Skip to main content

Targeting the myeloid microenvironment in neuroblastoma

Abstract

Myeloid cells (granulocytes and monocytes/macrophages) play an important role in neuroblastoma. By inducing a complex immunosuppressive network, myeloid cells pose a challenge for the adaptive immune system to eliminate tumor cells, especially in high-risk neuroblastoma. This review first summarizes the pro- and anti-tumorigenic functions of myeloid cells, including granulocytes, monocytes, macrophages, and myeloid-derived suppressor cells (MDSC) during the development and progression of neuroblastoma. Secondly, we discuss how myeloid cells are engaged in the current treatment regimen and explore novel strategies to target these cells in neuroblastoma. These strategies include: (1) engaging myeloid cells as effector cells, (2) ablating myeloid cells or blocking the recruitment of myeloid cells to the tumor microenvironment and (3) reprogramming myeloid cells. Here we describe that despite their immunosuppressive traits, tumor-associated myeloid cells can still be engaged as effector cells, which is clear in anti-GD2 immunotherapy. However, their full potential is not yet reached, and myeloid cell engagement can be enhanced, for example by targeting the CD47/SIRPα axis. Though depletion of myeloid cells or blocking myeloid cell infiltration has been proven effective, this strategy also depletes possible effector cells for immunotherapy from the tumor microenvironment. Therefore, reprogramming of suppressive myeloid cells might be the optimal strategy, which reverses immunosuppressive traits, preserves myeloid cells as effectors of immunotherapy, and subsequently reactivates tumor-infiltrating T cells.

Introduction

Neuroblastoma is a pediatric tumor originating from sympathoadrenal lineage dysregulation [1] which exhibits high heterogeneity in disease severity. While low- and intermediate-risk neuroblastoma patients achieve a 80–95% 5-year overall survival rate, high-risk cases only reach 45% [2]. Approximately half of the diagnosed neuroblastoma cases are classified as high-risk, with 20–30% featuring MYCN amplification [3]. MYCN, a proto-oncogenic transcription factor, fosters tumor cell proliferation, angiogenesis, and metastasis, concurrently suppressing immune activation [4]. In low-risk cases, no treatment or solely surgery is often curative [5, 6], while high-risk patients undergo induction chemotherapy, surgery, high-dose chemotherapy with autologous stem cell transplantation (ASCT), and radiation therapy [7]. Since 2015, anti-disialoganglioside (GD2) antibody immunotherapy and 13-cis retinoic acid (isotretinoin) is applied as consolidation therapy. Initially anti-GD2 was combined with IL-2 and Granulocyte/Macrophage-Colony Stimulating Factor (GM-CSF) (USA) or IL-2 only (Europe). Since the administration of IL-2 has been proven to be of no additional benefit this has been omitted [8].

Neuroblastoma has a low mutational burden [9] as well as low human leukocyte antigen (HLA) type I expression [10], limiting tumor recognition by the T cells. Due to the scarcity of neo-antigens, the development of immunotherapeutics, including anti-tumor antibodies, T cell vaccines and chimeric antigen receptor (CAR) T cells hampered initially. However, success was achieved by targeting the highly expressed GD2 [11] using antibody therapy, thereby activating NK cells and myeloid cells. Anti-GD2 antibody therapy significantly improved survival for high-risk neuroblastoma patients [12, 13]. Although GD2-targeting CAR T cells have been developed, they failed to enhance survival initially, likely due to T cell exhaustion by the immunosuppressive tumor microenvironment (TME) (reviewed in [13]). However, recently, a phase I/II trial showed that the use of GD2-targeting CAR T cells (GD2-CART01) was feasible and safe in treating high-risk neuroblastoma, resulting in 3-year overall survival and event-free survival of 60% and 36%, respectively [14].

Neuroblastomas, like many solid tumors, harbor a complex immunosuppressive microenvironment hindering immune-mediated tumor clearance. Tumor-infiltrating lymphocytes are often inactivated or exhausted due to immunosuppressive factors, including cytokines (IL-6, IL-10, TGF-β, galectin-1) secreted by tumor, stromal, and myeloid immune cells [15,16,17,18,19]. Furthermore, MYCN overexpression in neuroblastoma cells diminishes NKG2D ligands, impeding NK cell activation [20]. Gangliosides like GD2 (cell-bound or soluble) suppress immunity by binding to myeloid checkpoint Siglec-7 [21], and CD8+ T cell cytotoxicity is curtailed via intracellular granule interference [22]. Lymphocyte activation is further hampered by recruitment and induction of immunosuppressive cells, such as regulatory T cells (Tregs), tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs) [23].

While often implicated in immunosuppression, myeloid cell subsets also possess potent anti-tumorigenic properties. This review comprehensively outlines the dualistic roles of myeloid cells in neuroblastoma. The first part summarizes their pro- and anti-tumorigenic functions during the development and progression of neuroblastoma. The second part discusses the involvement of myeloid cells in current treatment regimen and explores novel strategies for their targeting, including: (1) engaging myeloid cells as effectors, (2) ablating myeloid cells, (3) blocking recruitment to the TME and (4) reprogramming of myeloid cells.

Myeloid subpopulations in the neuroblastoma microenvironment

While the field of tumor immunology initially focused on dissecting the role of lymphocytes, there is a growing awareness of the significance of myeloid cells. Myeloid cells comprise monocytes, macrophages, granulocytes (mainly neutrophils and eosinophils), MDSC and certain DC subsets. Though some DCs are of myeloid origin, they are beyond the scope of this review, as they have been extensively covered elsewhere (reviewed in [24]). MDSC can be categorized into three subsets: polymorphonuclear MDSC (PMN-MDSC, CD33+CD14CD15+LOX1+), monocytic MDSC (M-MDSC, CD33+CD14+CD15HLA-DR−/low) and early MDSCs (E-MDSC, CD11b+CD33+CD14CD15) [25,26,27]. Identifying specific MDSC subsets can be challenging due to overlapping lineage markers with other immune cells such as neutrophils and monocytes (reviewed in [28]). However, the main defining characteristic of MDSCs is their immunosuppressive function. Recent markers such as LOX-1 and CD84 expression on PMN-MDSCs [26, 29] or gradient centrifugation [30] can aid in distinguishing PMN-MDSCs from neutrophils, as PMN-MDSCs reside in the low-density (peripheral blood mononuclear cells; PBMC) fraction, while neutrophils are high-density cells. Furthermore, M-MDSCs can be differentiated from monocytes by lower or absent HLA class II expression and increased C-X-C motif chemokine receptor 1 (CXCR1) expression [25, 31]. The challenges in accurately identifying MDSCs often lead to inconsistencies in nomenclature across studies. In this review, we adhere to the nomenclature used in the referenced literature, considering the mentioned immune cell subsets as they are described. Now, we will first discuss the involvement of each of these myeloid immune subsets in neuroblastoma, starting with neutrophils (Fig. 1).

Fig. 1
figure 1

Overview of the interactions of myeloid cell subsets in the tumor microenvironment of neuroblastoma. A neuroblastoma tumor is shown in the center, surrounded by various myeloid immune cells: neutrophils, MDSC, monocytes, TAM, and eosinophils. For each of these cell types, pertinent interactions within the TME are depicted. For neutrophils, a high NLR is associated with unfavorable clinical outcomes. Reported tumor-neutrophil interactions involve the induction of neutrophil adhesion to neuroblastoma cell lines through IFNγ and IL-1, resulting in increased ICAM-1 expression on neutrophils. MDSCs are linked to T cell activation inhibition, notably through P2X7 receptor activation leading to increased ATP levels within the TME. Additionally, neuroblastoma-conditioned medium induces MDSCs through M-CSF and Arg-1/2. While most mouse MDSCs exhibit an M-MDSC phenotype, human MDSCs present a PMN-MDSC signature. For monocytes, LMR was not correlated with clinical outcomes in neuroblastoma patients. In neuroblastoma, TAMs undergo polarization from M1 to M2 as the disease progresses. Additionally, TAMs contribute to the preparation of the metastatic niche by taking up tumor-secreted EVs, resulting in the upregulation of immunosuppressive cytokines and genes associated with tumor cell extravasation, and via the CXCL2/CXCR2 axis. In MYCN-amplified neuroblastoma, TAMs exhibit elevated expression of macrophage-related immune checkpoints CD47 and Siglec7, in contrast to MYCN-nonamplified tumors. MYCN-nonamplified neuroblastoma is characterized by elevated CCL2 secretion by TAMs, resulting in the recruitment of TAMs, myeloid cells, and plasmacytoid DC. Furthermore, TAM-induced hypoxia leads to the inhibition of NK cells through HIF-2α production. TAMs also collaborate with CAFs and MSCs to promote tumor progression. Although our understanding of eosinophils, mast cells, and basophils is limited, IGF-2 secreted by eosinophils is suggested to play a role in neuroblastoma tumor growth. Abbreviations: Arg1/2 = arginase-1/2, ATP = adenosine triphosphate, CAF = cancer-associated fibroblast, CAF = cancer-associated fibroblasts, CCL2 = C–C motif chemokine ligand 2, CXCL2 = C-X-C motif chemokine ligand 2, CXCR2 = C-X-C motif chemokine receptor 2, DC = dendritic cell, Eo = eosinophil, EV = extracellular vesicles, EVs = extracellular vesicles, HIF-2a = hypoxia inducible factor 2α, ICAM-1 = intercellular adhesion molecule-1, IFNγ = interferon gamma, IGF-2 = insulin-like growth factor 2, LMR = lymphocyte-to-monocyte ratio, M-CSF = macrophage-colony stimulating factor, M-MDSC = monocytic myeloid-derived suppressor cells, MDSC = myeloid-derived suppressor cell, Mono = monocyte, MSC = mesenchymal stromal cell, MSC = mesenchymal stromal cells, NB = neuroblastoma, NB-NA = MYCN non-amplified neuroblastoma, Neutro = neutrophil, NK = natural killer, NLR = neutrophil-to-lymphocyte ratio, PMN-MDSC = polymorphonuclear myeloid-derived suppressor cells, TAM = tumor-associated macrophage, TME = tumor microenvironment

Neutrophils

Neutrophils are short-lived phagocytes (though their exact lifespan is heavily under debate) and are the most abundant leukocyte population in the human body, primarily residing in the bone marrow [32, 33]. Approximately 1011 neutrophils are released from the bone marrow and enter circulation daily, with an additional release during inflammatory conditions, upon which they migrate towards the sites of inflammation [34].

The prognostic significance of tumor-infiltrating neutrophils or the neutrophil-to-lymphocyte ratio (NLR) in various cancer types typically indicates unfavorable outcomes [35]. However, in the case of neuroblastoma, conflicting and inconclusive findings have been reported regarding the correlation between neutrophils and disease progression. Initial studies by Morandi et al. demonstrated increased neutrophil counts in peripheral blood samples from patients with localized neuroblastoma compared to patients with metastasized neuroblastoma, hence neutrophils were associated with improved overall survival (OS) [36]. Similarly, Zeng et al. found a positive correlation between higher numbers of tumor-infiltrating macrophages and neutrophils and increased OS [37]. Conversely, other studies did not observe significant associations between neutrophils and survival [38], or even found the opposite [39,40,41]. Zhang et al. reported that a higher NLR was associated with reduced OS [39]. Erbe et al. did not find a significant correlation between NLR and survival, but they did note that a higher neutrophils count was associated with shortened event-free survival (EFS) [41]. Lee et al. observed that elevated neutrophil counts were associated with a higher cumulative incidence of disease progression, although not significantly impacting EFS or OS [40]. However, in a subgroup analysis of high-risk neuroblastoma patients, the correlation was stronger, with a high neutrophil count significantly associated with reduced EFS. Although these results appear conflicting, there are indications that high neutrophils counts in blood may indicate a positive prognosis in localized neuroblastoma (low-risk), whereas high neutrophil counts in high-risk neuroblastoma are associated with a negative prognosis, as suggested by the studies of Morandi and Lee. Nevertheless, further investigations, specifically differentiating between low- and high-risk patients, are required to confirm these hypotheses in the future.

The interaction between neutrophils and neuroblastoma cells was studied already decades ago, albeit mostly in vitro. Two studies showed that cytokines like tumor necrosis factor alpha (TNFα), Interleukin (IL)-1 and interferon gamma (IFNγ) induce neutrophil adhesion to SK-N-SH and LAN-1 neuroblastoma cell lines by upregulating intercellular adhesion molecule-1 (ICAM-1) expression [42, 43]. Blocking lymphocyte function-associated antigen 1 (LFA-1, formed by CD11a and CD18), the ligand of ICAM-1, prevented cytokine-induced neutrophil adhesion to neuroblastoma cells. However, neutrophil binding to SK-N-MC cells was very high and independent of stimulatory cytokines or LFA-1, indicating multiple adhesion mechanisms exist for different neuroblastoma cell lines. Interestingly, co-culture experiments demonstrated that endothelial cells down-regulated adhesion receptor CD33 upon interaction with neuroblastoma cells, thus impairing neutrophil adhesion and extravasation [44]. Nevertheless, Fultang et al. indicated the presence of CD15+ neutrophils in neuroblastoma tumors, particularly in high-risk disease, suggesting their ability to extravasate and localize around the vasculature [45]. Direct co-culture of neuroblastoma cell lines with neutrophils exhibited both pro-tumorigenic (SMS-KCN and SMS-LHN) and anti-tumorigenic (LAN-1) effects, in a cell–cell contact independent manner [46]. These studies highlight both pro-tumorigenic and anti-tumorigenic effects of neutrophils and touch upon the intricate interplay between neutrophils and the neuroblastoma cells in the TME.

More recently, Martínez-Sanz and colleagues identified upregulated neutrophil-related mRNA transcripts (including CGR3B, FPR1, S100A8/9, and SIGLEC9) in both early and late-stage neuroblastoma, compared to healthy adrenal gland tissue [47]. However, the exact interactions between neutrophils and tumor cells in the TME remain largely unknown. After the initial interest in neutrophils, immunotherapy research focused more on harnessing the adaptive immune system and engaging NK cells through antibody therapy in the early 2000s, which may explain the scarcity of data on neutrophils. Furthermore, a nomenclature change has led to (immunosuppressive) neutrophils being described more frequently as PMN-MDSC in cancer. Consequently, we will now delve into the role of these cells in neuroblastoma.

Myeloid-derived suppressor cells

MDSCs represent an immature immunosuppressive subset of myeloid cells that undergo expansion during pathological conditions such as inflammation and cancer [48, 49]. Their presence has been associated with poor clinical outcomes in various cancer types [50]. Under the influence of continuous cytokine production within the TME, undifferentiated myeloid cells from the bone marrow enter circulation and infiltrate the tumor, where they often fail to mature (reviewed in [50,51,52]). MDSCs exert immunosuppressive effects by suppressing T and NK cell activation and cytotoxicity, while promoting Treg and M2 macrophage polarization. They employ various mechanisms to induce immune suppression, including the production of reactive oxygen species (ROS), nitric oxide (NO), anti-inflammatory cytokines, and the depletion of L-arginine via arginase (Arg)-1 [48, 53, 54]. In general, M-MDSCs exhibit higher immunosuppressive activity than PMN-MDSCs, suppressing both antigen-specific and non-specific T cell responses [48].

In neuroblastoma-bearing mice, both PMN-MDSC and M-MDSC induce T cell suppression [55,56,57,58,59]. Notably, TH-MYCN mice, which exhibit spontaneous neuroblastoma development due to MYCN overexpression, showed a higher PMN-MDSC abundance compared to M-MDSC [60]. Conversely, in a commonly used neuroblastoma mouse model involving A/J mice injected with NXS2 neuroblastoma cells intravenously, M-MDSC displayed higher levels of Arg-1 and iNOS, and produced higher levels of TGF-β1 and ROS compared to PMN-MDSC [56]. This hints at M-MDSCs' superior suppressive potency compared to PMN-MDSC, a trend observed in other cancer types as well [61, 62]. Co-injection of M-MDSCs with NXS2 neuroblastoma cells promoted tumor growth more than PMN-MDSC co-injection, underscoring their distinct roles. MDSCs' immunosuppressive actions involved P2X7 receptor activation, which elevates extracellular adenosine triphosphate (ATP) levels within the TME. P2X7R agonist BzATP induced M-MDSCs to secrete chemoattractant C–C motif chemokine ligand (CCL)-2, and upregulated Arg-1, ROS, and TGF-β1 in MDSC cell lines MSC-1 and MSC-2.

Furthermore, neuroblastoma-conditioned medium has been shown to induce MDSCs. Cultivating hematopoietic progenitors or monocytes in such media generated immunosuppressive myeloid cells [57, 59, 63]. Intriguingly, Arg-2-expressing neuroblastoma cells depleted local and circulating arginine levels, similar to the mechanism employed by MDSCs [63]. Arg-1 in neuroblastoma hindered myeloid activation and suppressed CD34+ bone marrow progenitor cell proliferation, suggesting that arginine deprivation contributes to MDSC induction in neuroblastoma [64]. Additionally, the macrophage-colony stimulating factor (M-CSF)/colony stimulating factor 1 receptor (CSF-1R) axis was suggested to drive myeloid differentiation into M-MDSCs in neuroblastoma [57]. Elevated M-CSF, CSF-1R, CD68, and CD14 correlated with poor patient outcomes. Intriguingly, primary human monocytes polarized into suppressive CD14+CSF-1R+ M-MDSC upon co-culture with neuroblastoma cells, mediated by tumor-originating M-CSF and attenuated through CSF-1R blockade.

Limited neuroblastoma patient data exists on MDSC subsets in circulation. Santilli et al. found elevated PMN-MDSCs in the peripheral blood of patients with neuroblastoma vs. controls [65]. Furthermore, high-risk neuroblastoma exhibited higher levels of CD33+CD11b+HLA-DR MDSCs (M-MDSC) than low-risk neuroblastoma [60]. Unexpectedly, responsive high-risk patients had more circulating HLA-DR MDSCs compared to patients who were refractory to therapy [66]. Recent single-cell transcriptomics unveiled exclusive tumor-infiltrated immature myeloid/neutrophil cells in bone marrow metastasis of neuroblastoma patients, suggesting PMN-MDSC involvement [64]. PMN-MDSC signatures were observed in primary neuroblastoma tumors based on single-cell transcriptomic analysis [58], with higher proportions of MDSCs detected in MYCN-amplified tumors compared to MYCN-nonamplified tumors. At relapse, tumors had increased MDSC proportions compared to diagnosis [60].

In summary, in vivo studies implicate a more important role for M-MDSC in neuroblastoma tumor progression compared to PMN-MDSC due to their prominent suppressive function. However, it remains uncertain if this translates to the human context. Furthermore, it is important to note that MDSC spatial distribution, abundance, and interactions with other immune cell subsets within the TME remain undisclosed.

Monocytes & macrophages

Monocytes and macrophages play a significant role within the myeloid compartment of the TME and possess phagocytic abilities. Unlike neutrophils, circulating monocyte levels or leukocyte-to-monocyte ratio (LMR) are not linked to overall survival [38, 41]. Tumor-derived chemokines and cytokines can recruit monocytes to the TME, where they can differentiate into monocyte-derived macrophages [67]. Additionally, tissue-resident macrophages can also be found in the TME, originating from yolk-sac precursors and exhibiting self-renewal capabilities [68]. Furthermore, recruited M-MDSCs can differentiate into macrophages at the tumor site [69]. Macrophages are traditionally categorized into M1 (pro-inflammatory) and M2 (anti-inflammatory) states [70], yet they exhibit notable plasticity and heterogeneity along this spectrum.

Macrophages in human neuroblastoma have a predominant M2-like phenotype, with some M1 presence [45, 58, 71,72,73,74]. In the TH-MYCN mouse model, TAMs transitioned from an M1 phenotype (MHCIIhigh/CD206low) to an M2 phenotype (MHCIIlow/CD206high) during tumor progression [55]. Additionally, the number of infiltrating immune cells and the proportion of TAMs (F4/80+/CD45+) among total infiltrating cells increased in advanced TH-MYCN tumors. In vitro, neuroblastoma-exposed monocytes/macrophages upregulated M2 markers (CD163, CD204, IL-10) [73, 75]. Conversely, Fultang et al. observed that neuroblastoma cells polarized macrophages toward an M1 phenotype (CD68+CD163) [45]. Consistent with these findings, low numbers of M2-polarized macrophages were observed to be correlated with unfavorable clinical outcome in neuroblastoma [37, 76]. Importantly, TAM emerge as the predominant PD-L1-expressing cells within the TMA, which was associated with improved clinical outcome in high-risk patients [77]. Finally, single-cell RNA sequencing revealed an augmented presence of an inflammatory monocyte cell state (IL1BhighS100Ahigh) in neuroblastoma compared to normal tissue [78].

Macrophages drive neuroblastoma dissemination by creating premetastatic niches, facilitating tumor cell colonization [79]. Studies utilizing SK-N-BE-derived primary tumors revealed the release of extracellular vesicles that are engulfed by macrophages at distant sites such as the liver and bone marrow. Consequently, this led to upregulation of immunosuppressive cytokines (IL-10), as well as genes involved in the recruitment of MDSC and tumor cell extravasation, rendering the premetastatic niche more attractive for tumor cell colonization. In accordance, high numbers of M2 macrophages in the primary neuroblastoma tumor correlated with the presence of bone marrow metastasis and poor clinical outcome [71, 73, 80]. Moreover, macrophage-derived CXCL2 promoted neuroblastoma invasiveness, but CXCL2/CXCR2 mechanism in this context remains unclear [73]. These are the only studies investigating the role of myeloid cells in neuroblastoma metastasis, though this has been described more extensively in other cancers, indicating there is a knowledge gap on this topic. However, recently it was described that macrophage migration inhibitory factor (MIF) and midkine (MDK) are expressed in neuroblastoma [81, 82], factors that are known to be involved in metastasis in other cancers [83, 84]. Unraveling the involvement of MIF and MDK in neuroblastoma metastasis could provide important information in the future.

MYCN-amplified and non-amplified neuroblastomas exhibit varying macrophage infiltration, reflecting distinct immune profiles due to specific genetic alterations. Chromosome 11q deletion or MYCN amplification in neuroblastomas exhibit higher numbers of M2-polarized (CD163+) macrophages and an activated Th2-lymphocytes/M2-macrophage axis compared to neuroblastoma lacking these mutations [72, 73, 85]. Interestingly, MYCN amplification is generally associated with lower immune infiltrate than non-amplified cases [86]. Theruvath et al. confirmed this by re-analyzing public databases, revealing higher Siglec 7 and CD47 expression on MYCN-amplified neuroblastoma macrophages [21]. In metastatic non-amplified neuroblastomas, TAM infiltration was higher compared to locoregional tumors, with a predominant M2 phenotype [71]. Furthermore, metastatic non-amplified tumors in patients ≥ 18 months showed elevated inflammation-related gene expression compared to those < 18 months. Consistent with these findings, a study involving 129 NA-NB patients reported upregulation expression of CD14, IL-6 and TGF-β in patients with poor clinical outcome [87].

The NB-tag mouse model [88], resembling human non-amplified neuroblastoma, recruits TAMs, myeloid cells, and plasmacytoid DCs through CCL2 signaling [88, 89]. Macrophage-secreted CCL2 is prominent in non-amplified tumors, and inversely correlated with MYCN amplification [87, 90]. The recruited DCs subsequently attracted CD4+ and CD8+ T cells via CCL22 and CCL19 [89]. In vitro and in vivo studies have demonstrated that CCL2-attracted TAMs in neuroblastoma enhance IL-6 expression, promote tumor growth and inhibit apoptosis [87, 88]. Furthermore, TAMs in non-amplified tumors activate STAT3 in neuroblastoma cells, upregulating c-Myc that reciprocally induces CCL2 secretion, forming a positive feedback loop [88]. Consistent with these findings, a recent in vivo study reported that depletion of macrophages expressing the CCL2 receptor (CCR2) prevented tumor formation in an ALK-mutated TH-MYCN mouse model [91].

Hypoxia, common in cancer due to insufficient oxygen supply (reviewed in [92]), upregulates membrane-bound TNFα on neuroblastoma cells, triggering TAMs to produce CCL20 [93]. This TAM-derived CCL20 recruits natural killer T (NKT) which eliminate pro-tumorigenic CD1d+ TAM at the tumor site [87]. Despite this anti-tumorigenic potential, hypoxia impairs NKT cell function against CD1d+ TAMs [93]. Additionally, hypoxia inducible factor 2α (HIF-2α)-producing TAMs were detected alongside neuroblastoma neural crest-like cells in the perivascular niche, where high levels of vascular endothelial growth factor (VEGF) were observed [94]. This suggests a cooperative interaction between neuroblastoma crest-like cells and macrophages to promote angiogenesis via HIF‐2α-mediated VEGF expression.

In neuroblastoma, TAMs collaborate with cancer-associated fibroblasts (CAFs) and mesenchymal stromal cells (MSC) to support tumor progression, especially in relapse patients. TAMs induce CAF proliferation [73], and MSCs/CAFs shield monocytes from apoptosis through in an IL-6-dependent manner [95]. The interactions of monocytes and MSC resulted in the significant upregulation of several pro-tumorigenic cytokines and chemokines, including TGF-β1, MCP-1, IL-6, IL-8, and IL-4. Furthermore, the abundance of MSC and CAFs was correlated with tumor progression, including high histological malignancy and low infiltration of T and NK cells [73, 96].

In summary, despite some conflicting findings, most studies suggest a notable role for M2-polarized TAMs in neuroblastoma progression, driving c-Myc expression, CAF recruitment, and angiogenesis.

Eosinophils

Currently, limited knowledge exists regarding the roles of granulocytes other than neutrophils in neuroblastoma, including eosinophils, basophils, and mast cells. To our knowledge, no literature is available discussing the role of basophils and mast cells in neuroblastoma. Eosinophils, usually associated with allergic reactions and immune responses against parasites [97], may have a minor role in neuroblastoma. In cancer, eosinophils generally display anti-tumorigenic properties, although some pro-tumorigenic effects have also been described (reviewed in [98]).

Eosinophils were first identified in neuroblastoma in the 1990s when a study of 21 tumors revealed their presence, along with insulin-like growth factor 2 (IGF-2) expression in these cells [99]. The authors proposed a mechanism whereby IGF-2, expressed both autocrinally and paracrinally (including by eosinophils), promotes tumor growth, but this mechanism was never confirmed. A subsequent study reported a positive link between low eosinophil count and patient survival [100]. Though not yet reported in neuroblastoma, tumor-infiltrating eosinophils are common in various cancer types [98], suggesting they are possibly present in the neuroblastoma TME as well.

The role of epigenetics in myeloid cells and therapy resistance

Myeloid cells exhibit a high degree of plasticity, characterized by their ability to deviate from typical differentiation pathways and generate diverse tolerogenic myeloid cell states within the TME. A substantial component of this plasticity can be attributed to epigenetic mechanisms, with myeloid cells displaying marked responsiveness to histone modifications, alterations in DNA methylation patterns, chromatin remodeling, and the regulatory impact of non-coding RNA molecules [101,102,103]. Importantly, abnormal epigenetic modifications have been reported in the development and progression of cancer and therapy resistance [104, 105], and are closely related to altered glucose, lipid, and amino acid metabolism in the TME as thoroughly reviewed elsewhere [103, 105, 106]. For instance, histone citrullination, catalyzed by peptide arginine deiminase (PAD), is associated with the creation of neutrophil extracellular traps (NETs) in cancer, contributing to both innate immunity and tumor progression [107]. In addition, Hypoxia and M1 macrophages drive histone lactylation through lactate accumulation in the TME. This upregulated methyltransferase-like 3 (METTL3) in infiltrating myeloid cells in colon cancer via H3K18 lactylation, crucial for the transcription of immunosuppressive genes [108]. Furthermore, reduction of methylation at the Arg1 and STAT3 promoter regions has been reported in ex vivo induced MDSCs, resulting in the release of STAT3-associated cytokines IL-6 and IL-10, which subsequently activated STAT3 phosphorylation. The phosphorylated STAT3, in turn, bound to the Arg1 and S100A8 promoter regions, leading to upregulation of Arg1 and S100A8, thereby augmenting the immunosuppressive capacity of MDSCs [109, 110]. Fetahu et al. identified key transcription factors in myeloid cells of neuroblastoma bone marrow metastases linked to open chromatin regions in genes associated with M2 polarization, tumor growth, and metastases, including IL-10, TIMP1, and EREG [111]. Finally, several studies suggest that PD-1 and PD-L1 expression in tumors is decreased due to epigenetic changes, resulting in resistance to immune checkpoint therapy. Interestingly, the DNA-demethylating agent azacytidine increased PD-L1 expression in non-small cell lung cancer patients [112, 113].

In neuroblastoma, two different epigenetic states have been defined: mesenchymal or neuro crest cell-like cells (MES) with a less-differentiated state, and adrenergic or sympathetic noradrenergic cells (ADRN) with a more differentiated state [114,115,116]. In primary neuroblastomas, the initial tumor mainly consists of the ADRN state [114, 115], but the MES state becomes more prominent during relapse and metastasis [117]. Studies showed that human neuroblastoma cells with stronger MES signature have a higher basal inflammatory state, promote T cell infiltration by secreting inflammatory cytokines, and respond to immune checkpoint therapy in an immunocompetent mouse model [118, 119]. Specifically, PRRX1 a component of the mesenchymal core regulatory circuitry, activates the transcription of MHC class I and APP genes which subsequently enhance the immunogenic state of neuroblastoma cells [119]. Furthermore, sensing of double-stranded RNA in MES state neuroblastoma cell lines resulted in secretion of proinflammatory cytokines, enrichment of inflammatory transcriptomic signatures, and increased tumor killing by T cells in vitro [118]. Interestingly, genetically switching unresponsive ADRN state neuroblastoma cell lines towards the MES state fully restored responsiveness. These studies did, however, not investigate the role of myeloid cells in relation to the epigenetic states of neuroblastoma cells.

In summary, epigenetic alterations are important for immunotherapy resistance, governing not just the expression of immune checkpoint inhibitors, but also influencing immune cell infiltration, antigen presentation, and the expression and release of cytokine profiles within the tumor. Furthermore, they drive myeloid cell reprogramming toward immunosuppressive subsets within the TME, amplifying their suppressive potential.

The role of myeloid cells in immunotherapy

As previously discussed, myeloid cells have been shown to predominantly exhibit a pro-tumorigenic role in neuroblastoma, making them attractive targets for immunotherapeutic interventions. In this section, we provide a summary of established and emerging therapeutic strategies that specifically target myeloid cells in neuroblastoma (Fig. 2).

Fig. 2
figure 2

Three main strategies of targeting myeloid cells in neuroblastoma. Myeloid cells can be 1.) engaged as effector cells of immunotherapy via ADCC. 2.) removed from the TME by depletion with antibodies (anti-CD11b, anti-CD105, anti-CD33, or anti-CSF-1R) or myoablative chemotherapy. Additionally, myeloid cells can be depleted via inhibition of cytokines via IL6R blockade, or inhibition of TGFβ or STAT3. Finally, their recruitment can be blocked with CCR2 inhibitors, or anti-CCL2 or anti- TGFβ. 3.) reprogrammed to an anti-tumor, immunostimulatory phenotype, leading to reactivation of T cells. Myeloid cell reprogramming can be achieved by inducing epigenetic modifications, inhibiting cytokines and chemokines, suppressing COX activity, stimulating CD40 ligation, and preventing arginine uptake. Abbreviations: ADCC = antigen-dependent cellular cytotoxicity, Arg-1 = arginase-1, CCL2 = C–C motif chemokine ligand 2, CCR2 = CCL2 receptor, COX = cyclooxygenase, CSF-1R = colony stimulating factor 1 receptor, FcγRs = Fc gamma receptors, IFNγ = interferon gamma, ROS = reactive oxygen species, TGFβ = transforming growth factor beta, TME = tumor microenvironment, TNFα = tumor necrosis factor alpha, VEGF = vascular endothelial growth factor

Myeloid cells as effector cells of immunotherapy

Myeloid effector cells in anti-GD2 immunotherapy

It is widely recognized that in addition to NK cells, macrophages possess the potential to serve as effector cells in current anti-GD2 antibody therapy. As early as 1990, it was demonstrated that M-CSF-differentiated macrophages could phagocytose neuroblastoma tumor cells upon stimulation with the 3F8 anti-GD2 antibody [120]. Recent studies have further emphasized the efficacy of macrophages as effector cells in antibody immunotherapy [21, 77, 121]. However, within the TME, macrophages (or TAMs) often exhibit inactivation, refractoriness, or highly immunosuppressive traits, thereby contributing to antibody therapy resistance. Interestingly, in two neuroblastoma patient cohorts, the presence of TAMs expressing PD-L1, as well as the expression of the SLAMF7 pathway and CD163 in TAMs, was found to be beneficial for high-risk patients and correlated with improved survival following immunotherapy [77]. While this may appear counterintuitive, it suggests that despite their immunosuppressive characteristics, TAMs can still be engaged as effector cells in immunotherapy.

Similarly, despite their association with tumor progression in high-risk neuroblastoma, neutrophils and monocytes (or MDSCs) can still be effectively engaged in anti-GD2 antibody therapy (Table 1) [46]. In the late 1980s and early 1990s, it was demonstrated that neutrophils could perform antigen-dependent cellular cytotoxicity (ADCC) against human neuroblastoma cells in vitro with anti-GD2 antibodies (ch14.18 or 3F8 IgG) [122,123,124,125]. Remarkably, Barker and colleagues discovered that granulocytes, displayed superior killing capacity against NMB-7 neuroblastoma cells compared to PBMC [124], which is generally the opposite in IgG antibody therapy. Furthermore, neutrophils derived from neuroblastoma patients (stage IV) retained potent ex vivo killing ability against neuroblastoma cells, matching that of neutrophils from healthy donors. This neutrophil-mediated killing is marked by the release of lytic granules [125] and dependent on CD11b/CD18 (collectively known as membrane-activated complex 1 (Mac-1) or complement receptor 3 (CR3)), as well as FcγRII and FcγRIII [126].

Table 1 Comprehensive overview of treatments in neuroblastoma engaging myeloid cells as effector cells

Subsequently, in two clinical trials investigating 3F8 anti-GD2 antibody therapy, it was demonstrated that neutrophil-mediated ADCC was indeed responsible for therapeutic efficacy in neuroblastoma patients [128, 129]. Activated neutrophils, characterized by the CD11b activation epitope CBRM1/5, were associated with prolonged progression-free survival [129]. Moreover, an FCGR2A polymorphism in patients showed a correlation with clinical outcome following anti-GD2 antibody and GM-CSF therapy [130, 144]. The FcγRIIa-R131 variant exhibited higher affinity for the murine IgG3 3F8 antibody than FcγRIIa-H131, resulting in enhanced ADCC. Consequently, patients homozygous for the FCGR2A-R131 allele demonstrated extended progression-free survival after treatment with murine 3F8 antibody and GM-CSF compared to patients who were heterozygous or homozygous for FCGR2A-H131 [130]. It is noteworthy that the affinity of these polymorphisms is reversed for human IgG1 antibodies, thus leading to an opposite correlation with dinutuximab (anti-GD2, CH14.18 antibody), which is of the human IgG1 isotype [144]. Given the high expression of FcγRIIa on neutrophils, macrophages, and monocytes and its absence on NK cells, the prognostic value of this FcγRIIa polymorphism indicates that myeloid cells constitute a major effector cell population in antibody therapy for neuroblastoma.

Although the introduction of anti-GD2 antibody therapy significantly improved survival rates, resistance still develops in many patients. In vitro studies have revealed that GD2 expression levels do not correlate with neutrophil-mediated ADCC, suggesting abundant GD2 levels on all neuroblastoma cells [127]. However, the effectiveness of neutrophil-mediated tumor cell killing was diminished in tumor cells displaying high rates of antibody internalization, thus proposing a potential mechanism for treatment resistance.

Previously, anti-GD2 immunotherapy was combined with IL-2. However, this was discontinued, since a recent randomized phase III trial of patients with high-risk neuroblastoma showed that the addition of IL-2 to anti-GD2 antibody therapy did not improve outcome and increased treatment toxicity [8]. Besides its stimulatory effects on NK-cell mediated ADCC [145], IL-2 was shown to upregulate expression of the adhesion molecule CD18 on neutrophils, but to decrease total neutrophil numbers [131, 146]. Additionally, two immune monitoring studies in neuroblastoma observed that IL-2 cycles increased eosinophil counts [131, 147]. It is generally known that eosinophils can be pre-activated by IL-2 and subsequently exert ADCC [148]. However, eosinophils are likely not involved in tumor killing, since similar levels of ADCC were found in patients with high compared to low eosinophil count and eosinophil counts were not significantly correlated with survival. Furthermore, long-term infusion of anti-GD2 in combination with IL-2 resulted in induction of Tregs, which inversely correlated with IFN-y levels and progression-free survival [135].

In the United States, anti-GD2 antibody therapy is enhanced with GM-CSF, but its accessibility and availability outside the country are restricted due to regulatory constraints. GM-CSF increases numbers of circulating neutrophils, monocytes, and eosinophils and promotes the release of myeloid cell-associated factors such as CXCL11, CCL17, CCL23, and MCP4 [131]. However, the primary mechanism of GM-CSF is augmenting neutrophil-mediated ADCC and to a lesser extent activating macrophages and monocytes. Both in vitro and in patients, the combination of GM-CSF with anti-GD2 antibodies activates neutrophils and upregulates CD11b expression [12, 123, 125, 126, 128]. Particularly in patients with refractory or minimal residual disease, the addition of GM-CSF to the treatment regimen has shown favorable responses [12, 132]. Interestingly, the route of GM-CSF administration influences the degree of neutrophil activation, with subcutaneous injection resulting in a higher percentage of activated neutrophils compared to intravenous injection [129]. However, in other cancer types, such as liver carcinoma and glioblastoma, GM-CSF is involved in the induction of MDSCs [149, 150], but during immunotherapy in neuroblastoma, GM-CSF appears to activate neutrophils instead. This may be attributed to the concurrent antibody therapy skewing neutrophils toward an anti-tumor phenotype and/or the intermittent cycles of GM-CSF administration rather than a continuous treatment. It is plausible that low-concentration, chronic GM-CSF stimulation induces MDSCs through negative feedback loops, whereas high-concentration, acute GM-CSF stimulation as applied in neuroblastoma therapy, activates neutrophils. Since GM-CSF is not available in Europe, several research groups in the Netherlands have explored the potential use of G-CSF as an alternative to GM-CSF [133]. They concluded that G-CSF exhibited comparable potency to GM-CSF in enhancing ADCC by neutrophils from both healthy donors and neuroblastoma patients. Similar to GM-CSF-enhanced ADCC, the killing mechanism was dependent on FcγRIIa and CD11b/CD18. Given the similarity in performance between G-CSF and GM-CSF, the authors proposed that the addition of G-CSF to anti-GD2 immunotherapy should be evaluated in patients.

Next to GM-CSF, the current treatment regimen for high-risk neuroblastoma includes 13-cis retinoic acid, also known as isotretinoin. In vitro studies have demonstrated that retinoic acid inhibits neuroblastoma cell growth and promotes cellular differentiation [151]. While retinoic acid alone does not improve overall survival [152], pretreatment with retinoic acid enhances the susceptibility of neuroblastoma cells to antibody therapy. Furthermore, differentiated neuroblastoma cells treated with retinoic acid produce IL-8, a crucial cytokine involved in the attraction of neutrophils [134]. IL-8-mediated neutrophil attraction may augment neutrophil-mediated killing induced by anti-GD2 antibody therapy. However, studies conducted in other advanced cancers have reported that IL-8 production and subsequent recruitment of neutrophils/PMN-MDSC can be detrimental, particularly in the context of checkpoint inhibitor treatment [153, 154]. Additionally, retinoic acid can exhibit both immunosuppressive effects, such as differentiation of immunosuppressive TAM [155, 156], and immunostimulatory effects, such as induction of inflammatory anti-tumor macrophages in other cancer types [157, 158]. It is important to note though that most of these studies involved an isomer of 13-cis retinoic acid—all-trans retinoic acid—which may have a slightly different mechanism of action. These studies combined underscore the potential effects of 13-cis retinoic acid treatment on the TME, although its specific impact on the neuroblastoma TME remains largely unknown.

Improving myeloid engagement in anti-GD2 immunotherapy

Several strategies have been developed to improve efficacy of anti-GD2 antibody therapy by targeting myeloid cells. For example, neutrophils exhibit stronger activation in response to IgA antibodies, via their Fc alpha receptor (FcαRI or CD89), compared to IgG antibodies. Our laboratory demonstrated the superior ability of IgA1 anti-GD2 antibodies, which share the same variable region as dinutuximab (ch14.18), in mediating neutrophil-mediated tumor cell killing compared to IgG1 ch14.18 [138]. Importantly, IgA anti-GD2 antibodies did not induce neuropathic pain in mice, unlike IgG anti-GD2 antibodies, primarily due to the absence of the complement factor C1q-binding site. Consequently, IgA antibodies do not activate complement on GD2 expressing nerves, a factor implicated in IgG-induced neuropathic pain [159]. Moreover, we developed a novel form of IgA anti-GD2 antibody (IgA3.0 ch14.18) that is suitable for clinical application, lacking O-glycosylation and featuring mutations that enhance antibody stability and prolong half-life [139]. In long-term mouse models, including xenograft and immunocompetent models with IMR32 and 9464D cells, IgA3.0 ch14.18 demonstrated remarkable anti-tumor efficacy. Furthermore, IgA3.0 ch14.18 effectively induced tumor cell killing by patient neutrophils, while IgG1 ch14.18-mediated tumor cell killing was significantly diminished with patient PBMC compared to healthy donor PBMC. In summary, harnessing and enhancing the anti-tumorigenic activity of neutrophils holds significant promise as a novel approach in the treatment of neuroblastoma.

Another novel strategy to enhance anti-GD2 antibody therapy is to block the CD47/SIRPα axis, analogous to immune checkpoints in T cell biology such as CTLA-4 and PD1/PD-L1. CD47, known as the 'don't eat me' signal, is frequently upregulated in various cancers, including neuroblastoma [47]. Through its interaction with the SIRPα (signal regulatory protein alpha or CD172a) receptor, CD47 inhibits myeloid cell-mediated killing. In the context of neuroblastoma, Theruvath et al. demonstrated that the combination of IgG anti-GD2 and anti-CD47 therapy enhanced macrophage-mediated phagocytosis of neuroblastoma cells in vitro synergistically and led to tumor eradication in mouse xenograft and syngeneic models [21]. The efficacy of this combination therapy primarily relied on phagocytosis of tumor cells by macrophages. Furthermore, the study revealed that ligation of anti-GD2 antibodies upregulated surface calreticulin, which primes macrophages for phagocytosis and disrupts the interaction between GD2 and Siglec 7, another myeloid checkpoint. Consequently, the authors found that the synergy was specific to the combination of anti-GD2 antibodies, as anti-CD47 therapy showed less synergy with antibodies targeting other antigens, such as B7-H3. Encouraged by these findings, a clinical trial investigating the combination of anti-GD2 (dinutuximab) and anti-CD47 (magrolimab) antibodies in neuroblastoma relapse patients was initiated (NCT04751383).

Similarly, anti-CD47 synergizes with antibodies solely recognizing the O-acetyl variant of GD2, developed by the lab of Stephane Birklé [121]. Interestingly, anti-O-acetyl-GD2 antibody therapy enhanced CD47 expression on tumor cells and induced influx of F4/80+ macrophages in a NXS2 liver metastasis model, which further explains the synergy of anti-GD2 and anti-CD47 therapy. Though the previous studies did not find a significant role for neutrophils in mouse models upon anti-GD2 and anti-CD47 combination treatment, Martínez-Sanz and colleagues showed that human neutrophils can be unleashed by this combination strategy in vitro [47]. Dinutuximab induced neutrophil-mediated killing of unmodified neuroblastoma cell lines up to 5–20%, whereas up to 80% killing was achieved in CD47 knockout cell lines. It will be interesting to evaluate in patients whether only macrophages or also other myeloid cells, such as neutrophils are involved in tumor clearance upon combination therapy.

Recently, in other cancers it was described that neutrophils can be activated to their full killing potential by supplementing IgG antibody therapy with TNF and CD40 agonists [160]. Since Voeller and colleagues already described synergy between CD40 agonists and anti-GD2 therapy in neuroblastoma [161], the addition of TNF and CD40 agonists to the treatment regimen could prove a promising new strategy.

Novel strategies for antibody immunotherapy engaging myeloid cells

Currently, alternative targets for antibody therapy in neuroblastoma are being explored, to circumvent the problem of GD2 downregulation, which is particularly prominent in a mesenchymal subset of neuroblastoma cells. Studies have identified B7-H3 as a viable alternative target, given its sustained expression in mesenchymal neuroblastoma cells [162, 163]. The laboratory of Paul Sondel is currently investigating a bispecific SNIPER antibody that targets both GD2 and B7-H3, demonstrating superior efficacy compared to monospecific anti-B7-H3 antibodies and lacking the induction of neuropathic pain [140]. Furthermore, Kendsersky et al. have demonstrated in vivo efficacy of the B7-H3-targeting antibody–drug conjugate (ADC) m276-SL-PBD in patient-derived xenograft (PDX) models of neuroblastoma [141]. However, the involvement of myeloid cells in the effector mechanisms of these novel therapies remains unknown.

Another promising target against which ADCs have been developed is the oncoprotein GPC2, which is overexpressed in neuroblastoma and drives tumor cell proliferation [142, 143]. Anti-GPC2 ADCs have been shown to induce immunogenic cell death in mouse models (NXS2 and 9464D) with GPC2 overexpression, leading to the recruitment of monocytes, macrophages, and MDSCs to the TME. Alongside T cells, macrophages have emerged as major mediators of the anti-GPC2 ADC treatment, and the combination of anti-GPC2 ADCs with anti-CD47 blockade has demonstrated a modest reduction in tumor burden [143].

Though myeloid cells have a negative image in the context of cancer due to their immunosuppressive traits, their potential as effector cells should not be underestimated. Though neutrophils and macrophages are already important mediators in antibody therapy their full potential is not yet reached and myeloid cell engagement can be enhanced, for example by targeting the CD47/SIRPα myeloid checkpoint.

Removal of suppressive myeloid cells from the TME

Direct depletion of myeloid cells

A potential strategy to counteract the suppressive function of myeloid cells in the TME is the depletion of these cell populations as summarized in Table 2. One approach involves using anti-CD11b antibodies to target and deplete all cells of the myeloid lineage. In a study conducted on NXS2 tumor-bearing mice treated with dinutuximab, anti-CD11b antibodies resulted in a modest delay in tumor growth and prolonged survival [164]. Another approach employs anti-CD105 antibodies, which specifically target a transmembrane co-receptor for both TGF-β and bone morphogenic protein-9 (BMP-9). By administering anti-CD105 antibodies, not only monocytes but also MDS and endothelial cells could be depleted [165]. This approach resulted in improved efficacy of anti-GD2 antibodies when combined with adoptively transferred activated human NK cells in neuroblastoma patient-derived xenograft (PDX) models.

Table 2 Comprehensive overview of treatments in neuroblastoma removing myeloid cells from the tumor microenvironment

Park et al. studied myeloid depletion methods in a neuroblastoma PDX model with bispecific GD2 antibodies (GD2-EATs) [166]. They employed various approaches including anti-Ly6G antibodies (targeting neutrophils and PMN-MDSC), anti-Ly6C antibodies (targeting M-MDSC and TAM), anti-CSF-1R antibodies or clodronate liposome (targeting macrophages) and dexamethasone (targeting monocytes). All improved T cell infiltration and survival when combined with GD2-EATs, with macrophage depletion being most effective. Similarly, depletion of human monocytes and macrophages through CSF-1R inhibition using BLZ945, in combination with anti-CSF-1R treatment, enhanced chemotherapeutic efficacy in immunodeficient NOD/SCID mice with neuroblastoma xenografts, independently of T cell contribution [167]. Additionally, Abraham et al. demonstrated that intratumoral injections of small interfering RNAs targeting mouse CSF-1 resulted in significant suppression of tumor growth in SK-N-AS and SK-N-DZ neuroblastoma xenografts, accompanied by decreased TAM infiltration [168]. Finally, treatment of human MDSC with the anti-CD33 ADC gemtuzumab ozogamicin led to cell death in vitro [169]. In co-culture experiments, gemtuzumab ozogamicin restored T cell proliferation and enhanced the activity of anti-GD2 CAR-T cells.

Importantly, depletion of myeloid cells is considered a ‘sledgehammer approach’ as myeloid effector cells that are required for immunotherapy are depleted as well. Targeting specific suppressive myeloid subsets might be more effective. NKT cells offer promise, as they can selectively eliminate immunosuppressive TAMs and MDSCs via CD1d interaction [87, 170]. NKT cells also produce GM-CSF, inducing M1-like TAMs [87, 171] and decreased IL-10 expression in PMN-MDSCs [170], thereby reverting their suppressive function. Although NKT cells are typically present in low numbers within the TME, GD2-CAR NKT cells have shown promise by maintaining their cytotoxic activity against suppressive TAMs [172]. Therefore, harnessing GD2-CAR NKT cells could serve as an indirect approach to deplete immunosuppressive TAMs and MDSCs in the TME.

Myeloablative chemotherapy

The chemotherapeutic doxorubicin (DOX) is currently the most specific drug for the selective removal of MDSC through ROS-mediated apoptosis induction [183], enhancing T cell activity and reducing Tregs in a BALB/c NB mouse model [173, 174]. Residual myeloid cells exhibited reduced expression of Arg-1, indoleamine-pyrrole 2,3-dioxygenase (IDO), as well as STAT3, STAT5, and STAT6, which are key signaling pathways involved in MDSC activation. Notably, in contrast to findings in a murine study on breast cancer [183], DOX affected macrophages in neuroblastoma by inhibiting their polarization from M1 to M2 phenotype [174]. Dopamine also yielded similar outcomes, albeit less potent than DOX. Furthermore, DOX enhanced antigen-specific CD8+ T cell cytotoxicity against neuroblastoma cells by upregulating CD3ζ and L-selectin [175]. Importantly, DOX improved efficacy of anti-GD2 therapy and adoptive T cell transfer, resulting in enhanced survival rates in BALB/c mice [173].

Platinum-based chemotherapeutics, such as cisplatin, have been demonstrated to inhibit STAT signaling, as evidenced by their ability to suppress cyclooxygenase (COX)-2-expressing M-MDSCs induced by melanoma tumor cells in vitro [184]. This inhibitory effect of cisplatin on M-MDSCs was further observed in patients with head and neck squamous cell carcinoma who underwent intravenous cisplatin treatment, leading to a drastic reduction in M-MDSC frequency. Furthermore, the surviving M-MDSCs exhibited decreased expression of COX2 and Arg-1, a result of attenuated STAT3 signaling, consequently impairing their ex vivo T cell inhibitory capacity. In a mouse model of SK-N-DX neuroblastoma, cisplatin was effective in reducing tumor burden. However, the specific mechanism underlying its action in this model was not investigated [176]. 5-FU, a chemotherapeutic agent, exerts its effects by targeting thymidylate synthase and depleting myeloid suppressive cells. In a syngeneic neuroblastoma mouse model, treatment with 5-FU resulted in a reduction of CD11b+ cells within the tumor and enhanced the efficacy of anti-GD2 antibodies [164]. Furthermore, depletion of MDSCs following 5-FU treatment improved the inhibitory effect of recipient leukocyte infusion on local tumor growth in murine neuroblastoma (Neuro2A)-bearing chimeras [177].

Importantly, it should be noted that one of the current chemotherapy regimens COJEC, has been associated with the induction of M2 macrophages in MYCN-amplified neuroblastoma [185]. Similarly, a neuroblastoma PDX model demonstrated that macrophage infiltration promoted the outgrowth of tumor cells that had survived COJEC-like chemotherapy. Consequently, the COJEC regimen could contribute to the development of resistance to anti-GD2 therapy in neuroblastoma and T cell-based therapeutics. Currently, the COJEC regimen, consisting of cisplatin, vincristine, carboplatin, etoposide, and cyclophosphamide, is frequently applied [186]. From a clinical point of view there is a strong wish to add anti-GD2 therapy during induction chemotherapy. Therefore, it could be important to explore alternative chemotherapeutic regimens that can synergize with immunotherapy.

Inhibition of cytokines

As described above, IL-6 stimulation and STAT signaling in myeloid cells are associated with immunosuppressive characteristics, including Arg-1 expression. STAT3 inhibition with AZD1480 or ruxolitinib reduced TAM-mediated MYC upregulation, restraining NBT2 tumor growth in NB-tag and NSG mice [88]. Moreover, Ara et al. demonstrated that IL-6 from monocytes activated STAT3 in neuroblastoma cells, causing drug resistance against etoposide and melphalan in vitro, which was counteracted by STAT3 inhibition or knockdown [178]. Studies in other cancer types suggest that STAT3 inhibition impedes the local proliferation of TAMs, thereby reducing their abundance within the tumor [187]. Furthermore, Louault et al. showed that IL-6 produced by MSC and neuroblastoma cells promoted the survival of TAMs ex vivo [95]. Apoptosis of TAMs was observed upon blocking the IL-6 receptor using the monoclonal antibody Tocilizumab. Upstream, TGF-β stimulated IL-6 production in neuroblastoma cells and MSC, contributing to the suppression of NK cell cytotoxic activity. Treatment with Galunisertib, a TGF-βR1 inhibitor, decreased IL-6 production in co-cultures and restored the activity of NK cells. Therefore, targeting the IL-6/TGF-β axis holds promise as a strategy to selectively deplete TAMs from the neuroblastoma TME.

Blocking recruitment of myeloid cells

Rather than eradicating the myeloid compartment entirely, an alternative strategy involves impeding myeloid cell recruitment to the tumor site. Overexpressed COX enzymes in neuroblastoma drive myeloid cell attraction by synthesizing prostaglandins, including PGE2 [55, 188, 189]. Blocking COX with aspirin in TH-MYCN mice led to diminished tumor burden and reduced tumor-associated myeloid cells like MDSCs, immature DCs, and TAMs [55, 180].

The CCL2/CCR2 axis is pivotal for recruiting monocytes, myeloid cells, and plasmacytoid DCs in MYCN-nonamplified neuroblastoma [89]. In other cancer types, inhibiting the CCL2/CCR2 axis with CCR2 inhibitors or anti-CCL2 antibodies has demonstrated reduced myeloid cell recruitment and improved clinical outcomes [190,191,192]. Sphingosine-1 (S1P), a bioactive lipid, induced CCL2 expression in neuroblastoma via S1P2 [181]. Inhibiting S1P2 with the stable derivative AB1, but not its precursor JTE-013, decreased macrophage infiltration in neuroblastoma xenografts, underscoring AB1's potential to inhibit TAM infiltration [182].

Furthermore, certain chemotherapeutic and immunotherapeutic drugs impact immune cell recruitment to tumors. For example, anti-TGF-β reduced M2 TAM recruitment in a neuroblastoma model, and low-dose oxaliplatin hindered neutrophil migration to tumors [179].

While depleting or blocking myeloid cells has been proven effective in neuroblastoma, it also depletes potential immunotherapy effectors. Additionally, targeting specific cytokines or immunosuppressive mediators produced by MDSC can be useful, but this only targets a small part of the problem. Reprogramming suppressive myeloid cells may be the optimal strategy, interrupting immunosuppressive traits while preserving myeloid cells as effectors of immunotherapy.

Reprogramming suppressive myeloid cells into an active phenotype

Finally, a highly elegant approach to target immunosuppressive myeloid cells involves reprogramming their suppressive phenotype into an antitumor, immunostimulatory state (Table 3). Various published therapeutic approaches have demonstrated successful myeloid cell reprogramming with consequent T cell activation. For instance, ibrutinib, an irreversible molecular inhibitor of Bruton's tyrosine kinase (BTK), was shown to reverse T cell suppression mediated by murine MDSC in a neuroblastoma mouse model [193]. This was evidenced by altered NO production and decreased mRNA expression of immunosuppressive factors Ido, Arg, and Tgfb. Moreover, ibrutinib-mediated BTK inhibition increased CD8+ T cell infiltration and enhanced response to anti-PD-L1 checkpoint inhibitor therapy. As previously discussed, neuroblastoma-derived factors hinder early myeloid cell differentiation to monocytes and macrophages, promoting monocyte suppressive function via M-CSF/CSF-1R interaction [57]. CSF-1R inhibitor BLZ945 effectively blocked CSF-1R-expressing suppressive myeloid cell generation and reversed tumor-educated monocyte suppression. Combining CSF-1R inhibition with anti-PD-1/PD-L1 immune checkpoint blockade in TH-MYCN mice enhanced activation of tumor-reactive T cells. Interestingly, PD-1 blockade induced enhanced T cell M-CSF secretion, enhancing monocyte suppressive capacity [194]. Thus, BLZ945-CSF-1R inhibition combined with PD-1 blockade synergistically controlled tumor growth. Additionally, catechins such as Polyphenon E exhibit neuroblastoma anticancer effects by inhibiting MDSC activity. Administered via drinking water in multiple neuroblastoma mouse models, Polyphenon E hindered MDSC development and mobility, and facilitated their differentiation into neutrophils through 67 kDa laminin receptor signaling and G-CSF induction [65]. Moreover, it lowered Arg-1 expression on MDSCs, promoting T cell proliferation in patient metastases. Another treatment strategy that potentiates T cells by reprogramming myeloid cells is addition of neutrophil-activating protein (NAP) in therapies like CAR T cell or oncolytic virus treatments. Tumino et al. reported an increase in circulating PMN-MDSC numbers upon GD2.CAR T-cell therapy [195]. Circulating PMN-MDSCs inversely correlated with GD2.CAR T-cell levels, potentially predicting treatment response. Moreover, Stroncek et al. demonstrated that monocytes inhibit the expansion of GD2.CAR T cells in neuroblastoma patients [196]. In accordance with this, GD2.CAR T cells were ineffective in a syngeneic NXS2 neuroblastoma model. However, GD2.CAR T cells designed to express NAP delayed tumor outgrowth by generating a ‘hot’ TME with high infiltration of neutrophils, M1 macrophages, NK cells, CD8+ T cells, DCs, and a reduced number of Tregs [197], which potentiated GD2.CAR T cell therapy. Likewise, oncolytic virus therapy can be enhanced by incorporating NAP into the virus load. Oncolytic Vaccinia viruses carrying a GD2 mimotope were ineffective against NXS2 tumors, but the inclusion of NAP led to tumor growth inhibition [198].

Table 3 Comprehensive overview of treatments in neuroblastoma reprogramming myeloid cells into an immunostimulatory phenotype

Other studies did report strategies to reprogram myeloid cells, but did not observe or investigate the indirect effects on T cells. COX inhibitors have shown promise in limiting myeloid infiltration by inhibiting PGE2, but their use is constrained by potential side effects. An alternative approach by Kock et al. selectively targeted microsomal prostaglandin E synthase-1 (mPGES-1) using a small molecule inhibitor in an immunocompetent transgenic neuroblastoma mouse model with MYCN oncogene expression [199]. This targeted inhibition of mPGES-1 suppressed PGE2 production specifically in CAFs, resulting in reduced tumor growth, impaired angiogenesis, and macrophage polarization towards a pro-inflammatory M1 phenotype. In addition, human FABP4-expressing macrophages were discovered to promote migration, invasion, and tumor growth of neuroblastoma cell lines [204]. This was facilitated through FABP4 binding to ATPB, which triggered ATPB ubiquitination, reduced ATP levels, and deactivated the NF-kB/RelA-IL-1a pathways, driving macrophages into an M2 phenotype. Blocking antibodies against IL-1a effectively countered FABP4-induced increased migration and invasion. Ligating CD40 offers another approach to target myeloid cells, as it activates antigen-presenting cells including macrophages. In murine NXS2 models, anti-CD40 treatment led to delayed tumor progression by inducing a proinflammatory M1 phenotype in macrophages and stimulating Th1 cytokine production [205]. This treatment also synergized with CpG-containing oligodeoxynucleotides (CpG), promoting upregulation of intracellular Toll-like receptor (TLR)-9 and generating NO, IFNγ, TNFα, and IL-12 in A/J mice bearing NXS2 tumors [206]. When combined with multidrug chemotherapy, radiation, anti-GD2, and anti-CTLA-4, this anti-CD40 and CpG therapy exhibited potent anti-tumor effects in mice with 9464D neuroblastoma tumors, repolarizing TAMs towards an M1 phenotype [161]. Furthermore, histone deacetylase (HDAC) inhibitors have been explored in cancer therapy due to their ability to prevent histone deacetylation, resulting in altered gene and protein expression [212]. In neuroblastoma, the HDAC inhibitor vorinostat has shown enhanced efficacy in combination with anti-GD2 antibody treatment in a TH-MYCN mouse model [200]. Vorinostat treatment increased GD2 expression in neuroblastoma tumors, improving responsiveness to anti-GD2 therapy. It also modified the myeloid composition of the TME, enhancing TAMs with mixed M1 and M2 phenotypes while reducing M-MDSC. Vorinostat-treated TAMs exhibited heightened ADCC capability, downregulated immunosuppressive genes, and upregulated FcyRI and FcyRII/III. Although clinical trials are lacking, the combination of anti-GD2 therapy and vorinostat holds promise for neuroblastoma treatment. Vorinostat was well-tolerated in a phase I trial with isotretinoin [201] and showed favorable response rates in a phase II trial with 131I-metaiodobenzylguanidine (MIBG) [202].

Furthermore, several therapeutic strategies have been reported that suggest alteration of gene expression of tumor cells or macrophages as a promising strategy to reprogram myeloid cells. BET bromodomain inhibitors like JQ1 and I-BET726 displace BRD4 from the MYCN promoter, leading to improved survival in various in vivo neuroblastoma models by inhibiting tumor growth and MYCN downregulation [209, 210]. Combined PI3K and BRD4 inhibition also shows promise, suppressing MYCN expression and inhibiting neuroblastoma cell growth and metastasis both in vitro and in vivo [211]. While not investigated in neuroblastoma, PI3K/BRD4 blockade has demonstrated immunomodulatory effects. In tumor-bearing mice, it reduces MDSC recruitment, enhances MHCII+ TAMs with a pro-inflammatory M1 phenotype, and promotes CD8+ T cell infiltration and activation within the tumor [213]. These findings suggest that aside from MYCN, PI3K/BRD4 blockade may influence myeloid cell migration and polarization, potentially contributing to its anti-tumor effects in neuroblastoma. Further research is needed to fully understand this mechanism. In addition, Rac2, a small GTPase, has been linked to the polarization of macrophages from M1 to M2 phenotype in vivo [208]. Rac2-deficient mice studies indicated that although macrophage infiltration into the TME remained unchanged, the absence of Rac2 led to a prevalent M1 phenotype in macrophages. In a syngeneic neuroblastoma (9464D) model, Rac2-deficient mice exhibited significantly reduced tumor growth, suggesting the contribution of M2 macrophages to tumor progression. Notably, Rac2 is also implicated in tumor-induced angiogenesis, implying that the impact of Rac2 knockdown on tumor inhibition might involve both M1 macrophage polarization and the suppression of tumor angiogenesis. Potential therapeutic strategies involving Rac2 inhibition, using inhibitors like NSC23766, wortmannin, and LY294002, warrant further exploration in neuroblastoma cancer development [214, 215].

Finally, some studies have been published that demonstrate the possibility to target cytokines to reprogram myeloid cells. Relation et al. introduced a novel strategy involving MSCs engineered to express IFNγ, injected directly into neuroblastoma tumors [203]. This led to decreased tumor proliferation and improved survival in a CHL-255 orthotopic neuroblastoma model, without increasing macrophage infiltration, but instead polarizing TAMs to an M1 phenotype. Additionally, incubating neuroblastoma cell lines with human monocytes increased IL-1β and TNFα-expressing macrophages, signifying an M1 phenotype via AKT phosphorylation [45]. IL-1β and TNFα from these macrophages stimulated arginine metabolism in neuroblastoma cells, promoting tumor cell proliferation. AKT inhibition using MK-2206 halted this effect by blocking IL-1β and TNFα secretion by macrophages. Blocking arginine uptake in tumor cells with L-NAME or depleting arginine with BCT-100 hindered neuroblastoma cell differentiation. BCT-100 treatment delayed neuroblastoma development in a TH-MYCN mouse model, prolonging survival. A phase I/II study with BCT-100 treatment is currently conducted in patients with neuroblastoma (NCT03455140). To our knowledge, results of this study have not been published yet.

Reprogramming the myeloid compartment in neuroblastoma offers an elegant strategy, preserving potential effector cells. This approach can also indirectly reactivate tumor-infiltrating T cells, and enhance T cell therapies, such as CAR T cells and checkpoint inhibitors. In addition to all myeloid-targeting treatments discussed in this review, many other treatment strategies are promising, but have not yet been studied in neuroblastoma. Some examples are the inhibition of myeloperoxidase, preventing lipid peroxidation in PMN-MDSC, inhibition of fatty acid transport protein 2 (FATP2), preventing uptake of arachidonic acid and synthesis of PGE2, DR5 agonists inducing MDSC apoptosis via TNF-related apoptosis-inducing ligand (TRAIL) and inhibition of Arg-1 [216,217,218,219]. Strategies involving IDO, NO, or TOLLIP inhibition, ATRA treatment or other HDAC inhibitors than vorinostat, such as entinostat and ricolinostat were proven effective as well and are thoroughly reviewed elsewhere [28, 220,221,222]. These approaches hold opportunities for new neuroblastoma treatments, and should be further investigated in the future.

Discussion

As in other solid tumors, the presence of neutrophils, MDSCs and macrophages in neuroblastoma is generally associated with worse survival and immunosuppression. Neutrophils appear to be associated with a worse prognosis in high-risk neuroblastoma, though exact mechanisms remain unknown and are obscured by changing nomenclature, with (suppressive) neutrophils now often referred to as PMN-MDSC. It is clear that MDSC are immunosuppressive in both patients and in mouse models. Interestingly, we observed that M-MDSC were more immunosuppressive than PMN-MDSC in mouse models, but due to limited patient studies it is uncertain whether this is the case in neuroblastoma patients as well. Regarding macrophages, research findings diverge. While some noted M2 macrophage induction by neuroblastoma, others primarily observed M1 macrophages, with both types potentially linked to poor clinical outcome. These discrepancies likely stem from distinct genetic profiles within neuroblastoma; tumors with chromosome 11q deletion or MYCN amplification tend to favor M2 phenotypes, whereas MYCN-nonamplified tumors have a higher prevalence of M1 macrophages [21, 72, 73, 85].

Because of their immunosuppressive nature and association with disease progression, myeloid cells are an excellent target for immunotherapy. Interestingly, immunosuppressive myeloid cells can still be engaged as effector cells, as is illustrated by the fact that neutrophils are important effector cells in anti-GD2 antibody therapy. This major role for neutrophils appears to be specific for anti-GD2 immunotherapy and/or neuroblastoma since neutrophils are much less involved as effectors of IgG antibody therapy in other cancers. One potential explanation for this phenomenon could be the timing of immunotherapy, often administered shortly after ASCT (90–120 days). Neutrophils, being highly abundant and among the first immune cells to recover post-ASCT, may possess a relative advantage compared to immune cells such as NK cells [223]. Additionally, post-ASCT NK cell recovery revealed a sustained enhanced metabolic immune cell profile relative to pre-ASCT levels in multiple myeloma patients [224, 225]. This is accompanied by a shift in NK cell subset distribution, correlating with reduced progression-free survival. These findings imply long-lasting ASCT-induced impacts on NK cell function.

In summary, substantial effort is put in developing T cell- and NKT cell-based therapeutics for neuroblastoma, among which CAR T cells and PD1/PD-L checkpoint inhibitors are the most prominent. However, results have been disappointing, apart from a breakthrough regarding GD2.CAR T cells for neuroblastoma patients with low tumor burden [14]. For adaptive immunity to be restored in neuroblastoma, first the immunosuppressive myeloid cells must be tackled. In this review, we highlighted several myeloid-targeting strategies that restored the ability of T cells to kill tumor cells, including depletion of myeloid cells using anti-Ly6G, anti-CD33 or the BTK inhibitor ibrutinib and myeloid reprogramming by DOX chemotherapy, anti-CSF1R, administration of Polyphenon E, or the addition of NAP to therapies. The combination of myeloid-targeting drugs and T cell-based therapeutics for neuroblastoma holds great promise for the future.

Availability of data and materials

This review article summarizes and discusses available literature and did not generate new data.

Abbreviations

ADC:

Antibody-drug conjugate

ADCC:

Antigen-dependent cellular cytotoxicity

ADNR:

Adrenergic noradrenergic cells

Arg-1:

Arginase 1

ASC:

Autologous stem cell transplantation

ATP:

Adenosine triphosphate

BMP-9:

Bone morphogenic protein-9

BTK:

Bruton's tyrosine kinase

CAFs:

Cancer-associated fibroblasts

CAR:

Chimeric antigen receptor

CCL:

C-C motif chemokine ligand

COX:

Cyclooxygenase

CpG:

CpG-containing oligodeoxynucleotides

CR3:

Complement receptor 3

CSF-1R:

Colony stimulating factor 1 receptor

CXCR:

C-X-C motif chemokine receptor

DC:

Dendritic cells

DOX:

Doxorubicin

E-MDSC:

Early myeloid-derived suppressor cells

EFS:

Event-free survival

FcαRI:

Fc alpha receptor

GD2:

Disialoganglioside

GM-CSF:

Granulocyte/macrophage-Colony Stimulating Factor

HDAC:

Histone deacetylase

HIF-2α:

Hypoxia inducible factor 2α

HLA:

Human leukocyte antigen

ICAM-1:

Intercellular adhesion molecule-1

IFNγ:

Interferon gamma

IGF-2:

Insulin-like growth factor 2

IL:

Interleukin

L-NAME:

N-nitro-L-arginine

LFA-1:

Lymphocyte function-associated antigen 1

LMR:

Leukocyte-to-monocyte ratio

M-CSF:

Macrophage-colony stimulating factor

MDK:

Midkine

MES:

Mesenchymal cell-like cells

METTL:

Methyltransferase-like 3

MIF:

Macrophage migration inhibitory factor

M-MDSC:

Monocytic myeloid-derived suppressor cells

Mac-1:

Membrane-activated complex 1

MDSC:

Myeloid-derived suppressor cells

mPGES-1:

Prostaglandin E synthase-1

MSC:

Mesenchymal stromal cells

NA-NB:

MYCN non-amplified neuroblastomas

NAP:

Neutrophil-activating protein

NET:

Neutrophil extracellular traps

NK:

Natural killer

NKT:

Natural killer T

NLR:

Neutrophil-to-lymphocyte ratio

NO:

Nitric oxide

OS:

Overall survival

PAD:

Peptide arginine deiminase

PBMC:

Peripheral blood mononuclear cells

PDX:

Patient-derived xenograft

PMN:

Polymorphonuclear cells

PMN-MDSC:

Polymorphonuclear myeloid-derived suppressor cells

ROS:

Reactive oxygen species

S1P:

Lipid sphingosine-1

SIRPα:

Signal regulatory protein alpha

TAMs:

Tumor-associated macrophages

TGF-β:

Transforming growth factor beta

TILs:

Tumor-infiltrating lymphocytes

TLR:

Toll-like receptor

TME:

Tumor microenvironment

TNFα:

Tumor necrosis factor alpha

VEGF:

Vascular endothelial growth factor

References

  1. De Preter K, Vandesompele J, Heimann P, et al. Human fetal neuroblast and neuroblastoma transcriptome analysis confirms neuroblast origin and highlights neuroblastoma candidate genes. Genome Biol. 2006;7:R84-r84.

    Article  Google Scholar 

  2. (2019) Neuroblastoma - Childhood: Statistics. 2020

  3. Brodeur GM, Seeger RC, Schwab M, et al. (1984) Amplification of N-myc in untreated human neuroblastomas correlates with advanced disease stage. Science. 1979;224:1121–4. https://doi.org/10.1126/science.6719137.

    Article  Google Scholar 

  4. Huang M, Weiss WA. Neuroblastoma and MYCN. Cold Spring Harb PerspectMed. 2013;3:a014415.

    Article  CAS  Google Scholar 

  5. De Bernardi B, Mosseri V, Rubie H, et al. Treatment of localised resectable neuroblastoma. Results of the LNESG1 study by the SIOP Europe Neuroblastoma Group. Br J Cancer. 2008;99:1027–33. https://doi.org/10.1038/sj.bjc.6604640.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Strother DR, London WB, Lou SM, et al. Outcome after surgery alone or with restricted use of chemotherapy for patients with low-risk neuroblastoma: results of Children’s Oncology Group study P9641. J Clin Oncol. 2012;30:1842–8. https://doi.org/10.1200/JCO.2011.37.9990.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Maris JM. Recent advances in neuroblastoma. N Engl J Med. 2010;362:2202–11. https://doi.org/10.1056/NEJMra0804577.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Ladenstein R, Potschger U, Valteau-Couanet D, et al. Interleukin 2 with anti-GD2 antibody ch14.18/CHO (dinutuximab beta) in patients with high-risk neuroblastoma (HR-NBL1/SIOPEN): a multicentre, randomised, phase 3 trial. Lancet Oncol. 2018;19:1617–29. https://doi.org/10.1016/S1470-2045(18)30578-3.

    Article  CAS  PubMed  Google Scholar 

  9. Pugh TJ, Morozova O, Attiyeh EF, et al. The genetic landscape of high-risk neuroblastoma. Nat Genet. 2013;45:279–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Lampson LA, Fisher CA, Whelan JP. Striking paucity of HLA-A, B, C and beta 2-microglobulin on human neuroblastoma cell lines. J Immunol. 1983;130:2471–8.

    Article  CAS  PubMed  Google Scholar 

  11. Kramer K, Gerald WL, Kushner BH, et al. Disialoganglioside G(D2) loss following monoclonal antibody therapy is rare in neuroblastoma. Clin Cancer Res. 1998;4:2135–9.

    CAS  PubMed  Google Scholar 

  12. Yu AL, Gilman AL, Ozkaynak MF, et al. Anti-GD2 antibody with GM-CSF, interleukin-2, and isotretinoin for neuroblastoma. N Engl J Med. 2010;363:1324–34. https://doi.org/10.1056/NEJMoa0911123.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Cheung NK, Cheung IY, Kushner BH, et al. Murine anti-GD2 monoclonal antibody 3F8 combined with granulocyte-macrophage colony-stimulating factor and 13-cis-retinoic acid in high-risk patients with stage 4 neuroblastoma in first remission. J Clin Oncol. 2012;30:3264–70. https://doi.org/10.1200/JCO.2011.41.3807.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Del Bufalo F, De Angelis B, Caruana I, et al. GD2-CART01 for relapsed or refractory high-risk neuroblastoma. N Engl J Med. 2023;388:1284–95. https://doi.org/10.1056/NEJMoa2210859.

    Article  PubMed  Google Scholar 

  15. Egler RA, Burlingame SM, Nuchtern JG, Russell HV. Interleukin-6 and soluble interleukin-6 receptor levels as markers of disease extent and prognosis in neuroblastoma. Clin Cancer Res. 2008;14:7028–34. https://doi.org/10.1158/1078-0432.CCR-07-5017.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Silverman AM, Nakata R, Shimada H, et al. A galectin-3-dependent pathway upregulates interleukin-6 in the microenvironment of human neuroblastoma. Cancer Res. 2012;72:2228–38. https://doi.org/10.1158/0008-5472.CAN-11-2165.

    Article  CAS  PubMed  Google Scholar 

  17. Zhen Z, Guo X, Liao R, et al. Involvement of IL-10 and TGF-beta in HLA-E-mediated neuroblastoma migration and invasion. Oncotarget. 2016;7:44340–9. https://doi.org/10.18632/oncotarget.10041.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Soldati R, Berger E, Zenclussen AC, et al. Neuroblastoma triggers an immunoevasive program involving galectin-1-dependent modulation of T cell and dendritic cell compartments. International journal of cancer Journal international du cancer. 2012;131:1131–41. https://doi.org/10.1002/ijc.26498[doi].

    Article  CAS  PubMed  Google Scholar 

  19. Buchel G, Schulte JH, Harrison L, et al. Immune response modulation by Galectin-1 in a transgenic model of neuroblastoma. Oncoimmunology. 2016;5:e1131378. https://doi.org/10.1080/2162402X.2015.1131378.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Brandetti E, Veneziani I, Melaiu O, et al. MYCN is an immunosuppressive oncogene dampening the expression of ligands for NK-cell-activating receptors in human high-risk neuroblastoma. Oncoimmunology. 2017;6:e1316439. https://doi.org/10.1080/2162402X.2017.1316439.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Theruvath J, Menard M, Smith BAH, et al. Anti-GD2 synergizes with CD47 blockade to mediate tumor eradication. Nat Med. 2022;28:333–44. https://doi.org/10.1038/s41591-021-01625-x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Lee HC, Wondimu A, Liu Y, et al. Ganglioside inhibition of CD8+ T cell cytotoxicity: interference with lytic granule trafficking and exocytosis. J Immunol. 2012;189:3521–7.

    Article  CAS  PubMed  Google Scholar 

  23. Binnewies M, Roberts EW, Kersten K, et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med. 2018;24:541–50. https://doi.org/10.1038/s41591-018-0014.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Zafari R, Razi S, Rezaei N. The role of dendritic cells in neuroblastoma: Implications for immunotherapy. Immunobiology. 2022;227:152293. https://doi.org/10.1016/j.imbio.2022.152293.

    Article  CAS  PubMed  Google Scholar 

  25. Bronte V, Brandau S, Chen SH, et al. Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards. Nat Commun. 2016;7:1–10.

    Article  Google Scholar 

  26. Condamine T, Dominguez GA, Youn J-I, 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. https://doi.org/10.1126/sciimmunol.aaf8943.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Cassetta L, Baekkevold ES, Brandau S, et al. Deciphering myeloid-derived suppressor cells: isolation and markers in humans, mice and non-human primates. Cancer Immunol Immunother. 2019;68:687–97. https://doi.org/10.1007/s00262-019-02302-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Veglia F, Sanseviero E, Gabrilovich DI. Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity. Nat Rev Immunol. 2021;21:485–98. https://doi.org/10.1038/s41577-020-00490-y.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. 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. https://doi.org/10.1126/sciimmunol.aay6017.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Sagiv JY, Michaeli J, Assi S, et al. Phenotypic diversity and plasticity in circulating neutrophil subpopulations in cancer. Cell Rep. 2015;10:562–73. https://doi.org/10.1016/j.celrep.2014.12.039.

    Article  CAS  PubMed  Google Scholar 

  31. 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;216:2150–69. https://doi.org/10.1084/jem.20181952.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Furze RC, Rankin SM. Neutrophil mobilization and clearance in the bone marrow. Immunology. 2008;125:281–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Tak T, Tesselaar K, Pillay J, et al. Whatˈs your age again? Determination of human neutrophil half-lives revisited. J Leukoc Biol. 2013;94:595–601. https://doi.org/10.1189/jlb.1112571.

    Article  CAS  PubMed  Google Scholar 

  34. Rankin SM. The bone marrow: a site of neutrophil clearance. J Leukoc Biol. 2010;88:241–51. https://doi.org/10.1189/jlb.0210112.

    Article  CAS  PubMed  Google Scholar 

  35. Gentles AJ, Newman AM, Liu CL, et al. The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat Med. 2015;21:938–45. https://doi.org/10.1038/nm.3909.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Morandi F, Barco S, Stigliani S, et al. Altered erythropoiesis and decreased number of erythrocytes in children with neuroblastoma. Oncotarget. 2017;8:53194–209. https://doi.org/10.18632/oncotarget.18285.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Zeng L, Liu X-Y, Chen K, et al. Phosphoserine phosphatase as an indicator for survival through potentially influencing the infiltration levels of immune cells in neuroblastoma. Front Cell Dev Biol. 2022;10:873710. https://doi.org/10.3389/fcell.2022.873710.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Zheng C, Liu S, Feng J, Zhao X. Prognostic value of inflammation biomarkers for survival of patients with neuroblastoma. Cancer Manag Res. 2020;12:2415–25. https://doi.org/10.2147/CMAR.S245622.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Zhang Y, Zhang C, Ma Y, et al. Prediction to the prognosis of children with neuroblastoma by nomogram based on the first-diagnosed inflammatory markers. Pediatr Surg Int. 2022;39:17. https://doi.org/10.1007/s00383-022-05302-z.

    Article  PubMed  Google Scholar 

  40. Lee JW, Bae JS, Kim JH, et al. Absolute neutrophil count after the first chemotherapy cycle as a surrogate marker for treatment outcomes in patients with neuroblastoma. Cancer Res Treat. 2022;54:259–68. https://doi.org/10.4143/crt.2021.010.

    Article  CAS  PubMed  Google Scholar 

  41. Erbe AK, Diccianni MB, Mody R, et al. KIR/KIR-ligand genotypes and clinical outcomes following chemoimmunotherapy in patients with relapsed or refractory neuroblastoma: a report from the Children’s Oncology Group. J Immunother Cancer. 2023;11(2):e006530.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Birdsall HH. Induction of ICAM-1 on human neural cells and mechanisms of neutrophil-mediated injury. Am J Pathol. 1991;139:1341–50.

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Chuluyan HE, Lang BJ, Issekutz AC. Differential mechanisms of neutrophil and monocyte adhesion on neuroblastoma cells: CD18 and VLA-4 integrins mediate adhesion to SK-N-SH, but not to SK-N-MC cell line. J Neurosci Res. 2000;60:649–55.

    Article  CAS  PubMed  Google Scholar 

  44. Blaheta RA, Powerski M, Hudak L, et al. Tumor-endothelium cross talk blocks recruitment of neutrophils to endothelial cells: a novel mechanism of endothelial cell anergy. Neoplasia. 2009;11:1054–63. https://doi.org/10.1593/neo.09762.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Fultang L, Gamble LD, Gneo L, et al. Macrophage-Derived IL1beta and TNFalpha Regulate Arginine Metabolism in Neuroblastoma. Cancer Res. 2019;79:611–24. https://doi.org/10.1158/0008-5472.CAN-18-2139.

    Article  CAS  PubMed  Google Scholar 

  46. Chen RL, Reynolds CP, Seeger RC. Neutrophils are cytotoxic and growth-inhibiting for neuroblastoma cells with an anti-GD2 antibody but, without cytotoxicity, can be growth-stimulating. Cancer Immunol Immunother. 2000;48:603–12. https://doi.org/10.1007/s002620050008.

    Article  CAS  PubMed  Google Scholar 

  47. Martínez-Sanz P, Hoogendijk AJ, Verkuijlen PJJH, et al. CD47-SIRPα Checkpoint Inhibition Enhances Neutrophil-Mediated Killing of Dinutuximab-Opsonized Neuroblastoma Cells. Cancers (Basel). 2021;13(17):4261. https://doi.org/10.3390/cancers13174261.

    Article  CAS  PubMed  Google Scholar 

  48. Gabrilovich DI, Ostrand-Rosenberg S, Bronte V. Coordinated regulation of myeloid cells by tumours. NatRevImmunol. 2012;12:253–68. https://doi.org/10.1038/nri3175.

    Article  CAS  Google Scholar 

  49. Veglia F, Perego M, Gabrilovich D. Myeloid-derived suppressor cells coming of age review-article. Nat Immunol. 2018;19:108–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Ostrand-Rosenberg S. Myeloid derived-suppressor cells: their role in cancer and obesity. Curr Opin Immunol. 2018;51:68–75. https://doi.org/10.1016/j.coi.2018.03.007.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Marvel D, Gabrilovich DI. Myeloid-derived suppressor cells in the tumor microenvironment: expect the unexpected. J Clin Invest. 2015;125:3356–64. https://doi.org/10.1172/JCI80005.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Gabrilovich DI. Myeloid-Derived Suppressor Cells CancerImmunolRes. 2017;5:3–8. https://doi.org/10.1158/2326-6066.CIR-16-0297.

    Article  CAS  Google Scholar 

  53. Mazzoni A, Bronte V, Visintin A, et al. Myeloid suppressor lines inhibit T cell responses by an NO-dependent mechanism. J Immunol. 2002;168:689–95. https://doi.org/10.4049/jimmunol.168.2.689.

    Article  CAS  PubMed  Google Scholar 

  54. Kusmartsev S, Nefedova Y, Yoder D, Gabrilovich DI. Antigen-specific inhibition of CD8+ T cell response by immature myeloid cells in cancer is mediated by reactive oxygen species. J Immunol. 2004;172:989–99. https://doi.org/10.4049/jimmunol.172.2.989.

    Article  CAS  PubMed  Google Scholar 

  55. Carlson LM, Rasmuson A, Idborg H, et al. Low-dose aspirin delays an inflammatory tumor progression in vivo in a transgenic mouse model of neuroblastoma. Carcinogenesis. 2013;34:1081–8. https://doi.org/10.1093/carcin/bgt009.

    Article  CAS  PubMed  Google Scholar 

  56. Bianchi G, Vuerich M, Pellegatti P, et al. ATP/P2X7 axis modulates myeloid-derived suppressor cell functions in neuroblastoma microenvironment. Cell Death Dis. 2014;5:e1135.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Mao Y, Eissler N, Blanc KL, et al. Targeting suppressive myeloid cells potentiates checkpoint inhibitors to control spontaneous neuroblastoma. Clin Cancer Res. 2016;22:3849–59. https://doi.org/10.1158/1078-0432.CCR-15-1912.

    Article  CAS  PubMed  Google Scholar 

  58. Costa A, Thirant C, Kramdi A, et al. Single-cell transcriptomics reveals shared immunosuppressive landscapes of mouse and human neuroblastoma. J Immunother Cancer. 2022;10(8):e004807. https://doi.org/10.1136/jitc-2022-004807.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Eissler N, Sveinbjörnsson B, Kock A, et al. Immune suppression by myeloid-derived suppressor cells, MDSCs, in MYCN-driven neuroblastoma provides a potential target for cancer immunotherapy. J Immunother Cancer. 2014;2:P203. https://doi.org/10.1186/2051-1426-2-S3-P203.

    Article  PubMed Central  Google Scholar 

  60. Gowda M, Godder K, Kmieciak M, et al. Distinct signatures of the immune responses in low risk versus high risk neuroblastoma. J Transl Med. 2011;9:170. https://doi.org/10.1186/1479-5876-9-170.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Dolcetti L, Peranzoni E, Ugel S, et al. Hierarchy of immunosuppressive strength among myeloid-derived suppressor cell subsets is determined by GM-CSF. Eur J Immunol. 2009;40:22–35. https://doi.org/10.1002/eji.200939903.

    Article  CAS  Google Scholar 

  62. Haverkamp JM, Smith AM, Weinlich R, et al. Myeloid-derived suppressor activity is mediated by monocytic lineages maintained by continuous inhibition of extrinsic and intrinsic death pathways. Immunity. 2014;41:947–59. https://doi.org/10.1016/j.immuni.2014.10.020.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Mussai F, Egan S, Hunter S, et al. Neuroblastoma arginase activity creates an immunosuppressive microenvironment that impairs autologous and engineered immunity. Cancer Res. 2015;75:3043–53. https://doi.org/10.1158/0008-5472.CAN-14-3443.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Lazic D, Kromp F, Rifatbegovic F, et al. Landscape of Bone Marrow Metastasis in Human Neuroblastoma Unraveled by Transcriptomics and Deep Multiplex Imaging. Cancers (Basel). 2021;13(17):4311. https://doi.org/10.3390/cancers13174311.

    Article  CAS  PubMed  Google Scholar 

  65. Santilli G, Piotrowska I, Cantilena S, et al. Polyphenon [corrected] E enhances the antitumor immune response in neuroblastoma by inactivating myeloid suppressor cells. Clin Cancer Res. 2013;19:1116–25. https://doi.org/10.1158/1078-0432.CCR-12-2528.

    Article  CAS  PubMed  Google Scholar 

  66. Gowda M, Payne KK, Godder K, Manjili MH. HLA-DR expression on myeloid cells is a potential prognostic factor in patients with high-risk neuroblastoma. Oncoimmunology. 2013;2:e26616. https://doi.org/10.4161/onci.26616.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Richards DM, Hettinger J, Feuerer M. Monocytes and macrophages in cancer: development and functions. Cancer Microenviron. 2013;6:179–91. https://doi.org/10.1007/s12307-012-0123-x.

    Article  CAS  PubMed  Google Scholar 

  68. Wu Y, Hirschi KK. Tissue-Resident Macrophage Development and Function. Front Cell Dev Biol. 2020;8:617879. https://doi.org/10.3389/fcell.2020.617879.

    Article  PubMed  Google Scholar 

  69. Kumar V, Patel S, Tcyganov E, Gabrilovich DI. The nature of myeloid-derived suppressor cells in the tumor microenvironment. Trends Immunol. 2016;37:208–20. https://doi.org/10.1016/j.it.2016.01.004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Mosser DM, Edwards JP. Exploring the full spectrum of macrophage activation. NatRevImmunol. 2008;8:958–69. https://doi.org/10.1038/nri2448.

    Article  CAS  Google Scholar 

  71. Asgharzadeh S, Salo JA, Ji L, et al. Clinical significance of tumor-associated inflammatory cells in metastatic neuroblastoma. J Clin Oncol. 2012;30:3525–32. https://doi.org/10.1200/JCO.2011.40.9169.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Larsson K, Kock A, Idborg H, et al. COX/mPGES-1/PGE2 pathway depicts an inflammatory-dependent high-risk neuroblastoma subset. Proc Natl Acad Sci U S A. 2015;112:8070–5. https://doi.org/10.1073/pnas.1424355112.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Hashimoto O, Yoshida M, Koma Y, et al. Collaboration of cancer-associated fibroblasts and tumour-associated macrophages for neuroblastoma development. J Pathol. 2016;240:211–23. https://doi.org/10.1002/path.4769.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Batchu S. Immunological landscape of Neuroblastoma and its clinical significance. Cancer Treat Res Commun. 2021;26:100274. https://doi.org/10.1016/j.ctarc.2020.100274.

    Article  PubMed  Google Scholar 

  75. Challagundla KB, Wise PM, Neviani P, et al. Exosome-mediated transfer of microRNAs within the tumor microenvironment and neuroblastoma resistance to chemotherapy. J Natl Cancer Inst. 2015;107(7):djv135. https://doi.org/10.1093/jnci/djv135.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Liu Q, Wang Z, Jiang Y, et al. Single-cell landscape analysis reveals distinct regression trajectories and novel prognostic biomarkers in primary neuroblastoma. Genes Dis. 2022;9:1624–38. https://doi.org/10.1016/j.gendis.2021.12.020.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Tang XX, Shimada H, Ikegaki N. Macrophage-mediated anti-tumor immunity against high-risk neuroblastoma. Genes Immun. 2022;23:129–40. https://doi.org/10.1038/s41435-022-00172-w.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Verhoeven BM, Mei S, Olsen TK, et al. The immune cell atlas of human neuroblastoma. Cell Rep Med. 2022;3:100657. https://doi.org/10.1016/j.xcrm.2022.100657.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Blavier L, Nakata R, Neviani P, et al. The capture of extracellular vesicles endogenously released by xenotransplanted tumours induces an inflammatory reaction in the premetastatic niche. J Extracell Vesicles. 2023;12:e12326. https://doi.org/10.1002/jev2.12326.

    Article  CAS  PubMed  Google Scholar 

  80. Yu Y, Zeng Y, Xia X, et al. Establishment and Validation of a Prognostic Immune Signature in Neuroblastoma. Cancer Control. 2021;28:10732748211033752. https://doi.org/10.1177/10732748211033751.

    Article  Google Scholar 

  81. Fiegel HC, Kaifi JT, Wachowiak R, et al. Midkine is highly expressed in neuroblastoma tissues. Pediatr Surg Int. 2008;24:1355–9. https://doi.org/10.1007/s00383-008-2263-0.

    Article  PubMed  Google Scholar 

  82. Garcia-Gerique L, García M, Garrido-Garcia A, et al. MIF/CXCR4 signaling axis contributes to survival, invasion, and drug resistance of metastatic neuroblastoma cells in the bone marrow microenvironment. BMC Cancer. 2022;22:669. https://doi.org/10.1186/s12885-022-09725-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Zhang Y, Zuo C, Liu L, et al. Single-cell RNA-sequencing atlas reveals an MDK-dependent immunosuppressive environment in ErbB pathway-mutated gallbladder cancer. J Hepatol. 2021;75:1128–41. https://doi.org/10.1016/j.jhep.2021.06.023.

    Article  CAS  PubMed  Google Scholar 

  84. Klemke L, De Oliveira T, Witt D, et al. Hsp90-stabilized MIF supports tumor progression via macrophage recruitment and angiogenesis in colorectal cancer. Cell Death Dis. 2021;12:155. https://doi.org/10.1038/s41419-021-03426-z.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Raieli S, Di Renzo D, Lampis S, et al. MYCN drives a tumor immunosuppressive environment which impacts survival in neuroblastoma. Front Oncol. 2021;11:625207. https://doi.org/10.3389/fonc.2021.625207.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Zhang P, Wu X, Basu M, et al. MYCN amplification is associated with repressed cellular immunity in neuroblastoma: an in silico immunological analysis of TARGET database. Front Immunol. 2017;8:1473. https://doi.org/10.3389/fimmu.2017.01473.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Song L, Asgharzadeh S, Salo J, et al. Valpha24-invariant NKT cells mediate antitumor activity via killing of tumor-associated macrophages. J Clin Invest. 2009;119:1524–36. https://doi.org/10.1172/JCI37869.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Hadjidaniel MD, Muthugounder S, Hung LT, et al. Tumor-associated macrophages promote neuroblastoma via STAT3 phosphorylation and up-regulation of c-MYC. Oncotarget. 2017;8:91516–29. https://doi.org/10.18632/oncotarget.21066.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Kacher J, Manches O, Aspord C, et al. Impaired antitumor immune response in MYCN-amplified neuroblastoma is associated with lack of CCL2 Secretion and poor dendritic cell recruitment. Cancer research communications. 2022;2:577–89. https://doi.org/10.1158/2767-9764.CRC-21-0134.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Metelitsa LS, Wu HW, Wang H, et al. Natural killer T cells infiltrate neuroblastomas expressing the chemokine CCL2. J Exp Med. 2004;199:1213–21. https://doi.org/10.1084/jem.20031462.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Van de Velde L-A, Allen EK, Crawford JC, et al. Neuroblastoma formation requires unconventional CD4 T cells and arginase-1-dependent myeloid cells. Cancer Res. 2021;81:5047–59. https://doi.org/10.1158/0008-5472.CAN-21-0691.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Pahlman S, Mohlin S. Hypoxia and hypoxia-inducible factors in neuroblastoma. Cell Tissue Res. 2018;372:269–75. https://doi.org/10.1007/s00441-017-2701.

    Article  PubMed  Google Scholar 

  93. Liu D, Song L, Wei J, et al. IL-15 protects NKT cells from inhibition by tumor-associated macrophages and enhances antimetastatic activity. J Clin Invest. 2012;122:2221–33. https://doi.org/10.1172/JCI59535.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Pietras A, Gisselsson D, Ora I, et al. High levels of HIF-2alpha highlight an immature neural crest-like neuroblastoma cell cohort located in a perivascular niche. J Pathol. 2008;214:482–8. https://doi.org/10.1002/path.2304.

    Article  CAS  PubMed  Google Scholar 

  95. Louault K, Porras T, Lee M-H, et al. Fibroblasts and macrophages cooperate to create a pro-tumorigenic and immune resistant environment via activation of TGF-β/IL-6 pathway in neuroblastoma. Oncoimmunology. 2022;11:2146860. https://doi.org/10.1080/2162402X.2022.2146860.

    Article  PubMed  PubMed Central  Google Scholar 

  96. Borriello L, Nakata R, Sheard MA, et al. Cancer-associated fibroblasts share characteristics and protumorigenic activity with mesenchymal stromal cells. Cancer Res. 2017;77:5142–57. https://doi.org/10.1158/0008-5472.CAN-16-2586.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Uhm TG, Kim BS, Chung IY. Eosinophil development, regulation of eosinophil-specific genes, and role of eosinophils in the pathogenesis of asthma. Allergy Asthma ImmunolRes. 2012;4:68–79. https://doi.org/10.4168/aair.2012.4.2.68.

    Article  CAS  Google Scholar 

  98. Grisaru-Tal S, Itan M, Klion AD, Munitz A. A new dawn for eosinophils in the tumour microenvironment. Nat Rev Cancer. 2020;20:594–607. https://doi.org/10.1038/s41568-020-0283-9.

    Article  CAS  PubMed  Google Scholar 

  99. El-Badry O, Helman L, Chatten J, et al. Insulin-like growth-factor-ii-mediated proliferation of human neuroblastoma. J Clin Invest. 1991;87:648–57. https://doi.org/10.1172/JCI115042.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. El-Badry OM. Growth regulation of human neuroblastoma. In: Liu ET, editor. Benz C. US: Springer; 1993. p. 105–28.

    Google Scholar 

  101. Álvarez-Errico D, Vento-Tormo R, Sieweke M, Ballestar E. Epigenetic control of myeloid cell differentiation, identity and function. Nat Rev Immunol. 2015;15:7–17. https://doi.org/10.1038/nri3777.

    Article  CAS  PubMed  Google Scholar 

  102. Ivashkiv LB, Park SH. Epigenetic regulation of myeloid cells. Microbiol Spectr. 2016;4(3):10. https://doi.org/10.1128/microbiolspec.MCHD-0010-2015.

    Article  Google Scholar 

  103. Porta C, Riboldi E, Ippolito A, Sica A. Molecular and epigenetic basis of macrophage polarized activation. Semin Immunol. 2015;27:237–48. https://doi.org/10.1016/j.smim.2015.10.003.

    Article  CAS  PubMed  Google Scholar 

  104. Quagliano A, Gopalakrishnapillai A, Barwe SP. Understanding the mechanisms by which epigenetic modifiers avert therapy resistance in cancer. Front Oncol. 2020;10:992. https://doi.org/10.3389/fonc.2020.00992.

    Article  PubMed  PubMed Central  Google Scholar 

  105. Xu X, Peng Q, Jiang X, et al. Metabolic reprogramming and epigenetic modifications in cancer: from the impacts and mechanisms to the treatment potential. Exp Mol Med. 2023;55:1357–70. https://doi.org/10.1038/s12276-023-01020-1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Xu L, Zhou C, Liang Y, et al. Epigenetic modifications in the accumulation and function of myeloid-derived suppressor cells. Front Immunol. 2022;13:1016870. https://doi.org/10.3389/fimmu.2022.1016870.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Zhu D, Zhang Y, Wang S. Histone citrullination: a new target for tumors. Mol Cancer. 2021;20:90. https://doi.org/10.1186/s12943-021-01373-z.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Xiong J, He J, Zhu J, et al. Lactylation-driven METTL3-mediated RNA m6A modification promotes immunosuppression of tumor-infiltrating myeloid cells. Mol Cell. 2022;82:1660-1677.e10. https://doi.org/10.1016/j.molcel.2022.02.033.

    Article  CAS  PubMed  Google Scholar 

  109. Sido JM, Yang X, Nagarkatti PS, Nagarkatti M. Δ9-Tetrahydrocannabinol-mediated epigenetic modifications elicit myeloid-derived suppressor cell activation via STAT3/S100A8. J Leukoc Biol. 2015;97:677–88. https://doi.org/10.1189/jlb.1A1014-479R.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Hegde VL, Singh UP, Nagarkatti PS, Nagarkatti M. Critical role of mast cells and peroxisome proliferator-activated receptor γ in the induction of myeloid-derived suppressor cells by marijuana cannabidiol in vivo. J Immunol. 2015;194:5211–22. https://doi.org/10.4049/jimmunol.1401844.

    Article  CAS  PubMed  Google Scholar 

  111. Fetahu IS, Esser-Skala W, Dnyansagar R, et al. Single-cell transcriptomics and epigenomics unravel the role of monocytes in neuroblastoma bone marrow metastasis. Nat Commun. 2023;14:3620. https://doi.org/10.1038/s41467-023-39210-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Wrangle J, Wang W, Koch A, et al. Alterations of immune response of non-small cell lung cancer with Azacytidine. Oncotarget. 2013;4:2067–79. https://doi.org/10.18632/oncotarget.1542.

    Article  PubMed  PubMed Central  Google Scholar 

  113. Juergens RA, Wrangle J, Vendetti FP, et al. Combination epigenetic therapy has efficacy in patients with refractory advanced non-small cell lung cancer. Cancer Discov. 2011;1:598–607. https://doi.org/10.1158/2159-8290.CD-11-0214.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. van Groningen T, Koster J, Valentijn LJ, et al. Neuroblastoma is composed of two super-enhancer-associated differentiation states. Nat Genet. 2017;49:1261–6. https://doi.org/10.1038/ng.3899.

    Article  CAS  PubMed  Google Scholar 

  115. Boeva V, Louis-Brennetot C, Peltier A, et al. Heterogeneity of neuroblastoma cell identity defined by transcriptional circuitries. Nat Genet. 2017;49:1408–13. https://doi.org/10.1038/ng.3921.

    Article  CAS  PubMed  Google Scholar 

  116. van Groningen T, Akogul N, Westerhout EM, et al. A NOTCH feed-forward loop drives reprogramming from adrenergic to mesenchymal state in neuroblastoma. Nat Commun. 2019;10:1530. https://doi.org/10.1038/s41467-019-09470-w.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. van Nes J, Chan A, van Groningen T, et al. A NOTCH3 transcriptional module induces cell motility in neuroblastoma. Clin Cancer Res. 2013;19:3485–94. https://doi.org/10.1158/1078-0432.CCR-12-3021.

    Article  CAS  PubMed  Google Scholar 

  118. Wolpaw AJ, Grossmann LD, Dessau JL, et al. Epigenetic state determines inflammatory sensing in neuroblastoma. Proc Natl Acad Sci U S A. 2022;119(6):e2102358119. https://doi.org/10.1073/pnas.2102358119.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Sengupta S, Das S, Crespo AC, et al. Mesenchymal and adrenergic cell lineage states in neuroblastoma possess distinct immunogenic phenotypes. Nat Cancer. 2022;3:1228–46. https://doi.org/10.1038/s43018-022-00427-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Munn DH, Cheung NK. Antibody-dependent antitumor cytotoxicity by human monocytes cultured with recombinant macrophage colony-stimulating factor. Induction of efficient antibody-mediated antitumor cytotoxicity not detected by isotope release assays. J Exp Med. 1989;170:511–26. https://doi.org/10.1084/jem.170.2.511.

    Article  CAS  PubMed  Google Scholar 

  121. Bahri M, Kailayangiri S, Vermeulen S, et al. SIRPα-specific monoclonal antibody enables antibody-dependent phagocytosis of neuroblastoma cells. Cancer Immunol Immunother. 2022;71:71–83. https://doi.org/10.1007/s00262-021-02968-7.

    Article  CAS  PubMed  Google Scholar 

  122. Bruchelt G, Handgretinger R, Kimmig A, et al. Effects of granulocytes on human neuroblastoma cells measured by chemiluminescence and chromium-51 release assay. J Biolumin Chemilumin. 1989;3:93–6. https://doi.org/10.1002/bio.1170030212.

    Article  CAS  PubMed  Google Scholar 

  123. Kushner BH, Cheung NK. GM-CSF enhances 3F8 monoclonal antibody-dependent cellular cytotoxicity against human melanoma and neuroblastoma. Blood. 1989;73:1936–41.

    Article  CAS  PubMed  Google Scholar 

  124. Barker E, Mueller BM, Handgretinger R, et al. Effect of a chimeric anti-ganglioside GD2 antibody on cell-mediated lysis of human neuroblastoma cells. Cancer Res. 1991;51:144–9.

    CAS  PubMed  Google Scholar 

  125. Barker E, Reisfeld RA. A mechanism for neutrophil-mediated lysis of human neuroblastoma cells. Cancer Res. 1993;53:362–7.

    CAS  PubMed  Google Scholar 

  126. Kushner BH, Cheung NK. Absolute requirement of CD11/CD18 adhesion molecules, FcRII and the phosphatidylinositol-linked FcRIII for monoclonal antibody-mediated neutrophil antihuman tumor cytotoxicity. Blood. 1992;79:1484–90.

    Article  CAS  PubMed  Google Scholar 

  127. Tibbetts R, Yeo KK, Muthugounder S, et al. Anti-disialoganglioside antibody internalization by neuroblastoma cells as a mechanism of immunotherapy resistance. Cancer Immunol Immunother. 2022;71:153–64. https://doi.org/10.1007/s00262-021-02963-y.

    Article  CAS  PubMed  Google Scholar 

  128. Cheung NK, Cheung IY, Kramer K, et al. Key role for myeloid cells: phase II results of anti-G(D2) antibody 3F8 plus granulocyte-macrophage colony-stimulating factor for chemoresistant osteomedullary neuroblastoma. Int J Cancer. 2014;135:2199–205. https://doi.org/10.1002/ijc.28851.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Cheung IY, Hsu K, Cheung NK. Activation of peripheral-blood granulocytes is strongly correlated with patient outcome after immunotherapy with anti-GD2 monoclonal antibody and granulocyte-macrophage colony-stimulating factor. J Clin Oncol. 2012;30:426–32. https://doi.org/10.1200/JCO.2011.37.6236.

    Article  CAS  PubMed  Google Scholar 

  130. Cheung NK, Sowers R, Vickers AJ, et al. FCGR2A polymorphism is correlated with clinical outcome after immunotherapy of neuroblastoma with anti-GD2 antibody and granulocyte macrophage colony-stimulating factor. J Clin Oncol. 2006;24:2885–90. https://doi.org/10.1200/JCO.2005.04.6011.

    Article  CAS  PubMed  Google Scholar 

  131. Szanto CL, Cornel AM, Tamminga SM, et al. Immune Monitoring during Therapy Reveals Activitory and Regulatory Immune Responses in High-Risk Neuroblastoma. Cancers (Basel). 2021;13(9):2096. https://doi.org/10.3390/cancers13092096.

    Article  CAS  PubMed  Google Scholar 

  132. Yu AL, Uttenreuther-Fischer MM, Huang CS, et al. Phase I trial of a human-mouse chimeric anti-disialoganglioside monoclonal antibody ch14.18 in patients with refractory neuroblastoma and osteosarcoma. J Clin Oncol. 1998;16:2169–80. https://doi.org/10.1200/JCO.1998.16.6.2169.

    Article  CAS  PubMed  Google Scholar 

  133. Martinez Sanz P, van Rees DJ, van Zogchel LMJ, et al. G-CSF as a suitable alternative to GM-CSF to boost dinutuximab-mediated neutrophil cytotoxicity in neuroblastoma treatment. J Immunother Cancer. 2021;9(5):e002259.

    Article  PubMed  PubMed Central  Google Scholar 

  134. Yang KD, Cheng SN, Wu NC, Shaio MF. Induction of interleukin-8 expression in neuroblastoma cells by retinoic acid: implication of leukocyte chemotaxis and activation. Pediatr Res. 1993;34:720–4. https://doi.org/10.1203/00006450-199312000-00005.

    Article  CAS  PubMed  Google Scholar 

  135. Troschke-Meurer S, Siebert N, Marx M, et al. Low CD4+/CD25+/CD127 regulatory T cell- and high INF-γ levels are associated with improved survival of neuroblastoma patients treated with long-term infusion of ch14.18/CHO combined with interleukin-2. Oncoimmunology. 2019;8:1661194. https://doi.org/10.1080/2162402X.2019.1661194.

    Article  PubMed  PubMed Central  Google Scholar 

  136. Rousseau RF, Haight AE, Hirschmann-Jax C, et al. Local and systemic effects of an allogeneic tumor cell vaccine combining transgenic human lymphotactin with interleukin-2 in patients with advanced or refractory neuroblastoma. Blood. 2003;101:1718–26. https://doi.org/10.1182/blood-2002-08-2493.

    Article  CAS  PubMed  Google Scholar 

  137. Russell HV, Strother D, Mei Z, et al. A phase 1/2 study of autologous neuroblastoma tumor cells genetically modified to secrete IL-2 in patients with high-risk neuroblastoma. J Immunother. 2008;31:812–9. https://doi.org/10.1097/CJI.0b013e3181869893.

    Article  CAS  PubMed  Google Scholar 

  138. Evers M, Stip M, Keller K, et al. Anti-GD2 IgA kills tumors by neutrophils without antibody-associated pain in the preclinical treatment of high-risk neuroblastoma. J Immunother Cancer. 2021;9(10):e003163.

    Article  PubMed  PubMed Central  Google Scholar 

  139. Stip MC, Evers M, Nederend M, et al. IgA antibody immunotherapy targeting GD2 is effective in preclinical neuroblastoma models. J Immunother Cancer. 2023;11:e006948. https://doi.org/10.1136/jitc-2023-006948.

    Article  PubMed  PubMed Central  Google Scholar 

  140. Erbe A, Gerhardt D, Hernandez R, et al. 461 Improving specific targeting of tumors through bispecific SNIPER antibodies. J Immunother Cancer. 2020;8:A280-LP−A280. https://doi.org/10.1136/jitc-2020-SITC2020.0461.

    Article  Google Scholar 

  141. Kendsersky NM, Lindsay J, Kolb EA, et al. The B7–H3-targeting antibody-drug conjugate m276-SL-PBD Is potently effective against pediatric cancer preclinical solid tumor models. Clin Cancer Res. 2021;27:2938–46. https://doi.org/10.1158/1078-0432.CCR-20-4221.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Bosse KR, Raman P, Zhu Z, et al. Identification of GPC2 as an oncoprotein and candidate immunotherapeutic target in high-risk neuroblastoma. Cancer Cell. 2017;32:295-309.e12. https://doi.org/10.1016/j.ccell.2017.08.003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Pascual-Pasto G, McIntyre B, Shraim R, et al. GPC2 antibody-drug conjugate reprograms the neuroblastoma immune milieu to enhance macrophage-driven therapies. J Immunother Cancer. 2022;10(12):e004704.

    Article  PubMed  PubMed Central  Google Scholar 

  144. Siebert N, Jensen C, Troschke-Meurer S, et al. Neuroblastoma patients with high-affinity FCGR2A, -3A and stimulatory KIR 2DS2 treated by long-term infusion of anti-GD(2) antibody ch14.18/CHO show higher ADCC levels and improved event-free survival. Oncoimmunology. 2016;5:e1235108.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Hank JA, Robinson RR, Surfus J, et al. Augmentation of antibody dependent cell mediated cytotoxicity following in vivo therapy with recombinant interleukin 2. Cancer Res. 1990;50:5234–9.

    CAS  PubMed  Google Scholar 

  146. Li J, Gyorffy S, Lee S, Kwok CS. Effect of recombinant human interleukin 2 on neutrophil adherence to endothelial cells in vitro. Inflammation. 1996;20:361–72. https://doi.org/10.1007/bf01486739.

    Article  PubMed  Google Scholar 

  147. Troschke-Meurer S, Siebert N, Marx M, et al. Low CD4(+)/CD25(+)/CD127(-) regulatory T cell- and high INF-gamma levels are associated with improved survival of neuroblastoma patients treated with long-term infusion of ch14.18/CHO combined with interleukin-2. Oncoimmunology. 2019;8:1661194. https://doi.org/10.1080/2162402X.2019.1661194.

    Article  PubMed  PubMed Central  Google Scholar 

  148. Rivoltini L, Viggiano V, Spinazze S, et al. In vitro anti-tumor activity of eosinophils from cancer patients treated with subcutaneous administration of interleukin 2. Role of interleukin 5. International journal of cancer Journal international du cancer. 1993;54:8–15. https://doi.org/10.1002/ijc.2910540103.

    Article  CAS  PubMed  Google Scholar 

  149. Kohanbash G, McKaveney K, Sakaki M, et al. GM-CSF promotes the immunosuppressive activity of glioma-infiltrating myeloid cells through interleukin-4 receptor-alpha. Cancer Res. 2013;73:6413–23. https://doi.org/10.1158/0008-5472.CAN-12-4124.

    Article  CAS  PubMed  Google Scholar 

  150. Thorn M, Guha P, Cunetta M, et al. Tumor-associated GM-CSF overexpression induces immunoinhibitory molecules via STAT3 in myeloid-suppressor cells infiltrating liver metastases. Cancer Gene Ther. 2016;23:188–98. https://doi.org/10.1038/cgt.2016.19.

    Article  CAS  PubMed  Google Scholar 

  151. Sidell N. Retinoic acid-induced growth inhibition and morphologic differentiation of human neuroblastoma cells in vitro. J Natl Cancer Inst. 1982;68:589–96.

    CAS  PubMed  Google Scholar 

  152. Matthay KK, Reynolds CP, Seeger RC, et al. Long-term results for children with high-risk neuroblastoma treated on a randomized trial of myeloablative therapy followed by 13-cis-retinoic acid: a children’s oncology group study. J Clin Oncol. 2009;27:1007–13. https://doi.org/10.1200/JCO.2007.13.8925.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Yuen KC, Liu L-F, Gupta V, et al. High systemic and tumor-associated IL-8 correlates with reduced clinical benefit of PD-L1 blockade. Nat Med. 2020;26:693–8. https://doi.org/10.1038/s41591-020-0860-1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Schalper KA, Carleton M, Zhou M, et al. Elevated serum interleukin-8 is associated with enhanced intratumor neutrophils and reduced clinical benefit of immune-checkpoint inhibitors. Nat Med. 2020;26:688–92. https://doi.org/10.1038/s41591-020-0856-x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Devalaraja S, To TKJ, Folkert IW, et al. Tumor-derived retinoic acid regulates intratumoral monocyte differentiation to promote immune suppression. Cell. 2020;180:1098-1114.e16. https://doi.org/10.1016/j.cell.2020.02.042.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. (2020) Retinoic Acid Mediates Monocyte Differentiation and Immune Response. Cancer Discov 10:OF7. https://doi.org/10.1158/2159-8290.CD-RW2020-044

  157. Rao E, Hou Y, Huang X, et al. All-trans retinoic acid overcomes solid tumor radioresistance by inducing inflammatory macrophages. Sci Immunol. 2021;6(60):eaba8426. https://doi.org/10.1126/sciimmunol.aba8426.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Tilsed CM, Casey TH, de Jong E, et al. Retinoic acid induces an IFN-driven inflammatory tumour microenvironment, sensitizing to immune checkpoint therapy. Front Oncol. 2022;12:849793. https://doi.org/10.3389/fonc.2022.849793.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Sorkin LS, Otto M, Baldwin WM 3rd, et al. Anti-GD(2) with an FC point mutation reduces complement fixation and decreases antibody-induced allodynia. Cancer Res. 2010;149:362–7. https://doi.org/10.1016/j.pain.2010.01.024.

    Article  CAS  Google Scholar 

  160. Linde IL, Prestwood TR, Qiu J, et al. Neutrophil-activating therapy for the treatment of cancer. Cancer Cell. 2023;41:356-372.e10. https://doi.org/10.1016/j.ccell.2023.01.002.

    Article  CAS  PubMed  Google Scholar 

  161. Voeller J, Erbe AK, Slowinski J, et al. Combined innate and adaptive immunotherapy overcomes resistance of immunologically cold syngeneic murine neuroblastoma to checkpoint inhibition. J Immunother Cancer. 2019;7:344. https://doi.org/10.1186/s40425-019-0823-6.

    Article  PubMed  PubMed Central  Google Scholar 

  162. Castriconi R, Dondero A, Augugliaro R, et al. Identification of 4Ig-B7-H3 as a neuroblastoma-associated molecule that exerts a protective role from an NK cell-mediated lysis. Proc Natl Acad Sci U S A. 2004;101:12640–5. https://doi.org/10.1073/pnas.0405025101.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Dondero A, Morini M, Cangelosi D, et al. Multiparametric flow cytometry highlights B7–H3 as a novel diagnostic/therapeutic target in GD2neg/low neuroblastoma variants. J Immunother Cancer. 2021;9(4):e002293.

    Article  PubMed  PubMed Central  Google Scholar 

  164. Siebert N, Zumpe M, von Lojewski L, et al. Reduction of CD11b(+) myeloid suppressive cells augments anti-neuroblastoma immune response induced by the anti-GD(2) antibody ch14.18/CHO. Oncoimmunology. 2020;9:1836768. https://doi.org/10.1080/2162402X.2020.1836768.

    Article  PubMed  PubMed Central  Google Scholar 

  165. Wu H-W, Sheard MA, Malvar J, et al. Anti-CD105 antibody eliminates tumor microenvironment cells and enhances Anti-GD2 antibody immunotherapy of neuroblastoma with activated natural killer cells. Clin Cancer Res. 2019;25:4761–74. https://doi.org/10.1158/1078-0432.CCR-18-3358.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Park JA, Wang L, Cheung N-KV. Modulating tumor infiltrating myeloid cells to enhance bispecific antibody-driven T cell infiltration and anti-tumor response. J Hematol Oncol. 2021;14:142. https://doi.org/10.1186/s13045-021-01156-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Webb MW, Sun J, Sheard MA, et al. Colony stimulating factor 1 receptor blockade improves the efficacy of chemotherapy against human neuroblastoma in the absence of T lymphocytes. Int J Cancer. 2018;143:1483–93. https://doi.org/10.1002/ijc.31532.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Abraham D, Zins K, Sioud M, et al. Stromal cell-derived CSF-1 blockade prolongs xenograft survival of CSF-1-negative neuroblastoma. Int J Cancer. 2010;126:1339–52. https://doi.org/10.1002/ijc.24859.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Fultang L, Panetti S, Ng M, et al. MDSC targeting with Gemtuzumab ozogamicin restores T cell immunity and immunotherapy against cancers. EBioMedicine. 2019;47:235–46. https://doi.org/10.1016/j.ebiom.2019.08.025.

    Article  PubMed  PubMed Central  Google Scholar 

  170. Mussai F, De Santo C, Cerundolo V. Interaction between invariant NKT cells and myeloid-derived suppressor cells in cancer patients: evidence and therapeutic opportunities. J Immunother. 2012;35:449–59. https://doi.org/10.1097/CJI.0b013e31825be926.

    Article  CAS  PubMed  Google Scholar 

  171. Courtney AN, Tian G, Marinova E, et al. NKT cells control tumor associated macrophages and metastatic growth in neuroblastoma. J Immun. 2017;198:204.24-204.24. https://doi.org/10.4049/jimmunol.198.Supp.204.24.

    Article  Google Scholar 

  172. Heczey A, Liu D, Tian G, et al. Invariant NKT cells with chimeric antigen receptor provide a novel platform for safe and effective cancer immunotherapy. Blood. 2014;124:2824–33. https://doi.org/10.1182/blood-2013-11-541235.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Xu W-L, Shi B-J, Li S-L, et al. Targeted inhibition of myeloid-derived suppressor cells in the tumor microenvironment by low-dose doxorubicin to improve immune efficacy in murine neuroblastoma. Chin Med J (Engl). 2020;134:334–43. https://doi.org/10.1097/CM9.0000000000001234.

    Article  CAS  PubMed  Google Scholar 

  174. Xu W, Li S, Li M, et al. Targeted elimination of myeloid-derived suppressor cells via regulation of the STAT pathway alleviates tumor immunosuppression in neuroblastoma. Immunol Lett. 2021;240:31–40. https://doi.org/10.1016/j.imlet.2021.09.011.

    Article  CAS  PubMed  Google Scholar 

  175. Xu W, Li S, Li M, et al. Upregulation of CD3ζ and L-selectin in antigen-specific cytotoxic T lymphocytes by eliminating myeloid-derived suppressor cells with doxorubicin to improve killing efficacy of neuroblastoma cells in vitro. J Clin Lab Anal. 2022;36:e24158. https://doi.org/10.1002/jcla.24158.

    Article  CAS  PubMed  Google Scholar 

  176. D’Incalci M, Colombo T, Ubezio P, et al. The combination of yondelis and cisplatin is synergistic against human tumor xenografts. Eur J Cancer. 2003;39:1920–6. https://doi.org/10.1016/s0959-8049(03)00490-8.

    Article  PubMed  Google Scholar 

  177. Dierckx de Casterlé I, Fevery S, Rutgeerts O, et al. Reduction of myeloid-derived suppressor cells reinforces the anti-solid tumor effect of recipient leukocyte infusion in murine neuroblastoma-bearing allogeneic bone marrow chimeras. Cancer Immunol Immunother. 2018;67:589–603. https://doi.org/10.1007/s00262-017-2114-8.

    Article  CAS  PubMed  Google Scholar 

  178. Ara T, Nakata R, Sheard MA, et al. Critical role of STAT3 in IL-6-mediated drug resistance in human neuroblastoma. Cancer Res. 2013;73:3852–64. https://doi.org/10.1158/0008-5472.CAN-12-2353.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Lucarini V, Melaiu O, D’Amico S, et al. Combined mitoxantrone and anti-TGFβ treatment with PD-1 blockade enhances antitumor immunity by remodelling the tumor immune landscape in neuroblastoma. J Exp Clin Cancer Res. 2022;41:326. https://doi.org/10.1186/s13046-022-02525-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Carlson LM, Kogner P. Neuroblastoma-related inflammation: May small doses of aspirin be suitable for small cancer patients? Oncoimmunology. 2013;2:e24658. https://doi.org/10.4161/onci.24658.

    Article  PubMed  PubMed Central  Google Scholar 

  181. Li M-H, Harel M, Hla T, Ferrer F. Induction of chemokine (C-C motif) ligand 2 by sphingosine-1-phosphate signaling in neuroblastoma. J Pediatr Surg. 2014;49:1286–91. https://doi.org/10.1016/j.jpedsurg.2014.04.001.

    Article  PubMed  PubMed Central  Google Scholar 

  182. Li M-H, Swenson R, Harel M, et al. Antitumor activity of a novel sphingosine-1-Phosphate 2 Antagonist, AB1, in Neuroblastoma. J Pharmacol Exp Ther. 2015;354:261–8. https://doi.org/10.1124/jpet.115.224519.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Alizadeh D, Trad M, Hanke NT, et al. Doxorubicin eliminates myeloid-derived suppressor cells and enhances the efficacy of adoptive T-cell transfer in breast cancer. Cancer Res. 2014;74:104–18. https://doi.org/10.1158/0008-5472.CAN-13-1545.

    Article  CAS  PubMed  Google Scholar 

  184. Van Wigcheren GF, De Haas N, Mulder TA, et al. Cisplatin inhibits frequency and suppressive activity of monocytic myeloid-derived suppressor cells in cancer patients. Oncoimmunology. 2021;10:1935557. https://doi.org/10.1080/2162402X.2021.1935557.

    Article  PubMed  PubMed Central  Google Scholar 

  185. Valind A, Verhoeven BM, Enoksson J, et al. Macrophage infiltration promotes regrowth in MYCN-amplified neuroblastoma after chemotherapy. Oncoimmunology. 2023;12:2184130. https://doi.org/10.1080/2162402X.2023.2184130.

    Article  PubMed  PubMed Central  Google Scholar 

  186. Peinemann F, Tushabe DA, van Dalen EC, Berthold F. Rapid COJEC versus standard induction therapies for high-risk neuroblastoma. Cochrane Database Syst Rev. 2015;2015(5):CD010774.

    Article  PubMed  PubMed Central  Google Scholar 

  187. Fletcher JS, Springer MG, Choi K, et al. STAT3 inhibition reduces macrophage number and tumor growth in neurofibroma. Oncogene. 2019;38:2876–84. https://doi.org/10.1038/s41388-018-0600-x.

    Article  CAS  PubMed  Google Scholar 

  188. Johnsen JI, Lindskog M, Ponthan F, et al. Cyclooxygenase-2 is expressed in neuroblastoma, and nonsteroidal anti-inflammatory drugs induce apoptosis and inhibit tumor growth in vivo. Cancer Res. 2004;64:7210–5. https://doi.org/10.1158/0008-5472.CAN-04-1795.

    Article  CAS  PubMed  Google Scholar 

  189. Rasmuson A, Kock A, Fuskevag OM, et al. Autocrine prostaglandin E2 signaling promotes tumor cell survival and proliferation in childhood neuroblastoma. PLoS ONE. 2012;7:e29331. https://doi.org/10.1371/journal.pone.0029331.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Pienta KJ, Machiels J-P, Schrijvers D, et al. Phase 2 study of carlumab (CNTO 888), a human monoclonal antibody against CC-chemokine ligand 2 (CCL2), in metastatic castration-resistant prostate cancer. Invest New Drugs. 2013;31:760–8. https://doi.org/10.1007/s10637-012-9869-8.

    Article  CAS  PubMed  Google Scholar 

  191. Sandhu SK, Papadopoulos K, Fong PC, et al. A first-in-human, first-in-class, phase I study of carlumab (CNTO 888), a human monoclonal antibody against CC-chemokine ligand 2 in patients with solid tumors. Cancer Chemother Pharmacol. 2013;71:1041–50. https://doi.org/10.1007/s00280-013-2099-8.

    Article  CAS  PubMed  Google Scholar 

  192. Nywening TM, Wang-Gillam A, Sanford DE, et al. Targeting tumour-associated macrophages with CCR2 inhibition in combination with FOLFIRINOX in patients with borderline resectable and locally advanced pancreatic cancer: a single-centre, open-label, dose-finding, non-randomised, phase 1b trial. Lancet Oncol. 2016;17:651–62. https://doi.org/10.1016/S1470-2045(16)00078-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  193. Ishfaq M, Pham T, Beaman C, et al. BTK Inhibition Reverses MDSC-Mediated Immunosuppression and Enhances Response to Anti-PDL1 Therapy in Neuroblastoma. Cancers (Basel). 2021;13(4):817. https://doi.org/10.3390/cancers13040817.

    Article  CAS  PubMed  Google Scholar 

  194. Eissler N, Mao Y, Brodin D, et al. Regulation of myeloid cells by activated T cells determines the efficacy of PD-1 blockade. Oncoimmunology. 2016;5:e1232222. https://doi.org/10.1080/2162402X.2016.1232222.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Tumino N, Weber G, Besi F, et al. Polymorphonuclear myeloid-derived suppressor cells impair the anti-tumor efficacy of GD2.CAR T-cells in patients with neuroblastoma. J Hematol Oncol. 2021;14:191.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Stroncek DF, Ren J, Lee DW, et al. Myeloid cells in peripheral blood mononuclear cell concentrates inhibit the expansion of chimeric antigen receptor T cells. Cytotherapy. 2016;18:893–901. https://doi.org/10.1016/j.jcyt.2016.04.003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Jin C, Ma J, Ramachandran M, et al. CAR T cells expressing a bacterial virulence factor trigger potent bystander antitumour responses in solid cancers. Nat Biomed Eng. 2022;6:830–41. https://doi.org/10.1038/s41551-022-00875-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Ma J, Jin C, Čančer M, et al. Concurrent expression of HP-NAP enhances antitumor efficacy of oncolytic vaccinia virus but not for Semliki Forest virus. Mol Ther Oncolytics. 2021;21:356–66. https://doi.org/10.1016/j.omto.2021.04.016.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Kock A, Larsson K, Bergqvist F, et al. Inhibition of microsomal prostaglandin e synthase-1 in cancer-associated fibroblasts suppresses neuroblastoma tumor growth. EBioMedicine. 2018;32:84–92. https://doi.org/10.1016/j.ebiom.2018.05.008.

    Article  PubMed  PubMed Central  Google Scholar 

  200. Kroesen M, Bull C, Gielen PR, et al. Anti-GD2 mAb and Vorinostat synergize in the treatment of neuroblastoma. Oncoimmunology. 2016;5:e1164919. https://doi.org/10.1080/2162402X.2016.1164919.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Pinto N, DuBois SG, Marachelian A, et al. Phase I study of vorinostat in combination with isotretinoin in patients with refractory/recurrent neuroblastoma: a new approaches to Neuroblastoma Therapy (NANT) trial. Pediatr Blood Cancer. 2018;65:e27023. https://doi.org/10.1002/pbc.27023.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. DuBois SG, Granger MM, Groshen S, et al. Randomized Phase II Trial of MIBG Versus MIBG, Vincristine, and irinotecan versus mibg and vorinostat for patients with relapsed or refractory neuroblastoma: a report from NANT consortium. J Clin Oncol. 2021;39:3506–14. https://doi.org/10.1200/JCO.21.00703.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Relation T, Yi T, Guess AJ, et al. Intratumoral delivery of interferonγ-secreting mesenchymal stromal cells repolarizes tumor-associated macrophages and suppresses neuroblastoma proliferation in vivo. Stem Cells. 2018;36:915–24. https://doi.org/10.1002/stem.2801.

    Article  CAS  PubMed  Google Scholar 

  204. Miao L, Zhuo Z, Tang J, et al. FABP4 deactivates NF-κB-IL1α pathway by ubiquitinating ATPB in tumor-associated macrophages and promotes neuroblastoma progression. Clin Transl Med. 2021;11:e395. https://doi.org/10.1002/ctm2.395.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  205. Lum HD, Buhtoiarov IN, Schmidt BE, et al. In vivo CD40 ligation can induce T-cell-independent antitumor effects that involve macrophages. J Leukoc Biol. 2006;79:1181–92. https://doi.org/10.1189/jlb.0405191.

    Article  CAS  PubMed  Google Scholar 

  206. Buhtoiarov IN, Lum HD, Berke G, et al. Synergistic activation of macrophages via CD40 and TLR9 results in T cell independent antitumor effects. J Immunol. 2006;176:309–18. https://doi.org/10.4049/jimmunol.176.1.309.

    Article  CAS  PubMed  Google Scholar 

  207. Buhtoiarov IN, Sondel PM, Wigginton JM, et al. Anti-tumour synergy of cytotoxic chemotherapy and anti-CD40 plus CpG-ODN immunotherapy through repolarization of tumour-associated macrophages. Immunology. 2011;132:226–39. https://doi.org/10.1111/j.1365-2567.2010.03357.x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. Joshi S, Singh AR, Zulcic M, et al. Rac2 controls tumor growth, metastasis and M1–M2 macrophage differentiation in vivo. PLoS ONE. 2014;9:e95893. https://doi.org/10.1371/journal.pone.0095893.

    Article  PubMed  PubMed Central  Google Scholar 

  209. Wyce A, Ganji G, Smitheman KN, et al. BET inhibition silences expression of MYCN and BCL2 and induces cytotoxicity in neuroblastoma tumor models. PLoS ONE. 2013;8:e72967. https://doi.org/10.1371/journal.pone.0072967.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Puissant A, Frumm SM, Alexe G, et al. Targeting MYCN in neuroblastoma by BET bromodomain inhibition. Cancer Discov. 2013;3:308–23. https://doi.org/10.1158/2159-8290.CD-12-0418.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Andrews FH, Singh AR, Joshi S, et al. Dual-activity PI3K-BRD4 inhibitor for the orthogonal inhibition of MYC to block tumor growth and metastasis. Proc Natl Acad Sci U S A. 2017;114:E1072–80. https://doi.org/10.1073/pnas.1613091114.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  212. Shankar S, Srivastava RK. Histone deacetylase inhibitors: mechanisms and clinical significance in cancer: HDAC inhibitor-induced apoptosis. Adv Exp Med Biol. 2008;615(261–98):261–98. https://doi.org/10.1007/978-1-4020-6554-5_13.

    Article  CAS  PubMed  Google Scholar 

  213. 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;18:1036–44. https://doi.org/10.1158/1535-7163.MCT-18-1206.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  214. Mizukawa B, Wei J, Shrestha M, et al. Inhibition of Rac GTPase signaling and downstream prosurvival Bcl-2 proteins as combination targeted therapy in MLL-AF9 leukemia. Blood. 2011;118:5235–45. https://doi.org/10.1182/blood-2011-04-351817.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  215. Akasaki T, Koga H, Sumimoto H. Phosphoinositide 3-kinase-dependent and -independent activation of the small GTPase Rac2 in human neutrophils. J Biol Chem. 1999;274:18055–9. https://doi.org/10.1074/jbc.274.25.18055.

    Article  CAS  PubMed  Google Scholar 

  216. Ugolini A, Tyurin VA, Tyurina YY, et al. Polymorphonuclear myeloid-derived suppressor cells limit antigen cross-presentation by dendritic cells in cancer. JCI Insight. 2020;5(15):e138581.

    Article  PubMed  PubMed Central  Google Scholar 

  217. Veglia F, Tyurin VA, Blasi M, et al. Fatty acid transport protein 2 reprograms neutrophils in cancer. Nature. 2019;569:73–8. https://doi.org/10.1038/s41586-019-1118-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  218. Tang Y, Zhou C, Li Q, et al. Targeting depletion of myeloid-derived suppressor cells potentiates PD-L1 blockade efficacy in gastric and colon cancers. Oncoimmunology. 2022;11:2131084. https://doi.org/10.1080/2162402X.2022.2131084.

    Article  PubMed  PubMed Central  Google Scholar 

  219. Steggerda SM, Bennett MK, Chen J, et al. Inhibition of arginase by CB-1158 blocks myeloid cell-mediated immune suppression in the tumor microenvironment. J Immunother Cancer. 2017;5:101. https://doi.org/10.1186/s40425-017-0308-4.

    Article  PubMed  PubMed Central  Google Scholar 

  220. Barry ST, Gabrilovich DI, Sansom OJ, et al. Therapeutic targeting of tumour myeloid cells. Nat Rev Cancer. 2023;23:216–37. https://doi.org/10.1038/s41568-022-00546-2.

    Article  CAS  PubMed  Google Scholar 

  221. Chaib M, Chauhan SC, Makowski L. Friend or foe? recent strategies to target myeloid cells in cancer. Front Cell Dev Biol. 2020;8:351. https://doi.org/10.3389/fcell.2020.00351.

    Article  PubMed  PubMed Central  Google Scholar 

  222. Wang Y, Johnson KCC, Gatti-Mays ME, Li Z. Emerging strategies in targeting tumor-resident myeloid cells for cancer immunotherapy. J Hematol Oncol. 2022;15:118. https://doi.org/10.1186/s13045-022-01335-y.

    Article  PubMed  PubMed Central  Google Scholar 

  223. Ozkaynak MF, Sondel PM, Krailo MD, et al. Phase I study of chimeric human/murine anti-ganglioside G(D2) monoclonal antibody (ch14.18) with granulocyte-macrophage colony-stimulating factor in children with neuroblastoma immediately after hematopoietic stem-cell transplantation: a Children’s Cancer. J Clin Oncol. 2000;18:4077–85. https://doi.org/10.1200/JCO.2000.18.24.4077.

    Article  CAS  PubMed  Google Scholar 

  224. Orrantia A, Terrén I, Astarloa-Pando G, et al. NK Cell reconstitution after autologous hematopoietic stem cell transplantation: association between nk cell maturation stage and outcome in multiple myeloma. Front Immunol. 2021;12:748207.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  225. Orrantia A, Terrén I, Astarloa-Pando G, et al. Human NK Cells in autologous hematopoietic stem cell transplantation for cancer treatment. Cancers (Basel). 2021;13:1589. https://doi.org/10.3390/cancers13071589.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This study was funded by TKI-Health Holland (TumMyTOF) (to Jeanette Leusen).

Author information

Authors and Affiliations

Authors

Contributions

MS initiated this project and wrote the manuscript. LT conducted literature research and wrote the first draft of the manuscript. MD provided clinical feedback and was involved in reviewing and editing the manuscript. JL provided immunological and biological feedback and was involved in reviewing and editing the manuscript. DK wrote the manuscript and was responsible for its execution. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Daniëlle Krijgsman.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

Jeanette Leusen is scientific founder and shareholder of TigaTx.

Additional information

Publisher’s Note

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

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Stip, M.C., Teeuwen, L., Dierselhuis, M.P. et al. Targeting the myeloid microenvironment in neuroblastoma. J Exp Clin Cancer Res 42, 337 (2023). https://doi.org/10.1186/s13046-023-02913-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13046-023-02913-9

Keywords