marker in atezolizumab treated advanced non‐small cell lung cancer patients. Cancer Biomark. 2018;22(3):467‐476. [DOI] [PubMed] [Google Scholar]
- 12. de Coaña YP, Wolodarski M, Poschke I, et al. Ipilimumab treatment decreases monocytic MDSCs and increases CD8 effector memory T cells in long‐term survivors with advanced melanoma. Oncotarget. 2017;8(13):21539‐21553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Ko JS, Rayman P, Ireland J, et al. Direct and differential suppression of myeloid‐derived suppressor cell subsets by sunitinib is compartmentally constrained. Cancer Res. 2010;70(9):3526‐3536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Gabrilovich DI, Ostrand‐Rosenberg S, Bronte V. Coordinated regulation of myeloid cells by tumours. Nat Rev Immunol. 2012;12(4):253‐268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Dumitru CA, Moses K, Trellakis S, Lang S, Brandau S. Neutrophils and granulocytic myeloid‐derived suppressor cells: immunophenotyping, cell biology and clinical relevance in human oncology. Cancer Immunol Immunother. 2012;61(8):1155‐1167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Coillard A, Segura E. In vivo differentiation of human monocytes. Front Immunol. 2019;10:1907. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Landskron G, De la Fuente M, Thuwajit P, Thuwajit C, Hermoso MA. Chronic inflammation and cytokines in the tumor microenvironment. J Immunol Res. 2014;2014:149185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Marvel D, Gabrilovich DI. Myeloid‐derived suppressor cells in the tumor microenvironment: expect the unexpected. J Clin Invest. 2015;125(9):3356‐3364. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Condamine T, Gabrilovich DI. Molecular mechanisms regulating myeloid‐derived suppressor cell differentiation and function. Trends Immunol. 2011;32(1):19‐25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Condamine T, Mastio J, Gabrilovich DI. Transcriptional regulation of myeloid‐derived suppressor cells. J Leukoc Biol. 2015;98(6):913‐922. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Nefedova Y, Huang M, Kusmartsev S, et al. Hyperactivation of STAT3 is involved in abnormal differentiation of dendritic cells in cancer. J Immunol. 2004;172(1):464‐474. [DOI] [PubMed] [Google Scholar]
- 22. Abad C, Nobuta H, Li J, Kasai A, Yong WH, Waschek JA. Targeted STAT3 disruption in myeloid cells alters immunosuppressor cell abundance in a murine model of spontaneous medulloblastoma. J Leukoc Biol. 2014;95(2):357‐367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Cubillos‐Ruiz JR, Mohamed E, Rodriguez PC. Unfolding anti‐tumor immunity: ER stress responses sculpt tolerogenic myeloid cells in cancer. J Immunother Cancer. 2017;5:5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Grootjans J, Kaser A, Kaufman RJ, Blumberg RS. The unfolded protein response in immunity and inflammation. Nat Rev Immunol. 2016;16(8):469‐484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Mastio J, Condamine T, Dominguez G, et al. Identification of monocyte‐like precursors of granulocytes in cancer as a mechanism for accumulation of PMN‐MDSCs. J Exp Med. 2019;216(9):2150‐2169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Movahedi K, Guilliams M, Van den Bossche J, et al. Identification of discrete tumor‐induced myeloid‐derived suppressor cell subpopulations with distinct T cell‐suppressive activity. Blood. 2008;111(8):4233‐4244. [DOI] [PubMed] [Google Scholar]
- 27. Damuzzo V, Pinton L, Desantis G, et al. Complexity and challenges in defining myeloid‐derived suppressor cells. Cytometry B Clin Cytom. 2015;88(2):77‐91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Haile LA, Gamrekelashvili J, Manns MP, Korangy F, Greten TF. CD49d is a new marker for distinct myeloid‐derived suppressor cell subpopulations in mice. J Immunol. 2010;185(1):203‐210. [DOI] [PubMed] [Google Scholar]
- 29. Mandruzzato S, Brandau S, Britten CM, et al. Toward harmonized phenotyping of human myeloid‐derived suppressor cells by flow cytometry: results from an interim study. Cancer Immunol Immunother. 2016;65(2):161‐169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Condamine T, Dominguez GA, Youn JI, et al. Lectin‐type oxidized LDL receptor‐1 distinguishes population of human polymorphonuclear myeloid‐derived suppressor cells in cancer patients. Sci Immunol. 2016;1(2):aaf8943. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Kattoor AJ, Kanuri SH, Mehta JL. Role of Ox‐LDL and LOX‐1 in atherogenesis. Curr Med Chem. 2019;26(9):1693‐1700. [DOI] [PubMed] [Google Scholar]
- 32. Veglia F, Tyurin VA, Blasi M, et al. Fatty acid transport protein 2 reprograms neutrophils in cancer. Nature. 2019;569(7754):73‐78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Alshetaiwi H, Pervolarakis N, McIntyre LL, et al. Defining the emergence of myeloid‐derived suppressor cells in breast cancer using single‐cell transcriptomics. Sci Immunol. 2020;5(44):eaay6017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Wu Y, Yi M, Niu M, Mei Q, Wu K. Myeloid‐derived suppressor cells: an emerging target for anticancer immunotherapy. Mol Cancer. 2022;21(1):184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Hegde S, Leader AM, Merad M. MDSC: markers, development, states, and unaddressed complexity. Immunity. 2021;54(5):875‐884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Safarzadeh E, Hashemzadeh S, Duijf PHG, et al. Circulating myeloid‐derived suppressor cells: an independent prognostic factor in patients with breast cancer. J Cell Physiol. 2019;234(4):3515‐3525. [DOI] [PubMed] [Google Scholar]
- 37. Zhang B, Wang Z, Wu L, et al. Circulating and tumor‐infiltrating myeloid‐derived suppressor cells in patients with colorectal carcinoma. PLoS One. 2013;8(2):e57114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Cassetta L, Bruderek K, Skrzeczynska‐Moncznik J, et al. Differential expansion of circulating human MDSC subsets in patients with cancer, infection and inflammation. J Immunother Cancer. 2020;8(2):e001223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Cole KE, Ly QP, Hollingsworth MA, et al. Human splenic myeloid derived suppressor cells: phenotypic and clustering analysis. Cell Immunol. 2021;363:104317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Okła K, Czerwonka A, Wawruszak A, et al. Clinical relevance and immunosuppressive pattern of circulating and infiltrating subsets of myeloid‐derived suppressor cells (MDSCs) in epithelial ovarian cancer. Front Immunol. 2019;10:691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Yamauchi Y, Safi S, Blattner C, et al. Circulating and tumor myeloid‐derived suppressor cells in resectable non‐small cell lung cancer. Am J Respir Crit Care Med. 2018;198(6):777‐787. [DOI] [PubMed] [Google Scholar]
- 42. de Goeje PL, Bezemer K, Heuvers ME, et al. Immunoglobulin‐like transcript 3 is expressed by myeloid‐derived suppressor cells and correlates with survival in patients with non‐small cell lung cancer. Oncoimmunology. 2015;4(7):e1014242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Heuvers ME, Muskens F, Bezemer K, et al. Arginase‐1 mRNA expression correlates with myeloid‐derived suppressor cell levels in peripheral blood of NSCLC patients. Lung Cancer. 2013;81(3):468‐474. [DOI] [PubMed] [Google Scholar]
- 44. Nan J, Xing YF, Hu B, et al. Endoplasmic reticulum stress induced LOX‐1(+) CD15(+) polymorphonuclear myeloid‐derived suppressor cells in hepatocellular carcinoma. Immunology. 2018;154(1):144‐155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Li X, Xing YF, Lei AH, et al. Neutrophil count is associated with myeloid derived suppressor cell level and presents prognostic value of for hepatocellular carcinoma patients. Oncotarget. 2017;8(15):24380‐24388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Wu L, Liu H, Guo H, et al. Circulating and tumor‐infiltrating myeloid‐derived suppressor cells in cervical carcinoma patients. Oncol Lett. 2018;15(6):9507‐9515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Yu J, Li H, Zhang Z, Lin W, Wei X, Shao B. Targeting the MDSCs of tumors in situ with inhibitors of the MAPK signaling pathway to promote tumor regression. Front Oncol. 2021;11:647312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Youn JI, Kumar V, Collazo M, et al. Epigenetic silencing of retinoblastoma gene regulates pathologic differentiation of myeloid cells in cancer. Nat Immunol. 2013;14(3):211‐220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Cimen Bozkus C, Elzey BD, Crist SA, Ellies LG, Ratliff TL. Expression of cationic amino acid transporter 2 is required for myeloid‐derived suppressor cell‐mediated control of T cell immunity. J Immunol. 2015;195(11):5237‐5250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Kumar V, Cheng P, Condamine T, et al. CD45 phosphatase inhibits STAT3 transcription factor activity in myeloid cells and promotes tumor‐associated macrophage differentiation. Immunity. 2016;44(2):303‐315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Shao B, Wei X, Luo M, et al. Inhibition of A20 expression in tumor microenvironment exerts anti‐tumor effect through inducing myeloid‐derived suppressor cells apoptosis. Sci Rep. 2015;5:16437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Schlecker E, Stojanovic A, Eisen C, et al. Tumor‐infiltrating monocytic myeloid‐derived suppressor cells mediate CCR5‐dependent recruitment of regulatory T cells favoring tumor growth. J Immunol. 2012;189(12):5602‐5611. [DOI] [PubMed] [Google Scholar]
- 53. Kim R, Hashimoto A, Markosyan N, et al. Ferroptosis of tumour neutrophils causes immune suppression in cancer. Nature. 2022;612(7939):338‐346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Lechner MG, Liebertz DJ, Epstein AL. Characterization of cytokine‐induced myeloid‐derived suppressor cells from normal human peripheral blood mononuclear cells. J Immunol. 2010;185(4):2273‐2284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Sormendi S, Wielockx B. Hypoxia pathway proteins as central mediators of metabolism in the tumor cells and their microenvironment. Front Immunol. 2018;9:40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Roussel M, Ferrell PB, Jr. , Greenplate AR, et al. Mass cytometry deep phenotyping of human mononuclear phagocytes and myeloid‐derived suppressor cells from human blood and bone marrow. J Leukoc Biol. 2017;102(2):437‐447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Ma P, Beatty PL, McKolanis J, Brand R, Schoen RE, Finn OJ. Circulating myeloid derived suppressor cells (MDSC) that accumulate in premalignancy share phenotypic and functional characteristics with MDSC in cancer. Front Immunol. 2019;10:1401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Kimura T, McKolanis JR, Dzubinski LA, et al. MUC1 vaccine for individuals with advanced adenoma of the colon: a cancer immunoprevention feasibility study. Cancer Prev Res (Phila). 2013;6(1):18‐26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Crespo J, Sun H, Welling TH, Tian Z, Zou W. T cell anergy, exhaustion, senescence, and stemness in the tumor microenvironment. Curr Opin Immunol. 2013;25(2):214‐221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Veglia F, Perego M, Gabrilovich D. Myeloid‐derived suppressor cells coming of age. Nat Immunol. 2018;19(2):108‐119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Ghaebi M, Nouri M, Ghasemzadeh A, et al. Immune regulatory network in successful pregnancy and reproductive failures. Biomed Pharmacother. 2017;88:61‐73. [DOI] [PubMed] [Google Scholar]
- 62. Lola C, Bechara M, Marwan M, et al. TIM‐3 regulates innate immune cells to induce fetomaternal tolerance. J Immunol (Baltimore, Md : 1950). 2013;190(1):88‐96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Mengyu S, Ziyang C, Meiqi C, et al. Continuous activation of polymorphonuclear myeloid‐derived suppressor cells during pregnancy is critical for fetal development. Cell Mol Immunol. 2021;18(7):1692‐1707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Natascha K, Christian G. Myeloid‐derived suppressor cells in pregnancy and the neonatal period. Front Immunol. 2020;11:584712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Schwarz J, Scheckenbach V, Kugel H, et al. Granulocytic myeloid‐derived suppressor cells (GR‐MDSC) accumulate in cord blood of preterm infants and remain elevated during the neonatal period. Clin Exp Immunol. 2018;191(3):328‐337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Natascha K, Carolin S, Julian S, et al. Granulocytic myeloid‐derived suppressor cells (GR‐MDSC) in breast milk (BM); GR‐MDSC accumulate in human BM and modulate T‐cell and monocyte function. Front Immunol. 2018;9:1098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. He Y‐M, Li X, Perego M, et al. Transitory presence of myeloid‐derived suppressor cells in neonates is critical for control of inflammation. Nat Med. 2018;24(2):224‐231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Peñaloza HF, Alvarez D, Muñoz‐Durango N, et al. The role of myeloid‐derived suppressor cells in chronic infectious diseases and the current methodology available for their study. J Leukoc Biol. 2019;105(5):857‐872. [DOI] [PubMed] [Google Scholar]
- 69. Malavika M, Sanju S, Poorna MR, et al. Role of myeloid derived suppressor cells in sepsis. Int Immunopharmacol. 2022;104:108452. [DOI] [PubMed] [Google Scholar]
- 70. Zhang W, Fang X, Gao C, et al. MDSCs in sepsis‐induced immunosuppression and its potential therapeutic targets. Cytokine Growth Factor Rev. 2023;69:90‐103. [DOI] [PubMed] [Google Scholar]
- 71. Thakuri BKC, Zhang J, Zhao J, et al. HCV‐associated exosomes upregulate RUNXOR and RUNX1 expressions to promote MDSC expansion and suppressive functions through STAT3‐miR124 axis. Cells. 2020;9(12):2715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Heim CE, Vidlak D, Kielian T. Interleukin‐10 production by myeloid‐derived suppressor cells contributes to bacterial persistence during Staphylococcus aureus orthopedic biofilm infection. J Leukoc Biol. 2015;98(6):1003‐1013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Yamada KJ, Heim CE, Aldrich AL, Gries CM, Staudacher AG, Kielian T. Arginase‐1 expression in myeloid cells regulates Staphylococcus aureus planktonic but not biofilm infection. Infect Immun. 2018;86(7):e00206‐18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Ren JP, Zhao J, Dai J, et al. Hepatitis C virus‐induced myeloid‐derived suppressor cells regulate T‐cell differentiation and function via the signal transducer and activator of transcription 3 pathway. Immunology. 2016;148(4):377‐386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Tacke RS, Lee HC, Goh C, et al. Myeloid suppressor cells induced by hepatitis C virus suppress T‐cell responses through the production of reactive oxygen species. Hepatology. 2012;55(2):343‐353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Heim CE, Vidlak D, Scherr TD, et al. Myeloid‐derived suppressor cells contribute to Staphylococcus aureus orthopedic biofilm infection. J Immunol. 2014;192(8):3778‐3792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Bertrand BP, Heim CE, West SC, et al. Role of Staphylococcus aureus formate metabolism during prosthetic joint infection. Infect Immun. 2022;90(11):e0042822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Kumar N, Khan N, Cleveland D, Geiger JD. A common approach for fighting tuberculosis and leprosy: controlling endoplasmic reticulum stress in myeloid‐derived suppressor cells. Immunotherapy. 2021;13(18):1555‐1563. [DOI] [PubMed] [Google Scholar]
- 79. Kelly‐Scumpia KM, Choi A, Shirazi R, et al. ER stress regulates immunosuppressive function of myeloid derived suppressor cells in leprosy that can be overcome in the presence of IFN‐γ. iScience. 2020;23(5):101050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Ruan WS, Feng MX, Xu J, et al. Early activation of myeloid‐derived suppressor cells participate in sepsis‐induced immune suppression via PD‐L1/PD‐1 Axis. Front Immunol. 2020;11:1299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Zhang Y, Wu Y, Liu H, et al. Granulocytic myeloid‐derived suppressor cells inhibit T follicular helper cells during experimental Schistosoma japonicum infection. Parasit Vectors. 2021;14(1):497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Wang L, Zhao J, Ren JP, et al. Expansion of myeloid‐derived suppressor cells promotes differentiation of regulatory T cells in HIV‐1+ individuals. AIDS. 2016;30(10):1521‐1531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Vollbrecht T, Stirner R, Tufman A, et al. Chronic progressive HIV‐1 infection is associated with elevated levels of myeloid‐derived suppressor cells. AIDS. 2012;26(12):F31‐F37. [DOI] [PubMed] [Google Scholar]
- 84. Macatangay BJ, Landay AL, Rinaldo CR. MDSC: a new player in HIV immunopathogenesis. AIDS. 2012;26(12):1567‐1569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Takano T, Matsumura T, Adachi Y, et al. Myeloid cell dynamics correlating with clinical outcomes of severe COVID‐19 in Japan. Int Immunol. 2021;33(4):241‐247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Feldmann M, Brennan FM, Maini RN. Rheumatoid arthritis. Cell. 1996;85(3):307‐310. [DOI] [PubMed] [Google Scholar]
- 87. So EY, Sun C, Wu KQ, Dubielecka PM, Reginato AM, Liang OD. Inhibition of lipid phosphatase SHIP1 expands myeloid‐derived suppressor cells and attenuates rheumatoid arthritis in mice. Am J Physiol Cell Physiol. 2021;321(3):C569‐C584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Gizinski AM, Fox DA. T cell subsets and their role in the pathogenesis of rheumatic disease. Curr Opin Rheumatol. 2014;26(2):204‐210. [DOI] [PubMed] [Google Scholar]
- 89. Wang W, Jiao Z, Duan T, et al. Functional characterization of myeloid‐derived suppressor cell subpopulations during the development of experimental arthritis. Eur J Immunol. 2015;45(2):464‐473. [DOI] [PubMed] [Google Scholar]
- 90. Fujii W, Ashihara E, Hirai H, et al. Myeloid‐derived suppressor cells play crucial roles in the regulation of mouse collagen‐induced arthritis. J Immunol. 2013;191(3):1073‐1081. [DOI] [PubMed] [Google Scholar]
- 91. Kurkó J, Vida A, Glant TT, et al. Identification of myeloid‐derived suppressor cells in the synovial fluid of patients with rheumatoid arthritis: a pilot study. BMC Musculoskelet Disord. 2014;15:281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Crook KR, Jin M, Weeks MF, et al. Myeloid‐derived suppressor cells regulate T cell and B cell responses during autoimmune disease. J Leukoc Biol. 2015;97(3):573‐582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Park MJ, Lee SH, Kim EK, et al. Interleukin‐10 produced by myeloid‐derived suppressor cells is critical for the induction of Tregs and attenuation of rheumatoid inflammation in mice. Sci Rep. 2018;8(1):3753. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Perfilyeva YV, Abdolla N, Ostapchuk YO, Tleulieva R, Krasnoshtanov VC, Belyaev NN. Expansion of CD11b(+)Ly6G(high) and CD11b(+)CD49d(+) myeloid cells with suppressive potential in mice with chronic inflammation and light‐at‐night‐induced circadian disruption. Inflamm Res. 2017;66(8):711‐724. [DOI] [PubMed] [Google Scholar]
- 95. Wu X, Zhu D, Tian J, et al. Granulocytic myeloid‐derived suppressor cell exosomal prostaglandin E2 ameliorates collagen‐induced arthritis by enhancing IL‐10(+) B cells. Front Immunol. 2020;11:588500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Jiao Z, Hua S, Wang W, Wang H, Gao J, Wang X. Increased circulating myeloid‐derived suppressor cells correlated negatively with Th17 cells in patients with rheumatoid arthritis. Scand J Rheumatol. 2013;42(2):85‐90. [DOI] [PubMed] [Google Scholar]
- 97. Guo C, Hu F, Yi H, et al. Myeloid‐derived suppressor cells have a proinflammatory role in the pathogenesis of autoimmune arthritis. Ann Rheum Dis. 2016;75(1):278‐285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Zhang H, Wang S, Huang Y, et al. Myeloid‐derived suppressor cells are proinflammatory and regulate collagen‐induced arthritis through manipulating Th17 cell differentiation. Clin Immunol. 2015;157(2):175‐186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Chen S, Guo C, Wang R, et al. Monocytic MDSCs skew Th17 cells toward a pro‐osteoclastogenic phenotype and potentiate bone erosion in rheumatoid arthritis. Rheumatology (Oxford). 2021;60(5):2409‐2420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Wu H, Zhen Y, Ma Z, et al. Arginase‐1‐dependent promotion of TH17 differentiation and disease progression by MDSCs in systemic lupus erythematosus. Sci Transl Med. 2016;8(331):331ra40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Li X, Fei F, Yao G, et al. Notch1 signalling controls the differentiation and function of myeloid‐derived suppressor cells in systemic lupus erythematosus. Immunology. 2023;168(1):170‐183. [DOI] [PubMed] [Google Scholar]
- 102. Pang B, Zhen Y, Hu C, Ma Z, Lin S, Yi H. Myeloid‐derived suppressor cells shift Th17/Treg ratio and promote systemic lupus erythematosus progression through arginase‐1/miR‐322‐5p/TGF‐β pathway. Clin Sci (Lond). 2020;134(16):2209‐2222. [DOI] [PubMed] [Google Scholar]
- 103. Yi H, Guo C, Yu X, Zuo D, Wang XY. Mouse CD11b+Gr‐1+ myeloid cells can promote Th17 cell differentiation and experimental autoimmune encephalomyelitis. J Immunol. 2012;189(9):4295‐4304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Glenn JD, Liu C, Whartenby KA. Frontline Science: Induction of experimental autoimmune encephalomyelitis mobilizes Th17‐promoting myeloid derived suppressor cells to the lung. J Leukoc Biol. 2019;105(5):829‐841. [DOI] [PubMed] [Google Scholar]
- 105. Melero‐Jerez C, Fernández‐Gómez B, Lebrón‐Galán R, et al. Myeloid‐derived suppressor cells support remyelination in a murine model of multiple sclerosis by promoting oligodendrocyte precursor cell survival, proliferation, and differentiation. Glia. 2021;69(4):905‐924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Yang T, Li J, Li R, et al. Correlation between MDSC and immune tolerance in transplantation: cytokines, pathways and cell‐cell interaction. Curr Gene Ther. 2019;19(2):81–92. [DOI] [PubMed] [Google Scholar]
- 107. Jordi O, Patricia C, Alberto UR, Estela PA. Tolerogenic role of myeloid suppressor cells in organ transplantation. Front Immunol. 2019;10:374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Wang K, Lv M, Chang YJ, et al. Early myeloid‐derived suppressor cells (HLA‐DR(‐)/(low)CD33(+)CD16(‐)) expanded by granulocyte colony‐stimulating factor prevent acute graft‐versus‐host disease (GVHD) in humanized mouse and might contribute to lower GVHD in patients post allo‐HSCT. J Hematol Oncol. 2019;12(1):31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Tobias H, Anurag S, Berta SG, et al. Myeloid‐derived suppressor cells in lung transplantation. Front Immunol. 2019;10:900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Okano S, Abu‐Elmagd K, Kish DD, et al. Myeloid‐derived suppressor cells increase and inhibit donor‐reactive T cell responses to graft intestinal epithelium in intestinal transplant patients. Am J Transplant. 2018;18(10):2544‐2558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Iglesias‐Escudero M, Sansegundo‐Arribas D, Riquelme P, et al. Myeloid‐derived suppressor cells in kidney transplant recipients and the effect of maintenance immunotherapy. Front Immunol. 2020;11:643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Xu J, Pei S, Wang Y, et al. Tpl2 protects against fulminant hepatitis through mobilization of myeloid‐derived suppressor cells. Front Immunol. 2019;10:1980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Martins NS, de Campos Fraga‐Silva TF, Correa GF, et al. Artepillin C reduces allergic airway inflammation by induction of monocytic myeloid‐derived suppressor cells. Pharmaceutics. 2021;13(11):1763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Piñeros AR, de Lima MHF, Rodrigues T, et al. Green propolis increases myeloid suppressor cells and CD4Foxp3 cells and reduces Th2 inflammation in the lungs after allergen exposure. J Ethnopharmacol. 2020;252:112496. [DOI] [PubMed] [Google Scholar]
- 115. Zhou L, Miao K, Yin B, et al. Cardioprotective role of myeloid‐derived suppressor cells in heart failure. Circulation. 2018;138(2):181‐197. [DOI] [PubMed] [Google Scholar]
- 116. Morales JK, Kmieciak M, Knutson KL, Bear HD, Manjili MH. GM‐CSF is one of the main breast tumor‐derived soluble factors involved in the differentiation of CD11b‐Gr1‐ bone marrow progenitor cells into myeloid‐derived suppressor cells. Breast Cancer Res Treat. 2010;123(1):39‐49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Chen J, Deng C, Shi Q, et al. CpG oligodeoxynucleotide induces bone marrow precursor cells into myeloid‐derived suppressor cells. Mol Med Rep. 2013;8(4):1149‐1154. [DOI] [PubMed] [Google Scholar]
- 118. Liu Y, Wei J, Guo G, Zhou J. Norepinephrine‐induced myeloid‐derived suppressor cells block T‐cell responses via generation of reactive oxygen species. Immunopharmacol Immunotoxicol. 2015;37(4):359‐365. [DOI] [PubMed] [Google Scholar]
- 119. Yang F, Li Y, Zou W, et al. Adoptive transfer of IFN‐γ‐induced M‐MDSCs promotes immune tolerance to allografts through iNOS pathway. Inflamm Res. 2019;68(7):545‐555. [DOI] [PubMed] [Google Scholar]
- 120. Scheurer J, Kitt K, Huber HJ, et al. Graft‐versus‐host disease prevention by in vitro‐generated myeloid‐derived suppressor cells is exclusively mediated by the CD11b+CD11c+ MDSC subpopulation. Front Immunol. 2021;12:754316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Wang H, Ji J, Zhuang Y, Zhou X, Zhao Y, Zhang X. PMA induces the differentiation of monocytes into immunosuppressive MDSCs. Clin Exp Immunol. 2021;206(2):216‐225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Sarra M, Cupi ML, Bernardini R, et al. IL‐25 prevents and cures fulminant hepatitis in mice through a myeloid‐derived suppressor cell‐dependent mechanism. Hepatology. 2013;58(4):1436‐1450. [DOI] [PubMed] [Google Scholar]
- 123. Li J, Tu G, Zhang W, et al. CHBP induces stronger immunosuppressive CD127 M‐MDSC via erythropoietin receptor. Cell Death Dis. 2021;12(2):177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Oberholtzer N, Atkinson C, Nadig SN. Adoptive transfer of regulatory immune cells in organ transplantation. Front Immunol. 2021;12:631365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Qiu J, Yang G, Feng M, et al. Extracellular vesicles as mediators of the progression and chemoresistance of pancreatic cancer and their potential clinical applications. Mol Cancer. 2018;17(1):2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Su YL, Banerjee S, White SV, Kortylewski M. STAT3 in tumor‐associated myeloid cells: multitasking to disrupt immunity. Int J Mol Sci. 2018;19(6):1803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Budhwar S, Verma P, Verma R, Rai S, Singh K. The Yin and Yang of myeloid derived suppressor cells. Front Immunol. 2018;9:2776. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Noman MZ, Desantis G, Janji B, et al. PD‐L1 is a novel direct target of HIF‐1alpha, and its blockade under hypoxia enhanced MDSC‐mediated T cell activation. J Exp Med. 2014;211(5):781‐790. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Tsukamoto H, Nishikata R, Senju S, Nishimura Y. Myeloid‐derived suppressor cells attenuate TH1 development through IL‐6 production to promote tumor progression. Cancer Immunol Res. 2013;1(1):64‐76. [DOI] [PubMed] [Google Scholar]
- 130. Li F, Zhao Y, Wei L, Li S, Liu J. Tumor‐infiltrating Treg, MDSC, and IDO expression associated with outcomes of neoadjuvant chemotherapy of breast cancer. Cancer Biol Ther. 2018;19(8):695‐705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Yang Z, Guo J, Weng L, Tang W, Jin S, Ma W. Myeloid‐derived suppressor cells—new and exciting players in lung cancer. J Hematol Oncol. 2020;13(1):10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Zhao H, Teng D, Yang L, et al. Myeloid‐derived itaconate suppresses cytotoxic CD8(+) T cells and promotes tumour growth. Nat Metab. 2022;4(12):1660‐1673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Szefel J, Danielak A, Kruszewski WJ. Metabolic pathways of L‐arginine and therapeutic consequences in tumors. Adv Med Sci. 2019;64(1):104‐110. [DOI] [PubMed] [Google Scholar]
- 134. Dar AA, Patil RS, Pradhan TN, Chaukar DA, D'Cruz AK, Chiplunkar SV. Myeloid‐derived suppressor cells impede T cell functionality and promote Th17 differentiation in oral squamous cell carcinoma. Cancer Immunol Immunother. 2020;69(6):1071‐1086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. DA A, PR S, PT N, CD A, DCA K, CS V. Myeloid‐derived suppressor cells impede T cell functionality and promote Th17 differentiation in oral squamous cell carcinoma. Cancer Immunol Immunother. 2020;69(6):1071‐1086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. 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. Oncoimmunology. 2018;7(9):e1469594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Ugel S, De Sanctis F, Mandruzzato S, Bronte V. Tumor‐induced myeloid deviation: when myeloid‐derived suppressor cells meet tumor‐associated macrophages. J Clin Invest. 2015;125(9):3365‐3376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Sinha P, Clements VK, Bunt SK, Albelda SM, Ostrand‐Rosenberg S. Cross‐talk between myeloid‐derived suppressor cells and macrophages subverts tumor immunity toward a type 2 response. J Immunol. 2007;179(2):977‐983. [DOI] [PubMed] [Google Scholar]
- 139. Yu J, Du W, Yan F, et al. Myeloid‐derived suppressor cells suppress antitumor immune responses through IDO expression and correlate with lymph node metastasis in patients with breast cancer. J Immunol. 2013;190(7):3783‐3797. [DOI] [PubMed] [Google Scholar]
- 140. Kwak T, Wang F, Deng H, et al. Distinct populations of immune‐suppressive macrophages differentiate from monocytic myeloid‐derived suppressor cells in cancer. Cell Rep. 2020;33(13):108571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Duhan V, Smyth MJ. Innate myeloid cells in the tumor microenvironment. Curr Opin Immunol. 2021;69:18‐28. [DOI] [PubMed] [Google Scholar]
- 142. Mabuchi S, Sasano T. Myeloid‐derived suppressor cells as therapeutic targets in uterine cervical and endometrial cancers. Cells. 2021;10(5):1073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Dysthe M, Parihar R. Myeloid‐Derived Suppressor Cells in the Tumor Microenvironment. Adv Exp Med Biol. 2020;1224:117‐140. [DOI] [PubMed] [Google Scholar]
- 144. Yang Y, Cao Y. The impact of VEGF on cancer metastasis and systemic disease. Semin Cancer Biol. 2022;86(Pt 3):251‐261. [DOI] [PubMed] [Google Scholar]
- 145. Yang J, Yan J, Liu B. Targeting VEGF/VEGFR to Modulate Antitumor Immunity. Front Immunol. 2018;9:978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Ridge SM, Sullivan FJ, Glynn SA. Mesenchymal stem cells: key players in cancer progression. Mol Cancer. 2017;16(1):31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Albini A, Bruno A, Noonan DM, Mortara L. Contribution to tumor angiogenesis from innate immune cells within the tumor microenvironment: implications for immunotherapy. Front Immunol. 2018;9:527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Parihar R, Rivas C, Huynh M, et al. NK Cells Expressing a Chimeric Activating Receptor Eliminate MDSCs and Rescue Impaired CAR‐T Cell Activity against Solid Tumors. Cancer Immunol Res. 2019;7(3):363‐375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Shojaei F, Wu X, Qu X, et al. G‐CSF‐initiated myeloid cell mobilization and angiogenesis mediate tumor refractoriness to anti‐VEGF therapy in mouse models. Proc Natl Acad Sci USA. 2009;106(16):6742‐6747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Wang J, Su X, Yang L, et al. The influence of myeloid‐derived suppressor cells on angiogenesis and tumor growth after cancer surgery. Int J Cancer. 2016;138(11):2688‐2699. [DOI] [PubMed] [Google Scholar]
- 151. Krebs R, Tikkanen JM, Raissadati A, Hollmen M, Dhaygude K, Lemstrom KB. Inhibition of vascular endothelial growth factor receptors 1 and 2 attenuates natural killer cell and innate immune responses in an experimental model for obliterative bronchiolitis. Am J Pathol. 2022;192(2):254‐269. [DOI] [PubMed] [Google Scholar]
- 152. MaruYama T, Chen W, Shibata H. TGF‐β and cancer immunotherapy. Biol Pharm Bull. 2022;45(2):155‐161. [DOI] [PubMed] [Google Scholar]
- 153. Horn LA, Chariou PL, Gameiro SR, et al. Remodeling the tumor microenvironment via blockade of LAIR‐1 and TGF‐beta signaling enables PD‐L1‐mediated tumor eradication. J Clin Invest. 2022;132(8):e155148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Gulley JL, Schlom J, Barcellos‐Hoff MH, et al. Dual inhibition of TGF‐beta and PD‐L1: a novel approach to cancer treatment. Mol Oncol. 2021;16(11):2117‐2134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Hao Y, Baker D, Ten Dijke P. TGF‐beta‐mediated epithelial‐mesenchymal transition and cancer metastasis. Int J Mol Sci. 2019;20(11):2767. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Navarro R, Tapia‐Galisteo A, Martin‐Garcia L, et al. TGF‐beta‐induced IGFBP‐3 is a key paracrine factor from activated pericytes that promotes colorectal cancer cell migration and invasion. Mol Oncol. 2020;14(10):2609‐2628. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157. Fu XG, Deng J, Xu WJ, Chen JY, Sun J, Deng H. Histidine decarboxylase‐expressing PMN‐MDSC‐derived TGF‐β1 promotes the epithelial‐mesenchymal transition of metastatic lung adenocarcinoma. Int J Clin Exp Pathol. 2020;13(6):1361‐1371. [PMC free article] [PubMed] [Google Scholar]
- 158. Jayakumar A, Bothwell ALM. Functional diversity of myeloid‐derived suppressor cells: the multitasking hydra of cancer. J Immunol. 2019;203(5):1095‐1103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159. Chan YLT, Zuo J, Inman C, et al. NK cells produce high levels of IL‐10 early after allogeneic stem cell transplantation and suppress development of acute GVHD. Eur J Immunol. 2018;48(2):316‐329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160. Stirm K, Leary P, Bertram K, et al. Tumor cell‐derived IL‐10 promotes cell‐autonomous growth and immune escape in diffuse large B‐cell lymphoma. Oncoimmunology. 2021;10(1):2003533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Lin Y, Li B, Yang X, et al. Fibroblastic FAP promotes intrahepatic cholangiocarcinoma growth via MDSCs recruitment. Neoplasia. 2019;21(12):1133‐1142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Subramaniam KS, Omar IS, Kwong SC, et al. Cancer‐associated fibroblasts promote endometrial cancer growth via activation of interleukin‐6/STAT‐3/c‐Myc pathway. Am J Cancer Res. 2016;6(2):200‐213. [PMC free article] [PubMed] [Google Scholar]
- 163. Weber R, Groth C, Lasser S, et al. IL‐6 as a major regulator of MDSC activity and possible target for cancer immunotherapy. Cell Immunol. 2021;359:104254. [DOI] [PubMed] [Google Scholar]
- 164. Liu H, Shen J, Lu K. IL‐6 and PD‐L1 blockade combination inhibits hepatocellular carcinoma cancer development in mouse model. Biochem Biophys Res Commun. 2017;486(2):239‐244. [DOI] [PubMed] [Google Scholar]
- 165. Pan S, Hu Y, Hu M, et al. S100A8 facilitates cholangiocarcinoma metastasis via upregulation of VEGF through TLR4/NFkappaB pathway activation. Int J Oncol. 2020;56(1):101‐112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. Eisenblaetter M, Flores‐Borja F, Lee JJ, et al. Visualization of tumor‐immune interaction ‐ target‐specific imaging of S100A8/A9 reveals pre‐metastatic niche establishment. Theranostics. 2017;7(9):2392‐2401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167. Chen X, Eksioglu EA, Zhou J, et al. Induction of myelodysplasia by myeloid‐derived suppressor cells. J Clin Invest. 2013;123(11):4595‐4611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168. Zhulai G, Oleinik E, Shibaev M, Ignatev K. Adenosine‐metabolizing enzymes, adenosine kinase and adenosine deaminase, in cancer. Biomolecules. 2022;12(3):418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169. Koh J, Kim Y, Lee KY, et al. MDSC subtypes and CD39 expression on CD8(+) T cells predict the efficacy of anti‐PD‐1 immunotherapy in patients with advanced NSCLC. Eur J Immunol. 2020;50(11):1810‐1819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170. Augustin RC, Leone RD, Naing A, Fong L, Bao R, Luke JJ. Next steps for clinical translation of adenosine pathway inhibition in cancer immunotherapy. J Immunother Cancer. 2022;10(2):e004089. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171. Mantovani A, Barajon I, Garlanda C. IL‐1 and IL‐1 regulatory pathways in cancer progression and therapy. Immunol Rev. 2018;281(1):57‐61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172. Voronov E, Shouval DS, Krelin Y, et al. IL‐1 is required for tumor invasiveness and angiogenesis. Proc Natl Acad Sci U S A. 2003;100(5):2645‐2650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173. Di Mitri D, Toso A, Chen JJ, et al. Tumour‐infiltrating Gr‐1+ myeloid cells antagonize senescence in cancer. Nature. 2014;515(7525):134‐137. [DOI] [PubMed] [Google Scholar]
- 174. Wolf JS, Chen Z, Dong G, et al. IL (interleukin)‐1alpha promotes nuclear factor‐kappaB and AP‐1‐induced IL‐8 expression, cell survival, and proliferation in head and neck squamous cell carcinomas. Clin Cancer Res. 2001;7(6):1812‐1820. [PubMed] [Google Scholar]
- 175. Shostak K, Chariot A. EGFR and NF‐kappaB: partners in cancer. Trends Mol Med. 2015;21(6):385‐393. [DOI] [PubMed] [Google Scholar]
- 176. Umansky V, Blattner C, Gebhardt C, Utikal J. The role of myeloid‐derived suppressor cells (MDSC) in cancer progression. Vaccines (Basel). 2016;4(4):36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177. Vaddepally RK, Kharel P, Pandey R, Garje R, Chandra AB. Review of indications of FDA‐approved immune checkpoint inhibitors per NCCN guidelines with the level of evidence. Cancers (Basel). 2020;12(3):738. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178. Oikawa D, Sato Y, Ohtake F, et al. Molecular bases for HOIPINs‐mediated inhibition of LUBAC and innate immune responses. Commun Biol. 2020;3(1):163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179. Noman MZ, Desantis G, Janji B, et al. PD‐L1 is a novel direct target of HIF‐1α, and its blockade under hypoxia enhanced MDSC‐mediated T cell activation. J Exp Med. 2014;211(5):781‐790. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180. Tzeng A, Diaz‐Montero CM, Rayman PA, et al. Immunological correlates of response to immune checkpoint inhibitors in metastatic urothelial carcinoma. Target Oncol. 2018;13(5):599‐609. [DOI] [PubMed] [Google Scholar]
- 181. Passaro A, Mancuso P, Gandini S, et al. Gr‐MDSC‐linked asset as a potential immune biomarker in pretreated NSCLC receiving nivolumab as second‐line therapy. Clin Transl Oncol. 2020;22(4):603‐611. [DOI] [PubMed] [Google Scholar]
- 182. Pires da Silva I, Lo S, Quek C, et al. Site‐specific response patterns, pseudoprogression, and acquired resistance in patients with melanoma treated with ipilimumab combined with anti‐PD‐1 therapy. Cancer. 2020;126(1):86‐97. [DOI] [PubMed] [Google Scholar]
- 183. Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune correlates of anti‐PD‐1 antibody in cancer. N Engl J Med. 2012;366(26):2443‐2454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184. Holokai L, Chakrabarti J, Lundy J, et al. Murine‐ and human‐derived autologous organoid/immune cell co‐cultures as pre‐clinical models of pancreatic ductal adenocarcinoma. Cancers (Basel). 2020;12(12):3816. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185. Görgün G, Samur MK, Cowens KB, et al. Lenalidomide enhances immune checkpoint blockade‐induced immune response in multiple myeloma. Clin Cancer Res. 2015;21(20):4607‐4618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186. Limagne E, Richard C, Thibaudin M, et al. Tim‐3/galectin‐9 pathway and mMDSC control primary and secondary resistances to PD‐1 blockade in lung cancer patients. Oncoimmunology. 2019;8(4):e1564505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187. Gao F, Yang C. Anti‐VEGF/VEGFR2 monoclonal antibodies and their combinations with PD‐1/PD‐L1 inhibitors in clinic. Curr Cancer Drug Targets. 2020;20(1):3‐18. [DOI] [PubMed] [Google Scholar]
- 188. Pico de Coaña Y, Wolodarski M, van der Haar Àvila I, et al. PD‐1 checkpoint blockade in advanced melanoma patients: NK cells, monocytic subsets and host PD‐L1 expression as predictive biomarker candidates. Oncoimmunology. 2020;9(1):1786888. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189. Pico de Coaña Y, Masucci G, Hansson J, Kiessling R. Myeloid‐derived suppressor cells and their role in CTLA‐4 blockade therapy. Cancer Immunol Immunother. 2014;63(9):977‐983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190. Meyer C, Cagnon L, Costa‐Nunes CM, et al. Frequencies of circulating MDSC correlate with clinical outcome of melanoma patients treated with ipilimumab. Cancer Immunol Immunother. 2014;63(3):247‐257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191. Tarhini AA, Edington H, Butterfield LH, et al. Immune monitoring of the circulation and the tumor microenvironment in patients with regionally advanced melanoma receiving neoadjuvant ipilimumab. PLoS One. 2014;9(2):e87705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192. Bjoern J, Juul Nitschke N, Zeeberg Iversen T, Schmidt H, Fode K, Svane IM. Immunological correlates of treatment and response in stage IV malignant melanoma patients treated with Ipilimumab. Oncoimmunology. 2016;5(4):e1100788. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193. Sade‐Feldman M, Kanterman J, Klieger Y, et al. Clinical significance of circulating CD33+CD11b+HLA‐DR‐ myeloid cells in patients with stage IV melanoma treated with ipilimumab. Clin Cancer Res. 2016;22(23):5661‐5672. [DOI] [PubMed] [Google Scholar]
- 194. Kim K, Skora AD, Li Z, et al. Eradication of metastatic mouse cancers resistant to immune checkpoint blockade by suppression of myeloid‐derived cells. Proc Natl Acad Sci USA. 2014;111(32):11774‐11779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195. Holmgaard RB, Brachfeld A, Gasmi B, et al. Timing of CSF‐1/CSF‐1R signaling blockade is critical to improving responses to CTLA‐4 based immunotherapy. Oncoimmunology. 2016;5(7):e1151595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196. Du Four S, Maenhout SK, Niclou SP, Thielemans K, Neyns B, Aerts JL. Combined VEGFR and CTLA‐4 blockade increases the antigen‐presenting function of intratumoral DCs and reduces the suppressive capacity of intratumoral MDSCs. Am J Cancer Res. 2016;6(11):2514‐2531. [PMC free article] [PubMed] [Google Scholar]
- 197. Rahma OE, Hodi FS. The intersection between tumor angiogenesis and immune suppression. Clin Cancer Res. 2019;25(18):5449‐5457. [DOI] [PubMed] [Google Scholar]
- 198. Melani C, Chiodoni C, Forni G, Colombo MP. Myeloid cell expansion elicited by the progression of spontaneous mammary carcinomas in c‐erbB‐2 transgenic BALB/c mice suppresses immune reactivity. Blood. 2003;102(6):2138‐2145. [DOI] [PubMed] [Google Scholar]
- 199. Tartour E, Pere H, Maillere B, et al. Angiogenesis and immunity: a bidirectional link potentially relevant for the monitoring of antiangiogenic therapy and the development of novel therapeutic combination with immunotherapy. Cancer Metastasis Rev. 2011;30(1):83‐95. [DOI] [PubMed] [Google Scholar]
- 200. Feng PH, Chen KY, Huang YC, et al. Bevacizumab reduces S100A9‐positive MDSCs linked to intracranial control in patients with EGFR‐mutant lung adenocarcinoma. J Thorac Oncol. 2018;13(7):958‐967. [DOI] [PubMed] [Google Scholar]
- 201. Peereboom DM, Alban TJ, Grabowski MM, et al. Metronomic capecitabine as an immune modulator in glioblastoma patients reduces myeloid‐derived suppressor cells. JCI Insight. 2019;4(22):e130748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202. Koinis F, Vetsika EK, Aggouraki D, et al. Effect of first‐line treatment on myeloid‐derived suppressor cells' subpopulations in the peripheral blood of patients with non‐small cell lung cancer. J Thorac Oncol. 2016;11(8):1263‐1272. [DOI] [PubMed] [Google Scholar]
- 203. Horikawa N, Abiko K, Matsumura N, et al. Anti‐VEGF therapy resistance in ovarian cancer is caused by GM‐CSF‐induced myeloid‐derived suppressor cell recruitment. Br J Cancer. 2020;122(6):778‐788. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204. Iwai T, Harada Y, Saeki H, Oki E, Maehara Y, Yonemitsu Y. Capecitabine reverses tumor escape from anti‐VEGF through the eliminating CD11b(high)/Gr1(high) myeloid cells. Oncotarget. 2018;9(25):17620‐17630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205. Shayan G, Kansy BA, Gibson SP, et al. Phase Ib study of immune biomarker modulation with neoadjuvant cetuximab and TLR8 stimulation in head and neck cancer to overcome suppressive myeloid signals. Clin Cancer Res. 2018;24(1):62‐72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206. Li J, Srivastava RM, Ettyreddy A, Ferris RL. Cetuximab ameliorates suppressive phenotypes of myeloid antigen presenting cells in head and neck cancer patients. J Immunother Cancer. 2015;3:54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207. Thakur A, Schalk D, Tomaszewski E, et al. Microenvironment generated during EGFR targeted killing of pancreatic tumor cells by ATC inhibits myeloid‐derived suppressor cells through COX2 and PGE2 dependent pathway. J Transl Med. 2013;11:35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208. McMichael EL, Benner B, Atwal LS, et al. A phase I/II trial of cetuximab in combination with interleukin‐12 administered to patients with unresectable primary or recurrent head and neck squamous cell carcinoma. Clin Cancer Res. 2019;25(16):4955‐4965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209. Sampath S, Won H, Massarelli E, et al. Combined modality radiation therapy promotes tolerogenic myeloid cell populations and STAT3‐related gene expression in head and neck cancer patients. Oncotarget. 2018;9(13):11279‐11290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210. Roskoski R, Jr. Sunitinib: a VEGF and PDGF receptor protein kinase and angiogenesis inhibitor. Biochem Biophys Res Commun. 2007;356(2):323‐328. [DOI] [PubMed] [Google Scholar]
- 211. Kujawski M, Kortylewski M, Lee H, Herrmann A, Kay H, Yu H. Stat3 mediates myeloid cell‐dependent tumor angiogenesis in mice. J Clin Invest. 2008;118(10):3367‐3377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212. Hellsten R, Lilljebjörn L, Johansson M, Leandersson K, Bjartell A. The STAT3 inhibitor galiellalactone inhibits the generation of MDSC‐like monocytes by prostate cancer cells and decreases immunosuppressive and tumorigenic factors. Prostate. 2019;79(14):1611‐1621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213. Pan PY, Wang GX, Yin B, et al. Reversion of immune tolerance in advanced malignancy: modulation of myeloid‐derived suppressor cell development by blockade of stem‐cell factor function. Blood. 2008;111(1):219‐228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214. Ozao‐Choy J, Ma G, Kao J, et al. The novel role of tyrosine kinase inhibitor in the reversal of immune suppression and modulation of tumor microenvironment for immune‐based cancer therapies. Cancer Res. 2009;69(6):2514‐22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215. Cohen PA, Ko JS, Storkus WJ, et al. Myeloid‐derived suppressor cells adhere to physiologic STAT3‐ vs STAT5‐dependent hematopoietic programming, establishing diverse tumor‐mediated mechanisms of immunologic escape. Immunol Invest. 2012;41(6‐7):680‐710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216. Maenhout SK, Van Lint S, Emeagi PU, Thielemans K, Aerts JL. Enhanced suppressive capacity of tumor‐infiltrating myeloid‐derived suppressor cells compared with their peripheral counterparts. Int J Cancer. 2014;134(5):1077‐1090. [DOI] [PubMed] [Google Scholar]
- 217. Kumar V, Patel S, Tcyganov E, Gabrilovich DI. The nature of myeloid‐derived suppressor cells in the tumor microenvironment. Trends Immunol. 2016;37(3):208‐220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218. Stiff A, Trikha P, Wesolowski R, et al. Myeloid‐derived suppressor cells express bruton's tyrosine kinase and can be depleted in tumor‐bearing hosts by ibrutinib treatmENT. Cancer Res. 2016;76(8):2125‐2136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219. Khaki Bakhtiarvand V, Ramezani‐Ali Akbari K, Amir Jalali S, et al. Myeloid‐derived suppressor cells (MDSCs) depletion by cabozantinib improves the efficacy of anti‐HER2 antibody‐based immunotherapy in a 4T1‐HER2 murine breast cancer model. Int Immunopharmacol. 2022;113(Pt B):109470. [DOI] [PubMed] [Google Scholar]
- 220. Heine A, Schilling J, Grünwald B, et al. The induction of human myeloid derived suppressor cells through hepatic stellate cells is dose‐dependently inhibited by the tyrosine kinase inhibitors nilotinib, dasatinib and sorafenib, but not sunitinib. Cancer Immunol Immunother. 2016;65(3):273‐282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221. Mollica Poeta V, Massara M, Capucetti A, Bonecchi R. Chemokines and chemokine receptors: new targets for cancer immunotherapy. Front Immunol. 2019;10:379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222. Korbecki J, Kojder K, Kapczuk P, et al. The effect of hypoxia on the expression of CXC chemokines and CXC chemokine receptors—a review of literature. Int J Mol Sci. 2021;22(2):843. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223. Greene S, Robbins Y, Mydlarz WK, et al. Inhibition of MDSC trafficking with SX‐682, a CXCR1/2 inhibitor, enhances NK‐cell immunotherapy in head and neck cancer models. Clin Cancer Res. 2020;26(6):1420‐1431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224. Sun L, Clavijo PE, Robbins Y, et al. Inhibiting myeloid‐derived suppressor cell trafficking enhances T cell immunotherapy. JCI Insight. 2019;4(7):e126853. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225. Steele CW, Karim SA, Leach JDG, et al. CXCR2 inhibition profoundly suppresses metastases and augments immunotherapy in pancreatic ductal adenocarcinoma. Cancer Cell. 2016;29(6):832‐845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226. Highfill SL, Cui Y, Giles AJ, et al. Disruption of CXCR2‐mediated MDSC tumor trafficking enhances anti‐PD1 efficacy. Sci Transl Med. 2014;6(237):237ra67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227. Chen JY, Lai YS, Chu PY, Chan SH, Wang LH, Hung WC. Cancer‐derived VEGF‐C increases chemokine production in lymphatic endothelial cells to promote CXCR2‐dependent cancer invasion and MDSC recruitment. Cancers (Basel). 2019;11(8):1120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228. Wu A, Maxwell R, Xia Y, et al. Combination anti‐CXCR4 and anti‐PD‐1 immunotherapy provides survival benefit in glioblastoma through immune cell modulation of tumor microenvironment. J Neurooncol. 2019;143(2):241‐249. [DOI] [PubMed] [Google Scholar]
- 229. Bockorny B, Semenisty V, Macarulla T, et al. BL‐8040, a CXCR4 antagonist, in combination with pembrolizumab and chemotherapy for pancreatic cancer: the COMBAT trial. Nat Med. 2020;26(6):878‐885. [DOI] [PubMed] [Google Scholar]
- 230. Zeng Y, Li B, Liang Y, et al. Dual blockade of CXCL12‐CXCR4 and PD‐1‐PD‐L1 pathways prolongs survival of ovarian tumor‐bearing mice by prevention of immunosuppression in the tumor microenvironment. Faseb j. 2019;33(5):6596‐6608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231. Rajkumar SV, Hayman SR, Lacy MQ, et al. Combination therapy with lenalidomide plus dexamethasone (Rev/Dex) for newly diagnosed myeloma. Blood. 2005;106(13):4050‐4053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232. Richardson PG, Blood E, Mitsiades CS, et al. A randomized phase 2 study of lenalidomide therapy for patients with relapsed or relapsed and refractory multiple myeloma. Blood. 2006;108(10):3458‐3464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233. Sakamaki I, Kwak LW, Cha SC, et al. Lenalidomide enhances the protective effect of a therapeutic vaccine and reverses immune suppression in mice bearing established lymphomas. Leukemia. 2014;28(2):329‐337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234. Romano A, Parrinello NL, La Cava P, et al. PMN‐MDSC and arginase are increased in myeloma and may contribute to resistance to therapy. Expert Rev Mol Diagn. 2018;18(7):675‐683. [DOI] [PubMed] [Google Scholar]
- 235. Wang J, McGuire TR, Britton HC, et al. Lenalidomide and cyclophosphamide immunoregulation in patients with metastatic, castration‐resistant prostate cancer. Clin Exp Metastasis. 2015;32(2):111‐124. [DOI] [PubMed] [Google Scholar]
- 236. Kuwahara‐Ota S, Shimura Y, Steinebach C, et al. Lenalidomide and pomalidomide potently interfere with induction of myeloid‐derived suppressor cells in multiple myeloma. Br J Haematol. 2020;191(5):784‐795. [DOI] [PubMed] [Google Scholar]
- 237. Busch A, Zeh D, Janzen V, et al. Treatment with lenalidomide induces immunoactivating and counter‐regulatory immunosuppressive changes in myeloma patients. Clin Exp Immunol. 2014;177(2):439‐453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238. Prendergast GC, Malachowski WJ, Mondal A, Scherle P, Muller AJ. Indoleamine 2,3‐dioxygenase and its therapeutic inhibition in cancer. Int Rev Cell Mol Biol. 2018;336:175‐203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239. Prendergast GC. Immune escape as a fundamental trait of cancer: focus on IDO. Oncogene. 2008;27(28):3889‐900. [DOI] [PubMed] [Google Scholar]
- 240. Munn DH, Zhou M, Attwood JT, et al. Prevention of allogeneic fetal rejection by tryptophan catabolism. Science. 1998;281(5380):1191‐1193. [DOI] [PubMed] [Google Scholar]
- 241. Opitz CA, Somarribas Patterson LF, Mohapatra SR, et al. The therapeutic potential of targeting tryptophan catabolism in cancer. Br J Cancer. 2020;122(1):30‐44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242. Holmgaard RB, Zamarin D, Li Y, et al. Tumor‐expressed IDO recruits and activates MDSCs in a Treg‐dependent manner. Cell Rep. 2015;13(2):412‐424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243. Yu J, Wang Y, Yan F, et al. Noncanonical NF‐κB activation mediates STAT3‐stimulated IDO upregulation in myeloid‐derived suppressor cells in breast cancer. J Immunol. 2014;193(5):2574‐2586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244. Liu X, Shin N, Koblish HK, et al. Selective inhibition of IDO1 effectively regulates mediators of antitumor immunity. Blood. 2010;115(17):3520‐3530. [DOI] [PubMed] [Google Scholar]
- 245. Komrokji RS, Wei S, Mailloux AW, et al. A phase II study to determine the safety and efficacy of the oral inhibitor of indoleamine 2,3‐dioxygenase (IDO) enzyme INCB024360 in patients with myelodysplastic syndromes. Clin Lymphoma Myeloma Leuk. 2019;19(3):157‐161. [DOI] [PubMed] [Google Scholar]
- 246. Alessi DR, James SR, Downes CP, et al. Characterization of a 3‐phosphoinositide‐dependent protein kinase which phosphorylates and activates protein kinase Balpha. Curr Biol. 1997;7(4):261‐269. [DOI] [PubMed] [Google Scholar]
- 247. Zhang Z, Richmond A. The role of PI3K inhibition in the treatment of breast cancer, alone or combined with immune checkpoint inhibitors. Front Mol Biosci. 2021;8:648663. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248. Zhang X, Shen L, Liu Q, Hou L, Huang L. Inhibiting PI3 kinase‐γ in both myeloid and plasma cells remodels the suppressive tumor microenvironment in desmoplastic tumors. J Control Release. 2019;309:173‐180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249. De Henau O, Rausch M, Winkler D, et al. Overcoming resistance to checkpoint blockade therapy by targeting PI3Kγ in myeloid cells. Nature. 2016;539(7629):443‐447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250. Liu G, Jin Z, Lu X. Differential targeting of Gr‐MDSCs, T cells and prostate cancer cells by dactolisib and dasatinib. Int J Mol Sci. 2020;21(7):2337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251. Joshi S, Singh AR, Liu KX, et al. SF2523: dual PI3K/BRD4 inhibitor blocks tumor immunosuppression and promotes adaptive immune responses in cancer. Mol Cancer Ther. 2019;18(6):1036‐1044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252. Das A, Durrant D, Salloum FN, Xi L, Kukreja RC. PDE5 inhibitors as therapeutics for heart disease, diabetes and cancer. Pharmacol Ther. 2015;147:12‐21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253. Booth L, Roberts JL, Cruickshanks N, et al. Phosphodiesterase 5 inhibitors enhance chemotherapy killing in gastrointestinal/genitourinary cancer cells. Mol Pharmacol. 2014;85(3):408‐419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254. Hamilton TK, Hu N, Kolomitro K, et al. Potential therapeutic applications of phosphodiesterase inhibition in prostate cancer. World J Urol. 2013;31(2):325‐330. [DOI] [PubMed] [Google Scholar]
- 255. Roberts JL, Booth L, Conley A, et al. PDE5 inhibitors enhance the lethality of standard of care chemotherapy in pediatric CNS tumor cells. Cancer Biol Ther. 2014;15(6):758‐767. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256. Noonan KA, Ghosh N, Rudraraju L, Bui M, Borrello I. Targeting immune suppression with PDE5 inhibition in end‐stage multiple myeloma. Cancer Immunol Res. 2014;2(8):725‐731. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257. Hassel JC, Jiang H, Bender C, et al. Tadalafil has biologic activity in human melanoma. Results of a pilot trial with Tadalafil in patients with metastatic Melanoma (TaMe). Oncoimmunology. 2017;6(9):e1326440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258. Serafini P, Meckel K, Kelso M, et al. Phosphodiesterase‐5 inhibition augments endogenous antitumor immunity by reducing myeloid‐derived suppressor cell function. J Exp Med. 2006;203(12):2691‐2702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259. Pauleau AL, Rutschman R, Lang R, Pernis A, Watowich SS, Murray PJ. Enhancer‐mediated control of macrophage‐specific arginase I expression. J Immunol. 2004;172(12):7565‐73. [DOI] [PubMed] [Google Scholar]
- 260. Califano JA, Khan Z, Noonan KA, et al. Tadalafil augments tumor specific immunity in patients with head and neck squamous cell carcinoma. Clin Cancer Res. 2015;21(1):30‐38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261. Weed DT, Vella JL, Reis IM, et al. Tadalafil reduces myeloid‐derived suppressor cells and regulatory T cells and promotes tumor immunity in patients with head and neck squamous cell carcinoma. Clin Cancer Res. 2015;21(1):39‐48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262. Kroesen M, Büll C, Gielen PR, et al. Anti‐GD2 mAb and Vorinostat synergize in the treatment of neuroblastoma. Oncoimmunology. 2016;5(6):e1164919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 263. Hashimoto A, Fukumoto T, Zhang R, Gabrilovich D. Selective targeting of different populations of myeloid‐derived suppressor cells by histone deacetylase inhibitors. Cancer Immunol Immunother. 2020;69(9):1929‐1936. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 264. Orillion A, Hashimoto A, Damayanti N, et al. Entinostat neutralizes myeloid‐derived suppressor cells and enhances the antitumor effect of PD‐1 inhibition in murine models of lung and renal cell carcinoma. Clin Cancer Res. 2017;23(17):5187‐5201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265. Christmas BJ, Rafie CI, Hopkins AC, et al. Entinostat converts immune‐resistant breast and pancreatic cancers into checkpoint‐responsive tumors by reprogramming tumor‐infiltrating MDSCs. Cancer Immunol Res. 2018;6(12):1561‐1577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266. Sidiropoulos DN, Rafie CI, Jang JK, et al. Entinostat decreases immune suppression to promote antitumor responses in a HER2+ breast tumor microenvironment. Cancer Immunol Res. 2022;10(5):656‐669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267. Lu Z, Zou J, Li S, et al. Epigenetic therapy inhibits metastases by disrupting premetastatic niches. Nature. 2020;579(7798):284‐290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268. Half E, Tang XM, Gwyn K, Sahin A, Wathen K, Sinicrope FA. Cyclooxygenase‐2 expression in human breast cancers and adjacent ductal carcinoma in situ. Cancer Res. 2002;62(6):1676‐1681. [PubMed] [Google Scholar]
- 269. Ristimäki A, Sivula A, Lundin J, et al. Prognostic significance of elevated cyclooxygenase‐2 expression in breast cancer. Cancer Res. 2002;62(3):632‐625. [PubMed] [Google Scholar]
- 270. Matsubayashi H, Infante JR, Winter J, et al. Tumor COX‐2 expression and prognosis of patients with resectable pancreatic cancer. Cancer Biol Ther. 2007;6(10):1569‐1575. [DOI] [PubMed] [Google Scholar]
- 271. Sinha P, Clements VK, Fulton AM, Ostrand‐Rosenberg S. Prostaglandin E2 promotes tumor progression by inducing myeloid‐derived suppressor cells. Cancer Res. 2007;67(9):4507‐4513. [DOI] [PubMed] [Google Scholar]
- 272. Xu Y, Zhao W, Xu J, et al. Activated hepatic stellate cells promote liver cancer by induction of myeloid‐derived suppressor cells through cyclooxygenase‐2. Oncotarget. 2016;7(8):8866‐8878. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 273. Zhang B, Ma X, Li Z, et al. Celecoxib enhances the efficacy of 15‐hydroxyprostaglandin dehydrogenase gene therapy in treating murine breast cancer. J Cancer Res Clin Oncol. 2013;139(5):797‐807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 274. Veltman JD, Lambers ME, van Nimwegen M, et al. COX‐2 inhibition improves immunotherapy and is associated with decreased numbers of myeloid‐derived suppressor cells in mesothelioma. Celecoxib influences MDSC function. BMC Cancer. 2010;10:464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 275. Zhao Q, Guo J, Wang G, Chu Y, Hu X. Suppression of immune regulatory cells with combined therapy of celecoxib and sunitinib in renal cell carcinoma. Oncotarget. 2017;8(1):1668‐1677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 276. Colligan SH, Amitrano AM, Zollo RA, et al. Inhibiting the biogenesis of myeloid‐derived suppressor cells enhances immunotherapy efficacy against mammary tumor progression. J Clin Invest. 2022;132(23):e158661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 277. Sykes DB, Kfoury YS, Mercier FE, et al. Inhibition of dihydroorotate dehydrogenase overcomes differentiation blockade in acute myeloid leukemia. Cell. 2016;167(1):171‐186.e15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 278. Du B, Han H, Wang Z, et al. targeted drug delivery to hepatocarcinoma in vivo by phage‐displayed specific binding peptide. Mol Cancer Res. 2010;8(2):135‐144. [DOI] [PubMed] [Google Scholar]
- 279. Pasqualini R, Koivunen E, Kain R, et al. Aminopeptidase N is a receptor for tumor‐homing peptides and a target for inhibiting angiogenesis. Cancer Res. 2000;60(3):722‐727. [PMC free article] [PubMed] [Google Scholar]
- 280. Zheng YB, Shang BY, Li Y, Zhen YS. An NGR‐integrated and enediyne‐energized apoprotein shows CD13‐targeting antitumor activity. Biomed Pharmacother. 2013;67(2):164‐171. [DOI] [PubMed] [Google Scholar]
- 281. Garde SV, Forté AJ, Ge M, et al. Binding and internalization of NGR‐peptide‐targeted liposomal doxorubicin (TVT‐DOX) in CD13‐expressing cells and its antitumor effects. Anticancer Drugs. 2007;18(10):1189‐1200. [DOI] [PubMed] [Google Scholar]
- 282. Di Matteo P, Hackl C, Jedeszko C, et al. NGR‐TNF, a novel vascular‐targeting agent, does not induce cytokine recruitment of proangiogenic bone marrow‐derived cells. Br J Cancer. 2013;109(2):360‐369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 283. Xia C, Li M, Ran G, et al. Redox‐responsive nanoassembly restrained myeloid‐derived suppressor cells recruitment through autophagy‐involved lactate dehydrogenase A silencing for enhanced cancer immunochemotherapy. J Control Release. 2021;335:557‐574. [DOI] [PubMed] [Google Scholar]
- 284. Mirando AC, Patil A, Rafie CI, et al. Regulation of the tumor immune microenvironment and vascular normalization in TNBC murine models by a novel peptide. Oncoimmunology. 2020;9(1):1760685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 285. Casella I, Feccia T, Chelucci C, et al. Autocrine‐paracrine VEGF loops potentiate the maturation of megakaryocytic precursors through Flt1 receptor. Blood. 2003;101(4):1316‐1323. [DOI] [PubMed] [Google Scholar]
- 286. D'Alterio C, Buoncervello M, Ieranò C, et al. Targeting CXCR4 potentiates anti‐PD‐1 efficacy modifying the tumor microenvironment and inhibiting neoplastic PD‐1. J Exp Clin Cancer Res. 2019;38(1):432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 287. Tobin RP, Jordan KR, Robinson WA, et al. Targeting myeloid‐derived suppressor cells using all‐trans retinoic acid in melanoma patients treated with Ipilimumab. Int Immunopharmacol. 2018;63:282‐291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 288. Knudson KM, Hicks KC, Ozawa Y, Schlom J, Gameiro SR. Functional and mechanistic advantage of the use of a bifunctional anti‐PD‐L1/IL‐15 superagonist. J Immunother Cancer. 2020;8(1):e000493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 289. Sumida K, Wakita D, Narita Y, et al. Anti‐IL‐6 receptor mAb eliminates myeloid‐derived suppressor cells and inhibits tumor growth by enhancing T‐cell responses. Eur J Immunol. 2012;42(8):2060‐2072. [DOI] [PubMed] [Google Scholar]
- 290. Haas MS, Kagey MH, Heath H, Schuerpf F, Rottman JB, Newman W. mDKN‐01, a novel anti‐DKK1 mAb, enhances innate immune responses in the tumor microenvironment. Mol Cancer Res. 2021;19(4):717‐725. [DOI] [PubMed] [Google Scholar]
- 291. Ishfaq M, Pham T, Beaman C, Tamayo P, Yu AL, Joshi S. BTK inhibition reverses MDSC‐mediated immunosuppression and enhances response to anti‐PDL1 therapy in neuroblastoma. Cancers (Basel). 2021;13(4):817. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 292. Ferrer G, Jung B, Chiu PY, et al. Myeloid‐derived suppressor cell subtypes differentially influence T‐cell function, T‐helper subset differentiation, and clinical course in CLL. Leukemia. 2021;35(11):3163‐3175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 293. Xin H, Zhang C, Herrmann A, Du Y, Figlin R, Yu H. Sunitinib inhibition of Stat3 induces renal cell carcinoma tumor cell apoptosis and reduces immunosuppressive cells. Cancer Res. 2009;69(6):2506‐2513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 294. Ko JS, Zea AH, Rini BI, et al. Sunitinib mediates reversal of myeloid‐derived suppressor cell accumulation in renal cell carcinoma patients. Clin Cancer Res. 2009;15(6):2148‐2157. [DOI] [PubMed] [Google Scholar]
- 295. Chen HM, Ma G, Gildener‐Leapman N, et al. Myeloid‐Derived Suppressor Cells as an Immune Parameter in Patients with Concurrent Sunitinib and Stereotactic Body Radiotherapy. Clin Cancer Res. 2015;21(18):4073‐4085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 296. Guislain A, Gadiot J, Kaiser A, et al. Sunitinib pretreatment improves tumor‐infiltrating lymphocyte expansion by reduction in intratumoral content of myeloid‐derived suppressor cells in human renal cell carcinoma. Cancer Immunol Immunother. 2015;64(10):1241‐1250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 297. Zahoor H, Mir MC, Barata PC, et al. Phase II trial of continuous treatment with sunitinib in patients with high‐risk (BCG‐refractory) non‐muscle invasive bladder cancer. Invest New Drugs. 2019;37(6):1231‐1238. [DOI] [PubMed] [Google Scholar]
- 298. Du Four S, Maenhout SK, De Pierre K, et al. Axitinib increases the infiltration of immune cells and reduces the suppressive capacity of monocytic MDSCs in an intracranial mouse melanoma model. Oncoimmunology. 2015;4(4):e998107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 299. Lu X, Horner JW, Paul E, et al. Effective combinatorial immunotherapy for castration‐resistant prostate cancer. Nature. 2017;543(7647):728‐732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 300. Viswanathan VS, Ryan MJ, Dhruv HD, et al. Dependency of a therapy‐resistant state of cancer cells on a lipid peroxidase pathway. Nature. 2017;547(7664):453‐457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 301. Hangauer MJ, Viswanathan VS, Ryan MJ, et al. Drug‐tolerant persister cancer cells are vulnerable to GPX4 inhibition. Nature. 2017;551(7679):247‐250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 302. Chen X, Kang R, Kroemer G, Tang D. Broadening horizons: the role of ferroptosis in cancer. Nat Rev Clin Oncol. 2021;18(5):280‐296. [DOI] [PubMed] [Google Scholar]
- 303. Song X, Xie Y, Kang R, et al. FANCD2 protects against bone marrow injury from ferroptosis. Biochem Biophys Res Commun. 2016;480(3):443‐449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 304. Wei J, Zhang M, Zhou J. Myeloid‐derived suppressor cells in major depression patients suppress T‐cell responses through the production of reactive oxygen species. Psychiatry Res. 2015;228(3):695‐701. [DOI] [PubMed] [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.
