Skip to main content

Microenvironmental regulation of tumor-associated neutrophils in malignant glioma: from mechanism to therapy

Abstract

Glioma is the most common primary intracranial tumor in adults, with high incidence, recurrence, and mortality rates. Tumor-associated neutrophils (TANs) are essential components of the tumor microenvironment (TME) in glioma and play a crucial role in glioma cell proliferation, invasion and proneural-mesenchymal transition. Besides the interactions between TANs and tumor cells, the multi-dimensional crosstalk between TANs and other components within TME have been reported to participate in glioma progression. More importantly, several therapies targeting TANs have been developed and relevant preclinical and clinical studies have been conducted in cancer therapy. In this review, we introduce the origin of TANs and the functions of TANs in malignant behaviors of glioma, highlighting the microenvironmental regulation of TANs. Moreover, we focus on summarizing the TANs-targeted methods in cancer therapy, aiming to provide insights into the mechanisms and therapeutic opportunities of TANs in the malignant glioma microenvironment.

Introduction

Glioma is the most common primary intracranial tumor in adults, with high incidence, recurrence, and mortality rates [1]. Overall, the broad category of gliomas (ICD-O-3 histology codes 9380–9384 and 9391–9460) represents approximately 24.5% of all primary brain tumors and 80.9% of all malignant tumors in adults [2]. Glioblastoma (GB), classified as grade 4, adult-type diffuse gliomas, represents the most common malignant brain tumor in adults with a 5-year survival rate of only 6.8% [2,3,4]. Current treatments for newly diagnosed patients suggested by newest guidelines mainly involve maximum safe surgical resection, followed by radiotherapy (RT) and radiochemotherapy combined with temozolomide (TMZ), as well as other combination treatments such as adjuvant chemotherapy and Tumor Treating Fields [4, 5]. Over the past decade, treatment strategies for glioma have continued to evolve. However, due to the highly invasive and drug resistance of glioma, the overall survival (OS) of glioma patients has not significantly improved, novel therapeutic targets and precision treatment strategies are urgently needed [6].

The tumor microenvironment (TME) is a key factor in tumor progression and has been one of the research hotspots in recent years. It is generally believed that the TME plays a crucial role in various malignant behaviors such as proliferation, invasion, angiogenesis, and drug resistance. In glioma TME, non-tumor cells mainly include tumor-associated macrophages (TAMs), dendritic cells (DCs), tumor-associated neutrophils (TANs), lymphocyte, astrocyte, endothelial cells, and others [7]. Previous studies focused more on the role of TAMs and other cells in the glioma microenvironment, the function of TANs was not appreciated for a long time because it was not as abundant as TAMs. With the development of bioinformatic analysis tools and single-cell transcriptome sequencing technology in recent years, the role of TANs in the progression of various tumors, including glioma, has been gradually discovered and attracted attention [8, 9].

Neutrophils are the most abundant myeloid cells in the circulation of humans and mice, with a ratio to monocytes of approximately 7:1 to 10:1 [10]. During the pathophysiological process of tumors, neutrophils proliferate, extravasate, and enter the TME from the peripheral blood through specific mechanisms, becoming TANs. Studies over the past decade have shown that TANs are not merely inert “bystanders” as traditionally believed, but rather important components that influence tumor progression and exhibit both pro-tumor and anti-tumor effects [11,12,13,14,15,16]. Histologically, it has been confirmed that neutrophils infiltrate more in malignant glioma [17]. Substances produced by TANs, such as matrix metalloproteinase 9 (MMP9), promote angiogenesis in glioma [18]. Furthermore, the role of TANs in resisting anti-angiogenic therapy and immune checkpoint therapy has been confirmed [19]. Conversely, other studies have shown that TANs also play an anti-tumor role, such as direct or indirect cytotoxic effects [20, 21]. The functions of TANs in glioma microenvironment are context-dependent and still need deep investigation.

In this review, we mainly discuss how TANs participate in the malignant behavior of glioma, including the origin of TANs, the impacts of TANs on glioma cells, and a particular focus on the microenvironmental regulation of TANs. We also summarize the research progress of TANs-targeted therapies in glioma treatment and novel neutrophil-based drug delivery systems in cancer therapy, with an attempt to highlight the important roles of TANs in glioma microenvironments and tumor progression, which finally shed light on TANs-targeted glioma therapy.

From neutrophils to tumor-associated neutrophils

Neutrophils develop and mature in the bone marrow

Neutrophils, recognized as crucial participants in acute inflammation, are a subset of polymorphonuclear leukocytes. They are typically the first leukocytes to be recruited to inflammatory sites, including tumors, exerting a significant impact on tumor progression [11, 22]. Neutrophils continuously originate from myeloid stem cells in the bone marrow. Granulocyte colony-stimulating factor (G-CSF) plays a pivotal role in promoting this process. During maturation, neutrophils go through the following stages: myeloblast, promyelocyte, myelocyte, metamyelocyte, band granulocyte, and eventually become mature polymorphonuclear/segmented granulocyte [22]. Mature neutrophils express G-CSF receptor (G-CSF-R), Toll-like receptors (TLRs), and CXC chemokine receptor 2 (CXCR2) on their surfaces. These receptors are closely associated with the egress of neutrophils from the bone marrow [22] (Fig. 1).

Fig. 1
figure 1

Neutrophil maturation and recruitment Neutrophils develop and mature in the bone marrow. Mature neutrophils travel out of the bone marrow into the peripheral blood in the presence of G-CSF, GM-CSF, TLR ligands and CXCLs. Subsequently, cytokines such as CXCLs, IL-1β and other cytokines recruit neutrophils into the TME. Neutrophils enter the TME and become TANs and differentiate into N1 and N2 types in response to IFN-γ, TGF-β signaling. New TANs classifications identified by dcTRAIL-R1 and CD101 statuses

Neutrophils are recruited to the TME and designated as TANs

In patients with solid tumors, mature neutrophils in the organism are classified into two major categories based on their localization—peripheral blood neutrophils (PBNs) and TANs infiltrating the tumor tissue [14]. The neutrophil-to-lymphocyte ratio (NLR), the ratio of neutrophil count to lymphocyte count in peripheral blood, is a biomarker linking the neutrophil-dominated innate immune response to the lymphocyte-supported adaptive immunity [23]. NLR can to some extent reflect the status of TME, serving as a commonly used indicator for clinical efficacy assessment and prognosis evaluation [24]. For example, patients with high NLR values are diagnosed with high-grade glioma and associated with poor OS [24].

Under the influence of several molecules (see below), neutrophils move out from the blood and are recruited into the TME. When neutrophils traffic into TME, they are referred to as TANs [14]. Studies indicate that CXCR2 is a critical molecular mediator of neutrophil recruitment. CXCR2, expressed on the surface of neutrophils, is functionally associated with mediating neutrophil activation [25, 26]. The level of CXCR2 is closely correlated with the prognosis of glioma. Yang et al. have demonstrated that elevated CXCR2 levels indicate a significantly higher recurrence and metastasis rate in glioma patients, serving as an adverse prognostic indicator [27]. Glioma cells produce G-CSF and granulocyte-macrophage colony-stimulating factor (GM-CSF), promoting the egress of neutrophils from the bone marrow and prolonging their lifespan [16, 28, 29]. This process is also closely associated with CXCR2 and its ligands CXC motif ligands (CXCLs) [29]. CXCR2 ligands include CXCL1, CXCL2, CXCL5, CXCL6, CXCL8 (Interleukin-8, IL-8), with IL-8 playing a crucial role in the recruitment of neutrophils to the microenvironment of glioma. Various components in the TME, such as IL-1β, TNF-α, can stimulate GB cells to produce IL-8, recruiting neutrophils (Fig. 2A) [30,31,32]. Fas in the TME, when bound to FasL on the surface of glioma cells, activates downstream signaling pathways, promoting the expression of IL-8 and recruiting neutrophils (Fig. 2B) [33]. Additionally, neutrophil extracellular traps (NETs) can promote the secretion of IL-8 by glioma cells through the HMGB1/RAGE/NF-κB axis, recruiting neutrophils [34]. These neutrophils, in turn, mediate the formation of NETs through the PI3K/AKT/ROS axis, creating a positive feedback loop (Fig. 2C) [34]. It has also been found that IL-8/CXCL1 interaction likewise plays an important role in TANs recruitment in the zebrafish glioma model [35]. Apart from IL-8, IL-1β is also an essential cytokine for promoting neutrophil recruitment. Wang et al. have suggested that the binding of LINC01116 to DDX5 promotes the expression of IL-1β in glioma cells, facilitating the recruitment of neutrophils (Fig. 2D) [36]. Lee et al. have indicated that ectopic expression of CD133 induces an increase in IL-1β expression, which promotes neutrophil recruitment (Fig. 2E) [37].

Fig. 2
figure 2

Recruitment of TANs IL-8 and IL-1β can recruit neutrophils into TME. (A) IL-1β, TNF-α in TME improve IL-8 production in glioma cells. (B) FasL in TME improves IL-8 production in glioma cells. (C) Positive feedback loop in TANs and glioma cells. NETs produce HMGB1 and promote IL-8 secretion in glioma cells by HMGB1/RAGE/NF-κB axis. IL-8 binds to CXCR2 on TANs promoting NETs formation by PI3K/AKT/ROS axis. (D) LINC01116 combined with DDX5 promote IL-1β secretion in glioma cells. (E) Ectopic expression of CD133 in glioma stem cells stimulates glioma cells secreting IL-1β

TANs are “educated” to play dual roles in TME

Numerous studies indicate that TANs exhibit both pro- and anti-tumor effects. In a considerable portion of research, like the differentiation of TAMs into M1-like and M2-like phenotypes, TANs are also described as N1-type (with anti-tumor effects) and N2-type (with pro-tumor effects) [11, 38, 39]. Several molecules have been identified to distinguish between N1 and N2. N1 markers include intercellular cell adhesion molecule-1 (ICAM-1), inducible nitric oxide synthase (iNOS), C-C motif ligand 3 (CCL3), tumor necrosis factor-α (TNF-α), etc.; N2 markers include CCL17, CCL2, arginine (Arg), CCL5, vascular endothelial growth factor (VEGF), etc [35]. What is clearer is that transforming growth factor-β (TGF-β) signaling in the TME tends to drive neutrophils towards a pro-tumor phenotype (N2), while type I interferon (IFN) signaling or inhibition of TGF-β signaling can drive neutrophils towards an anti-tumor phenotype (N1) [38]. In addition, N1 and N2-type TANs may also vary with TME: in early tumor stages, TANs exhibit stronger cytotoxic effects on tumor cells, whereas in advanced tumor stages, TANs exhibit more of a pro-tumor phenotype [40, 41]. (Fig. 1)

Some scRNAseq data suggest that N1/N2 may not be the so-called ‘polarization’ in different directions, but rather that TANs exhibit different effects on tumor cells at different degrees of activation. Using scRNAseq technology, they identified TANs as different from N1/N2 in multiple differentiation states. Wu and colleagues identified TANs as 10 distinct transcriptional states, including inflammation, angiogenesis, and antigen presentation, from 225 samples from 17 cancer types [8]. A newly published article in Science presents a novel perspective on TANs recruitment and reprogramming [42]. In pancreatic tumor model, researchers classified infiltrating neutrophils into three different neutrophil states, T1, T2 and T3 [42]. Their states can be determined by dcTRAIL-R1 and CD101, i.e., T1 (dcTRAIL-R1CD101), T2 (dcTRAIL-R1CD101+), and T3 (dcTRAIL-R1+) [42]. Of these, T1 and T2 neutrophils are transitional neutrophil populations, differentiated from immature and mature neutrophils, respectively, while T3 neutrophils are in a terminally differentiated state, formed by further reprogramming of T1 and T2 neutrophils [42]. This process is irreversible [42]. In addition, the researchers further found that TME stimuli are necessary for the upregulation of dcTRAIL-R1 i.e., the shift to T3 neutrophils, but this process does not require the intervention of any tumor-soluble factors [42]. This finding suggests a defined program within neutrophils that enables them to acquire the T3 phenotype independent of their maturation stage [42]. At the functional level, T3 neutrophils are located in hypoxic niche and highly express VEGFA, promoting angiogenesis and tumor growth [42]. (Fig. 1)

TANs involving in malignant behaviors of tumor

TANs promote tumor invasion

Several studies across different tumors have confirmed the correlation of TANs with the invasiveness, malignancy, and adverse prognosis of tumors, and that neutrophil depletion therapy contributes to promoting survival [43]. The invasion of tumor cells into normal brain tissue is a characteristic of malignant glioma [44]. Isocitrate dehydrogenase (IDH), a key enzyme in the Krebs cycle, catalyzes the oxidative decarboxylation of isocitrate. IDH has two isoforms, IDH1 and IDH2, with most mutations involving the substitution of arginine at position 132 by histidine in IDH1 (R132H) and arginine at position 172 by lysine in IDH2 (R172K) [45]. Based on the status of IDH, glioma can be classified into wild-type IDH (wt-IDH) and mutant IDH (mu-IDH) [45]. Compared with wt-IDH, mu-IDH exhibits weaker invasiveness and higher rates of progression-free survival (PFS) [45, 46]. Amankulor et al. compared the immune cell composition in wt-IDH1 and mu-IDH1 glioma and found that mu-IDH1 glioma had fewer immune cell infiltration, especially neutrophils [17]. Also, they noted a lower presence of neutrophil chemokines in the homogenates of mu-IDH1 glioma [17]. Further, neutrophil depletion using an anti-Ly6g antibody had a significant survival benefit in wt-IDH1 glioma mice and no significant effect in mu-IDH1 glioma mice [17, 47].

The awakening of dormant tumor cells is a common cause of tumor invasion, metastasis, and recurrence. In experimental tumor models, NETs formed during the inflammation can awaken tumor cells [48]. NETs, generated by neutrophils, are extracellular network structures constructed from DNA fiber (nuclear and/or mitochondrial DNA) and modified with histones and enzymes [48, 49]. Neutrophil elastase (NE) and MMP9 are pivotal among these. Using a model of lung cancer caused by chronic inflammation, Albrengues et al. have found that NETs DNA connects to the extracellular matrix (ECM), transports NE and MMP9 released by neutrophils to the vicinity of the substrate, facilitating the breakdown and reorganization of laminin [50]. This recombined laminin awakens dormant cancer cells via integrin activation and the FAK/ERK/MLCK/YAP signaling pathway, promoting aggressive tumor growth and metastasis [50]. Studies on various intracranial tumors similarly support the promoting role of these two enzymes in tumor invasion. NE is absent in normal brain tissue but is present in the border zones of high-grade glioma [51, 52]. Compared to low-grade glioma, the borders of GB exhibit more NE, correlating with stronger invasiveness [51]. A study including samples from 62 glioma patients (surgical or biopsy) has suggested that higher levels of MMP9 in glioma mean stronger invasiveness, higher grades, and poorer prognosis [53]. Additionally, MMP9 promotes the proliferation of glioma cells [53]. (Fig. 3A)

TANs promote tumor proliferation

TANs have been shown to directly promote tumor cell proliferation. Researchers co-cultured U87MG glioma cells with human neutrophils and observed neutrophil promotion of glioma cell proliferation in both 2D and 3D co-culture experiments [54]. Interestingly, soluble factors mediating glioma-neutrophil communication vary over time. After 24 h and 72 h of co-cultivation, the production of IL-8 in glioma-neutrophil cultures increased significantly and decreased sharply after 120 h. The production pattern of TNF-α is similar to IL-8. The opposite was observed for IL-1β. It was virtually unproduced in the first few hours, but its release rose after 120 h [54]. Correspondingly, glioma cells were attacked by neutrophils in the first 24 h of co-cultivation, and after 120 h, tumors responded to the presence of neutrophils with increased viability [54]. The crosstalk between neutrophils and glioma cells potentiates the differential release of immune cell recruiting and suppression molecules promoting tumor progression [54]. One study based on zebrafish glioblastoma model indicated that neutrophils are recruited in early stage during oncogenesis and increases tumor cell proliferation [35]. The possible explanation is that ROS released by TANs can cause DNA damage and induce tumorigenesis in peri-tumor cells [35, 54]. Both lung adenocarcinoma model and non-small cell lung cancer (NSCLC) model confirm that TANs produce NE, which promotes tumor cell proliferation by upregulating the PDGFR/PI3K pathway and PGE2 synthesis [55, 56]. CCK8 assay demonstrated that NETs significantly contributed to the proliferation rate of glioma cells, which was reduced by DNase I, hydrolase can degrade NETs [34]. Although NE is an important component of NETs, it is not clear whether the role of NETs in promoting tumor proliferation is wholly or partially dependent on NE. TANs also promote tumor proliferation indirectly through the secretion of soluble factors. Bone morphogenetic protein 2 (BMP2) and TGF-β2 secreted by TANs promote the expression of miR-301b-3p in hepatocellular carcinoma (HCC), which has been shown to promote hepatocellular carcinoma (HCC) proliferation and stem cell characteristics [57]. Meanwhile, miR-301b-3p can suppress CYLD expression in HCC cells, consequently leads to hyperactive NF-κB signaling, higher level of CXCL5 secretion, and more TANs recruitment, forming a positive feedback loop [57]. (Fig. 3B)

Many studies focus on the role of ferroptosis of tumor cells on tumor invasion and proliferation, but contradictions still exist. TANs are closely associated with ferroptosis of tumor cells. TANs transfer myeloperoxidase (MPO)-containing particles into GB cells, and MPO mediates ferroptosis of GB cells by increasing lipid-based ROS, clarifying the mechanism of necrosis in GB cells [58]. In triple-negative breast cancer (TNBC), TANs produce NETs, which mediate ferroptosis resistance of TNBC cells through the TLR9/Merlin signaling axis and promote TNBC proliferation [59]. Both TANs-mediated ferroptosis resistance and ferroptosis of tumor cells enhance the invasiveness of tumor and suggest a poor prognosis [58, 59]. (Fig. 3B)

TANs promote tumor angiogenesis

Like normal organs, tumor tissues obtain oxygen and nutrients through blood vessels, which is mainly achieved through angiogenesis. As a critical manifestation of glioma progression, angiogenesis mainly involves the following steps: (1) vascular rupture, (2) degradation of the vascular basement membrane and surrounding extracellular matrix (ECM), and (3) migration of vascular endothelial cells (ECs) and new blood vessel formation [60]. TANs predominantly influence steps (2) and (3). Various evidence indicates that neutrophils play a significant role in angiogenesis under normal physiological conditions [61]. In the context of cancer, TANs serve as a significant source of pro-angiogenic factors in the TME, no less than TAMs [18]. TANs promote tumor angiogenesis mainly through interaction with ECs.

VEGF/VEGFR interactions, one of the most classical pathways in ECs, activate downstream pathways including PLC/ERK pathway, PI3K/AKT/mTOR pathway, small GTPases, and others [62]. They are widely involved in vascular development, cell survival, regulation of vasomotion, cell shape, cell migration and polarization, and many other processes [62]. TANs can produce large amounts of VEGFA, which stimulates ECs and promotes tumor angiogenesis through the VEGFA/VEGFR2 pathway [18]. In addition, since a large amount of VEGF is sequestered in the ECM in an inactivated form, MMP9, produced by TANs, degrades ECM and releases a large amount of VEGF, which effects on the nearby ECs and promotes tumor angiogenesis [18, 61]. The activity of MMP9 is inhibited by tissue inhibitors of metalloproteinase-1 (TIMP-1). This inhibitor is often released in conjunction of MMP9 to form an MMP9/TIMP-1 complex thereby preventing its protein hydrolysis activity [61]. However, TANs do not express tissue inhibitors of metalloproteinase-1 (TIMP-1), which forms an MMP-9/TIMP-1 complex with MMP9 to inhibit its protein hydrolysis activity, thus requiring only a small amount of TANs to produce significant pro-tumor angiogenic effects [61, 63]. In addition, IL-8 secreted by tumor cells recruits TANs and facilitates the rapid release of MMP9 from TANs. Meanwhile, MMP9 can potentiate IL-8 via amino-terminal processing, resulting in a 10-fold higher potency in TANs activation, consequently promoting MMP9 release and IL-8 mediated recruitment of TANs. This creates a positive feedback loop, recruiting more TANs and promoting more angiogenesis [61, 64,65,66]. (Fig. 3C)

Bv8 (also known as prokineticin 2) is important in promoting tumor angiogenesis [67]. As a homolog of endocrine gland-derived VEGF (EG-VEGF), Bv8 plays the role of mitogen or survival factor of ECs in angiogenesis [68]. Studies have shown that G-CSF activates the JAK signaling and STAT3 pathways, promoting neutrophil proliferation [18]. Meanwhile, G-CSF induces the upregulation of Bv8 expression on neutrophils in a STAT3-dependent manner [69, 70]. Bv8 can activate STAT3 in a JAK2-dependent manner, establishing a positive feedback loop that generates a large amount of Bv8 in the TME and promotes tumor angiogenesis [69, 70]. Blocking Bv8, in turn, can inhibit glioma angiogenesis [71]. Unlike VEGFR (a kind of receptor tyrosine kinases, RTKs), the receptors for Bv8 and EG-VEGF belong to the GPCR family and are not drug targets for tyrosine kinase inhibitors (TKIs) [68, 69]. This may also be one of the reasons why tumors get TKI therapy-resistant. (Fig. 3C)

Recent studies have found that TANs produce Osteopontin (OPN) and MMP14promoting EC chemotaxis and accelerating vascular network formation, respectively [72]. OPN maintains EC survival through activation of the PI3K-AKT pathway, followed by Bcl-xL upregulation and NF-κB activation [72, 73]. Meanwhile, OPN also enhances VEGF expression through the AKT and ERK pathways, which in turn acts as a positive feedback signal activating PI3K/AKT and ERK pathways, enhancing angiogenesis directly [73]. The pro-angiogenic effect of MMP14 is associated with VEGFR1 [74]. In ECs, VEGFR1 is considered as a negative regulator of VEGFR2 [62]. Compared to VEGFA/VEGFR2 interaction, VEGFA has a stronger affinity for VEGFR1 but a weaker ability to induce VEGFR1 phosphorylation [62]. MMP14 can bind selectively and then degrade VEGFR1 on the surface of ECs, thereby enhancing the sensitivity of VEGFR2 to VEGFA, resulting a strong proliferation effect on ECs at low concentrations of VEGFA [74]. (Fig. 3C)

In addition to their pro-angiogenic role, ECs can also affect neutrophil adhesion in TME. Co-culture experiments showed that crosstalk between neuroblastoma cells and ECs downregulates adhesion molecule CD44R expression impairing neutrophil adhesion and invasion, blocking neutrophil recruitment to ECs [75]. Researchers suggested that this might be a tumor escape strategy [75]. Interestingly, the addition of isolated tumor cell membrane fragments to ECs rather than soluble cell culture supernatant caused this effect, suggesting that the interaction of ECs with tumor cells is cell-cell direct contact [75]. This process may be related to the PKC signaling pathway, and blocking PKC can prevent the reduction in neutrophil binding capacity [75]. (Fig. 3C)

TANs promote PN-MES transition

GB has been classified into 4 molecular subtypes—Proneural (PN), Mesenchymal (MES), Classical (CL), and Neural (NL). The NL type has been recognized as a contamination of specimens by normal neurons and has been excluded from the most recent classifications [76,77,78]. Compared to the PN-type, MES-type GB is more aggressive and has a worse prognosis [79]. Presently, most studies have suggested that the critical signaling pathways driving the PN-MES transition (PMT) include STAT3, JAK, and NF-κB pathways, involving various activated molecules such as CXCLs, growth factors, and neurotransmitters [77, 79]. The prevailing view is that the crosstalk between TAMs and GB cells plays a pivotal role in PMT [77, 79, 80]. However, some recent evidence has suggested that TANs also play an indispensable and crucial role in PMT.

Dysregulated expression of the S100 protein family is a common feature in human cancers [81]. S100A4, an important member of the S100 protein family, plays a significant role in tumor progression, metastasis, and mesenchymal transition [82]. Liang et al. have indicated that MES-type GB significantly overexpresses S100A4, whereas the PN type downregulates S100A4 expression [83]. Meanwhile, consistent with the finding that neutrophils can increase S100A4 expression in vitro, S100A4 was significantly co-expressed with CD64 in samples from GB patients, which collectively suggests that TANs promote PMT in GB by facilitating the expression of S100A4 [83]. Chow et al. further identified S100A4 as an upstream regulator of other PMT modulators, such as SNAIL2 and ZEB, which have been explicitly identified to promote the invasion and mesenchymal transition of GB [84]. (Fig. 3D)

Another strong evidence comes from Chen et al. They used CRISPR/Cas9 technology to knock out monocyte chemoattractant protein (MCP) to abolish TME recruitment to monocytes [85]. In a platelet-derived growth factor B (PDGFB)-driven GB model, monocyte depletion was accompanied by a massive influx of neutrophils into the TME and elevation of the neutrophil chemokines CXCL1 and CXCL2. In contrast, these phenomena were not present in the Nf1-silenced GB model [85]. Further experiments demonstrated that neutrophil-derived TNF-α directly drives the mesenchymal transition of PDGFB-driven primary GB cells [85]. (Fig. 3D)

Fig. 3
figure 3

TANs participate in malignant behaviors of glioma cells (A) NETs assist NE and MMP9 in the catabolism of Laminin, which ultimately promotes the arousal of dormant glioma cells and promotes glioma invasion. (B) TANs produce mtROS, NE, BMP2, TGF-β2, regulate ferroptosis resistance or ferroptosis of tumor cells promoting tumor proliferation. (C) TANs secrete BV8, IL-8, VEGFA, MMP14 and OPN, promoting tumor angiogenesis via influencing EC. Tumor cell-EC interaction reduces neutrophil recruitment. (D) TANs promote PMT of glioma

Interaction of TANs with TME niches and components

Hypoxia

Hypoxia is a common niche in solid tumors and is no exception in glioma. The hypoxia-inducible factor (HIF) family, as crucial transcription factors, become activated under hypoxic conditions, regulating various hypoxia-responsive genes to support cell survival in hypoxic environments [86,87,88]. Other transcription factors such as NF-κB also play vital roles and crosstalk with HIF [89]. Studies have found that in highly heterogeneous tumors, low oxygen signaling, and hypoxia-related genes are more prevalent, correlating with poorer prognoses [90]. Therefore, elucidating the role of the hypoxic niche in tumor progression is both important and necessary.

Hypoxia typically arises due to rapid cancer cell proliferation and the lack of sufficient oxygen-supplying blood vessels [91]. Correspondingly, hypoxia can induce angiogenesis through various signaling pathways, including directly inducing angiogenic factors within tumor cells and indirectly recruiting pro-angiogenic cells [86]. Studies have suggested that hypoxia can induce tumor cells to produce IL-8, one of the key cytokines involved in TANs recruitment [92]. It has also been found that hypoxia induces tumor cells to produce many extracellular vesicles (EVs), which transport amounts of IL-8 and miR-451 [93]. Although it has not been described that hypoxia-derived EV directly affects neutrophil function, the EVs intrinsic IL-8 and miR-451 have been described to play opposite roles in neutrophil recruitment [93]. As mentioned above, IL-8/CXCR1/2 signaling promotes neutrophil recruitment to the TME. Moreover, IL-8 acts as a neutrophil activator that activates the MAPK signaling pathway, inhibits neutrophil apoptosis and enhances neutrophil proliferation [94]. On the contrary, miR-451 inhibits the phosphorylation of p38 MAPK and suppresses neutrophil chemotaxis [95]. Further studies on the specific effects of EVs are needed. Triner et al., in addition, have found that the CXCL1 gene is upregulated in colon cancer in a HIF2α-dependent manner, recruiting TANs through the CXCL1/CXCR2 signaling to promote colorectal cancer progression [96]. Recent studies have discovered that GB, under hypoxic conditions, can produce acrolein, inhibiting neutrophil activation and promoting N2 TANs phenotype [97]. Acrolein can produce TGF-β-like effects, promoting neutrophil expression of Arg1, inhibiting MPO expression, superoxide production and ROS generation, inhibiting tumor-killing activity, and inducing N2 phenotype polarization [97]. Dimercaprol, which is a kind of acrolein scavenger, can inhibit tumor growth significantly [97]. Further studies have shown that acrolein modifies the Cys310 residue in AKT, which is a key residue in the catalytic structural domain of AKT [97]. This modification by acrolein inhibits AKT phosphorylation, blocks the downstream signaling pathway of PI3K/AKT and inhibits neutrophil activation [97]. Overall, the interaction between hypoxia and neutrophils results in pro-tumor genesis. (Fig. 4A).

TAMs

Concerning brain tumors, the concept of TAMs is generally broadened into two cell types: circulating monocyte/macrophage and resident microglia within the central nervous system [98, 99]. TAMs represent most immune cells within the brain tumor microenvironment, constituting up to 30% of the tumor mass [7]. This process may be related to the PKC signaling pathway, and blocking PKC can prevent the reduction in neutrophil binding capacity [100]. The conventional view is that TAMs exist in two polarized states, M1-like (classically activated), which is usually anti-tumor, and M2-like (alternatively activated), which is usually pro-tumor [99]. A novel view, on the other hand, suggests that the M1/M2-like classification inadequately reflects the actual states of TAMs in vivo, thus classifying them into immunosuppressive cells and proliferative macrophages by molecular subtype and functional state [101]. This can help us better understand the role of TAMs in TME.

The interaction between TANs and TAMs has been established in various solid tumors. It promotes oncostatin M and IL-11 associated tumor progression in intrahepatic cholangiocarcinoma (ICC) and post-treatment progression in NSCLC [102, 103]. Researchers have also identified antitumor outcomes resulting from the TANs-TAMs interaction. In dermal sarcoma models, TANs promote TAMs to produce IL-12, subsequently activating the IFN-γ pathway in αβT cells, generating an antitumor effect [104]. But unfortunately, in glioma, studies on TANs-TAMs interactions remain largely unexplored. Researchers tend to focus more on the interactions of TANs or TAMs, separately, with glioma cells and other cells, even though there is overlap in the cytokines expressed or received by TANs and TAMs (such as CCR2, TGF-β, IFN-γ, VEGF, GM-CSF, etc.) [98, 99, 105]. It has been shown that OPN produced by glioma stem cells recruits both TANs and TAMs to glioma TME [106]. It has also been suggested that TANs can promote biopsy-induced glioma cell metastasis by recruiting TAMs [28]. Additionally, an interesting and yet unresolved issue is that, apart from the MES subtype of GBM, TANs recruitment in glioma appears to be repulsed by TAMs. This pattern differs from what is observed in infections or other tumors, where monocytes/macrophages produce neutrophil chemokines to recruit neutrophils [10]. In short, the TANs-TAMs interaction in glioma is a worth exploring research direction. (Fig. 4B)

T-cell

The dysfunction of T-cell is the most important mechanism behind the immune suppression in glioma [99]. TANs produce a high level of ROS that can enter T-cell, damaging telomeres in DNA, accelerating T-cell aging, consequently leading to T-cell dysfunction [107]. Arginine is a crucial factor for sustaining T-cell activation, and Arginase 1 (Arg1) released by TANs extracellularly significantly depletes extracellular L-arginine, causing T-cell dysfunction [108]. This mechanism often results in systemic T-cell suppression in GB patients [108].

The interaction between the programmed cell death receptor-1 (PD-1) and its ligand (PD-L1) represents the classical T-cell immune checkpoint that acts as a “brake” on immune function [109]. In glioma, this pathway serves as a significant mechanism causing T-cell suppression [110]. Sequential IF studies have shown that CD4+ T helper cell, Treg, and CD8+ T-cell are closer to TANs than to other TME cells [111]. Meanwhile, PD-L1+ TANs were closest to PD-1+CD8+ T-cell and the most abundant PD-L1+ cell type within the PD-1+CD8+ T-cell niche was TANs [111]. This stands as critical evidence of TANs-mediated immune suppression in glioma TME. Furthermore, TANs can induce PD-1 expression on CD4+ effector memory T-cell (TEM) within the GB microenvironment. Simultaneously, TANs exhibit high levels of PD-L1, depleting CD4+ TEM function through the PD-1/PD-L1 axis, promoting immune suppression [112]. In a certain GB model with defective type I IFN receptor, tumor-infiltrating CD11b+Ly6G+ cell increase, while CD4+FoxP3+ cell (immunosuppressive Treg) increase and CD8+ cytotoxic T cell (CTL) decrease [113]. Ferroptosis also plays an important role in T-cell related immunosuppression. Ferroptosis is a non-apoptotic, regulated cell death triggered by dysregulation of redox mechanisms. New studies have shown that TANs are highly susceptible to ferroptosis and even spontaneously undergoes ferroptosis [114]. Meanwhile, ferroptosis accounts for most of the death program in TANs [114]. Ferroptosis in TANs can generate a large amount of PGE2 and oxidized phosphatidylethanolamine (ox-PE), which collectively inhibit CD8 + T cell activity, producing immunosuppression [114] (Fig. 4C).

Interestingly, the interaction between TAN and T-cell also exhibits some antitumor effects. In lung cancer, TANs with an antigen-presenting cell (APC)-like phenotype trigger antitumor T-cell responses and support anticancer activity [21]. Lad et al. have reported a specific population of TANs with DC characteristics [20]. These particular TANs, originating from the local cranial bone marrow, accumulate in the GB microenvironment, can activate T-cell cytotoxicity and memory, inhibiting tumor growth in vivo [20]. Recent findings provide strong support for this anti-tumor effect and have made great strides. Wu et al. analyzed the transcriptional profile of TANs and classified them into 10 different states, of which HLA-DR+ TANs express MHC class II molecules and exhibit antigen-presenting properties [8]. Co-culture experiments have shown that HLA-DR+ TANs can present tumor neoantigens and activate T-cell directly through ligand-receptor interactions, then stimulate T-cell to express TNFα and generate reactive T-cell responses [8]. According to metabolic analysis, leucine catabolism leads to high HLA-DR expression and initiates antigen presentation mechanisms [8]. Leucine diet combined with anti-PD-1 therapy significantly reduces tumor volume and stabilizes the disease [8]. (Fig. 4C)

Fig. 4
figure 4

Crosstalk between TANs and glioma microenvironment niches and components (A) Hypoxia stimulates glioma cells to produce IL-8, CXCL1 and Acrolein, promoting neutrophil recruitment and N2-type transformation. (B) TANs and TAMs interaction upregulate STAT3 signaling in tumor cells resulting in protumor effect; CCL3+ TANs and SPP1+ TAMs interaction promotes post-therapy tumor progression; TANs promote TAMs IL-12 secretion activating IFN-γ signaling in αβ T-cell, leading to antitumor effect. (C) TANs led to immunosuppression via T-cell dysfunction, ICB and ferroptosis; DC-like TANs and HLA-DR+ TANs exert an antitumor effect

TANs promote glioma therapy resistance

TANs promote RT resistance in glioma

As the infiltration of tumor cells extends far beyond the lesion, postoperative RT is an important part of glioma treatment, helping to preserve patient function and improve survival [5]. Several studies have demonstrated that TANs are involved in promoting glioma RT resistance [115]. Radiation-induced senescent GB cells recruit TANs and promote angiogenesis via NF-κB signaling [115, 116]. Meanwhile, TANs support GB to glioma stem cells transformation through NOS2-NO-ID4 signaling [115, 116]. In addition, TANs secrete S100A4, which promotes glioma stem cells proliferation and stemness and facilitates RT resistance formation [83, 115]. In lung cancer model, researchers have found that lung cancer cells upregulate GLUT1 expression in TAN, which promotes TANs survival and RT resistance [117]. Increased sensitivity of lung cancer to RT was observed in the TANs GLUT1 knock-out lung cancer mouse model [117].

TANs promote anti-angiogenesis therapy resistance in glioma

TKIs, whose main targets are RTKs, such as EGFR, PDGFR, VEGFR, and are commonly used anti-angiogenic agents that effectively inhibit glioma proliferation and invasion [118]. TANs can produce substances such as VEGFA, MMP9, MMP14, OPN, etc., which enhance VEGFA/VEGFR interaction that promotes glioma angiogenesis. This pro-angiogenic effect largely cuts down the therapeutic efficacy of TKIs or requires larger doses of TKIs to achieve the same efficacy [19]. In addition to VEGF, TANs also produce Bv8 to promote tumor angiogenesis. Unlike VEGFR, the receptor for Bv8 belongs to the GPCR family, therefore, TKIs cannot inhibit the pro-angiogenic effect produced by Bv8, resulting in TKIs resistance in tumors [68, 69]. Several studies based on different solid tumors corroborate this and find that inhibiting TANs can enhance the efficacy of TKIs [119,120,121]. Additional studies have demonstrated that TANs mediate glioma resistance to anti-angiogenic therapy by promoting glioma mesenchymal transition. de Groot JF and colleagues compared acquired bevacizumab-resistant versus non-resistant GB subtypes and found a highly significant correlation between acquired resistance to anti-VEGF therapy and mesenchymal features in GB [122]. Subsequently, they determined that TANs promote GB mesenchymal transition through S100A4 and discovered that S100A4 knockdown neutrophils attenuated mesenchymal transformation and angiogenesis of GB by not affecting neutrophil proliferation [83]. Hence, a combination therapy involving S100A4 inhibitors may be a viable option to counteract resistance to anti-angiogenic therapy in glioma. No relevant clinical/preclinical trials were retrieved, and the specific effects of this regimen need to be further investigated.

TANs promote immune checkpoint blockade therapy resistance in glioma

Immune checkpoint blockade (ICB) has shown remarkable success in various solid tumors, but good efficacy has not been observed in glioma, an important reason being the presence of significant immunosuppression in glioma [123, 124]. Blocking the PD-1/PD-L1 axis represents one of the classic ICB therapies. PD-1 is primarily expressed in activated T-cells and its interaction with PD-L1 reduces the immune response of T-cells [109, 124]. Commonly used drugs include PD-1 antibodies such as Nivolumab, Pembrolizumab, and emerging PD-L1 antibodies such as Atezolizumab [124, 125]. However, a prerequisite for these therapies to be effective is that T-cells possess (at least to some extent) normal functionality. As mentioned earlier, TANs in the glioma microenvironment can cause T-cell dysfunction through various pathways, particularly depleting the functionality of CD4+TEM via the PD-1/PD-L1 axis [107, 108, 112, 113]. These can promote glioma treatment resistance to ICB.

Therapies targeting TANs

Anti-angiogenesis therapy

Presently, the standard therapy for newly diagnosed glioma involves surgical resection followed by radiotherapy plus TMZ combined chemotherapeutic treatment [5, 126]. However, almost all patients experience recurrence [5, 126]. Bevacizumab, an anti-VEGF antibody, has been approved for treating recurrent glioma in several countries, including the United States [5]. Nonetheless, current clinical trials have not shown distinct advantages of bevacizumab. Although the use of bevacizumab improves PFS, the OS of patients was not improved compared to standard therapy [127, 128]. Apart from bevacizumab, regorafenib has shown promising progress in recent studies. Regorafenib is an orally available multikinase inhibitor used against angiogenesis, stromal, and RTK [129]. In a phase II clinical trial, regorafenib, when compared to lomustine (TMZ analog), increased the median OS from 5.6 to 7.4 months, with a comparable rate of severe adverse events [129]. Regorafenib might be a potential novel therapy. (Table 1)

Table 1 Clinical trials targeting TANs and/or its correlating agents

PD-1/PD-L1 blockade

PD-1/PD-L1 blockade has shown significant efficacy in many tumors but remains limited effective in glioma. The classic PD-1 monoclonal antibody nivolumab has efficacy comparable to bevacizumab [130]. Combining nivolumab with standard therapy does not influence patient survival [126]. Single-agent use of the PD-1 antibody pembrolizumab is effective for only a small subset of recurrent GB patients [131]. The combination of the PD-L1 antibody durvalumab with bevacizumab has proven ineffective [132]. These less optimistic results encourage researchers to explore new directions. Several studies have shown that adjuvant or neoadjuvant PD-1 monoclonal antibodies, such as nivolumab and pembrolizumab, can enhance the anti-tumor immune response of glioma patients, resulting in a positive effect on glioma treatment [133, 134]. Pembrolizumab combined with DNX-2401, an oncolytic virus capable of inducing an anti-tumor immune response in patients, has shown significant survival benefits for some glioma patients [135]. Furthermore, innovative delivery methods are potential avenues of development. A phase I clinical trial has confirmed that intracerebral administration of nivolumab and CTLA-4 inhibitor ipilimumab after maximal safe resection is feasible, safe, and has a positive effect on OS [136]. Recent studies have found that leucine stimulates TANs to highly express HLA-DR+, present tumor neoantigens and stimulate T cells to produce reactive T cell responses [8]. Animal studies have shown that a leucine diet combined with anti-PD-1 therapy significantly reduces tumor volume and stabilizes the disease [8]. (Table 1)

Targeting CXCLs-CXCR1/2 signaling

IL-8 (CXCL8) plays an important role in neutrophil recruitment to TME and is involved in tumor malignant behavior. Pre-clinical studies have shown that the IL-8 antagonist BMS-986253 significantly reduces neutrophil recruitment and attenuates mesenchymal transition in triple-negative breast cancer [137]. Phase I studies have found that BMS-986253 significantly reduces IL-8 levels and is being evaluated in combination therapy with immune checkpoint inhibitors (NCT02536469) [138]. CXCR2 is an essential neutrophil chemotactic factor receptor, recruiting neutrophils to the TME and regulating TANs participation in tumor angiogenesis [25]. CXCR1 also plays an important role in neutrophil recruitment [35]. In addition, the efficacy of CXCR1/2 antagonists in the treatment of non-neoplastic diseases has been demonstrated. For example, a phase II study demonstrated that MK-7123, a CXCR2 antagonist, significantly reduced neutrophil counts and significantly improved lung function in COPD patients (NCT01006616) [139]. Using CXCR2 antagonists for tumor therapy is a promising strategy, and this has shown good progress in clinical trials of Reparixin for breast cancer (NCT02001974, NCT02370238) [140]. A study using a CXCR2 antagonist (SB225002) in high-grade glioma demonstrated its potential to inhibit tumor growth during tumor development and progression [141]. (Table 1)

Targeting TGF-β signaling

TGF-β signaling plays a crucial role in glioma, inducing TANs to polarize toward the N2 type (pro-tumor), and several downstream signaling pathways are involved in tumor progression [38]. The use of TGF-β antagonists for the treatment of glioma is theoretically a viable option. Galunisertib (LY2157299 monohydrate) is a small molecule TGF-β-R1 kinase inhibitor that displays anti-tumor effects in GB animal models [142,143,144]. A phase I/II clinical trial has indicated that combining 300 mg/d of Galunisertib with TMZ chemotherapy can improve OS for GB patients [143]. Another phase II clinical trial has demonstrated that in recurrent GB patients, the treatment with Galunisertib plus lomustine did not improve OS compared to placebo plus lomustine treatment [145]. The specific efficacy of TGF-β blockade in glioma requires further investigation. (Table 1)

Targeting the S100 protein family

Multiple members of the S100 protein family exhibit dysregulated expression in cancer [81]. As mentioned above, TANs promote glioma metastasis through S100A4 [83]. Single-cell analysis has suggested that S100A4 could be a target for glioma immunotherapy [146]. Feng et al. have found that Allopregnanolone, a physiological neural modulator, significantly downregulates several proteins in GB, including S100A4 and S100A11, inhibiting GB cell survival [147]. An in vitro study has indicated that downregulating S100A12 (primarily expressed by neutrophils) significantly inhibits glioma cell growth [148]. These studies imply that the S100 protein family might be key molecular targets to explore new directions in glioma treatment.

Targeting NETs

NETs play a crucial role in the progression of various cancers, and targeting NETs is a potential new treatment direction. However, most of these studies have remained at the mechanistic level and have not achieved breakthrough progress [48]. In one study, NETs have led to endothelial dysfunction in advanced glioma patients, promoting a hypercoagulable state, which is necessary to cause the complication of venous thromboembolism (VTE) [149]. Researchers subsequently found that in vitro, the combination of DNase I and activated protein C significantly reduced endothelial dysfunction [149]. Furthermore, there is evidence suggesting that NETs might exhibit an anti-tumor effect [150]. Overall, although there is substantial evidence supporting the therapeutic targeting of NETs, we recommend consulting in-depth reviews on neutrophils, NETs, and cancer for more information [48, 49, 150,151,152].

Neutrophil-based drug delivery systems

Neutrophil-based drug delivery systems are a promising targeted cancer therapy strategy, primarily divided into two types: those utilizing neutrophils as carriers and neutrophil membrane-driven nanovesicles [153]. The most significant advantage of this system is its natural ability to traverse the blood-brain barrier (BBB) as neutrophils naturally recruit to the tumor microenvironment. In recent years, studies utilizing this system for glioma treatment have made significant progress. Wang et al. have reported a neutrophil-exosome system targeting the inflamed tumor microenvironment, showing success in crossing the BBB and infiltrating tumor tissues in a GB animal model loaded with doxorubicin in response to inflammatory stimuli [154]. Zhang et al. have reported a neutrophil-based micro-robot (neutrobot) capable of actively delivering drugs to malignant glioma [155]. Using magnetic drug-loaded particles phagocytosed by neutrophils and their chemotactic movement along inflammatory gradients, precise drug delivery significantly suppressed tumor cell proliferation [155]. Although emerging chimeric antigen receptor (CAR)-T cell therapy has been successful in many cancers, its effectiveness in gliomas is limited due to the difficulty of T cells crossing the BBB [156]. Recently, Chang et al. have engineered CAR-neutrophils using CRISPR/Cas9 in human pluripotent stem cells, targeting GB, delivering and releasing nanomedicines to kill tumor cells, prolonging the lifespan of mouse models, contributing to the application of CAR therapy in intracranial tumors [156].

Conclusions and perspectives

In this review, we provide a comprehensive description of the functions and mechanisms of TANs in glioma progression. In most cases, TANs exhibit a pro-tumoral N2 phenotype, promoting glioma cell proliferation, invasion, PN-MES transition, and resistance to radiotherapy and chemotherapy through paracrine and other forms [54]. It is important to note that the role of TANs in tumor progression is context-dependent, TANs can also adopt an anti-tumoral N1 phenotype that inhibits tumor growth [11]. In addition to tumor heterogeneity, the different phenotypes of TANs may be related to the time at which they are recruited into the tumor microenvironment, where they are more likely to exhibit an N1 phenotype at early stages and are gradually induced by the tumor microenvironment to an N2 phenotype over time. Furthermore, a large number of bioinformatic analysis studies have reported a series of new genes or genesets associated with the proportion of TANs infiltration in gliomas and the prognosis of glioma patients, which we have summarized in this paper. However, most of these reports are phenotypic studies, the mechanisms of the above genes and genesets on the infiltration and function of TANs still need in-depth elucidation.

Gliomas are hallmarked by their complex tumor microenvironments [157]. The extensive crosstalk between tumor cells and microenvironmental components within the glioma microenvironment and the formation of positive feedbacks that promote tumor growth and malignant progression, which are important causes of recurrence and treatment resistance in gliomas [157]. In this review, we focus on elucidating the interactions between glioma microenvironment components and TANs and their effects on tumor progression, including the hypoxic microenvironment, vascular endothelial cells, TAMs and T cells. TANs have extensive interactions with multiple microenvironmental components and are essential for maintaining their pro-tumoral N2 phenotype. Therefore, further investigation into the role of TANs in tumor progression and the development of targeted therapies, whether for gliomas or other malignancies, should take full account of the microenvironment in which TANs and tumor cells reside in the tumor, in conjunction with existing tools such as spatial transcriptomics.

Finally, we summarize current advances in TANs-based anti-tumor therapies. Although TANs are highly promising anti-glioma targets, because the mechanism TANs in glioma progression is still largely unclear, there are currently fewer targets that can specifically target TANs within tumors, and even fewer corresponding clinical trials. As the mechanism TANs continues to be elucidated, more therapies targeting TANs will become available for the clinical treatment of gliomas and other tumors, both alone and in combination with therapies including immune checkpoint inhibitors. Additionally, the treatment of glioma with drug delivery systems based on neutrophils represents a promising research direction that requires further refinement through more clinical experiments.

Data availability

No datasets were generated or analysed during the current study.

Abbreviations

APC:

Antigen-presenting cell

Arg:

Arginine

Arg1:

Arginase 1

BBB:

Blood-brain barrier

BMP2:

Bone morphogenetic protein 2

CAR:

Chimeric antigen receptor

CCL:

C-C motif ligand 3

CL:

Classical

CTL:

Cytotoxic T cell

CXCLs:

CXC motif ligands

CXCR:

CXC chemokine receptor

DCs:

Dendritic cells

ECM:

Extracellular matrix

ECs:

Endothelial cells

EVs:

Extracellular vesicles

EG-VEGF:

Endocrine gland-derived vascular endothelial growth factor

GB:

Glioblastoma

G-CSF:

Granulocyte colony-stimulating factor

G-CSF-R:

Granulocyte colony-stimulating factor receptor

GM-CSF:

Granulocyte-macrophage colony-stimulating factor

HCC:

Hepatocellular carcinoma

HIF:

Hypoxia-inducible factor

ICAM-1:

Intercellular cell adhesion molecule-1

ICB:

Immune checkpoint blockade

ICC:

Intrahepatic cholangiocarcinoma

IDH:

Isocitrate dehydrogenase

IFN:

Interferon

IL:

Interleukin

iNOS:

Inducible nitric oxide synthase

IRS-1:

Insulin receptor substrate-1

MCP:

Monocyte chemoattractant protein

MES:

Mesenchymal

MMP9:

Matrix metalloproteinase 9

MPO:

Myeloperoxidase

mtROS:

Mitochondrial reactive oxygen species

mu-IDH:

Mutant isocitrate dehydrogenase

NE:

Neutrophil elastase

NETs:

Neutrophil extracellular traps

NL:

Neural

NLR:

Neutrophil-to-lymphocyte ratio

NSCLC:

Non-small cell lung cancer

OPN:

Osteopontin

OS:

Overall survival

ox-PE:

Oxidized phosphatidylethanolamine

PBNs:

Peripheral blood neutrophils

PD-1:

Programmed cell death receptor-1

PDGFB:

Platelet-derived growth factor B

PDGFR:

Platelet-derived growth factor receptor

PD-L1:

Programmed cell death receptor 1 ligand-1

PFS:

Progression-free survival

PGE2:

Prostaglandin E2

PMT:

Proneural-mesenchymal transition

PN:

Proneural

RT:

Radiotherapy

RTKs:

Receptor tyrosine kinases

TANs:

Tumor-associated neutrophils

TAMs:

Tumor-associated macrophages

TEM:

Effector memory T-cell

TGF-β:

Transforming growth factor-β

TIMP-1:

Tissue inhibitors of metalloproteinase-1

TKIs:

Tyrosine kinase inhibitors

TLRs:

Toll-like receptors

TME:

Tumor microenvironment

TMZ:

Temozolomide

TNBC:

Triple-negative breast cancer

TNF-α:

Tumor necrosis factor-α

VEGF:

Vascular endothelial growth factor

VEGFA:

Vascular endothelial growth factor A

VEGFR:

Vascular endothelial growth factor receptor

VTE:

Venous thromboembolism

wt-IDH:

Wild-type isocitrate dehydrogenase

References

  1. Ostrom QT, Bauchet L, Davis FG, Deltour I, Fisher JL, Langer CE, Pekmezci M, Schwartzbaum JA, Turner MC, Walsh KM, et al. The epidemiology of glioma in adults: a state of the science review. Neuro Oncol. 2014;16:896–913.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Ostrom QT, Patil N, Cioffi G, Waite K, Kruchko C, Barnholtz-Sloan JS. CBTRUS Statistical Report: primary brain and other Central Nervous System tumors diagnosed in the United States in 2013–2017. Neuro Oncol. 2020;22:iv1–96.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Louis DN, Perry A, Wesseling P, Brat DJ, Cree IA, Figarella-Branger D, Hawkins C, Ng HK, Pfister SM, Reifenberger G, et al. The 2021 WHO classification of tumors of the Central Nervous System: a summary. Neuro Oncol. 2021;23:1231–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Kotecha R, Odia Y, Khosla AA, Ahluwalia MS. Key clinical principles in the management of Glioblastoma. JCO Oncol Pract. 2023;19:180–9.

    Article  PubMed  Google Scholar 

  5. Weller M, van den Bent M, Preusser M, Le Rhun E, Tonn JC, Minniti G, Bendszus M, Balana C, Chinot O, Dirven L, et al. EANO guidelines on the diagnosis and treatment of diffuse gliomas of adulthood. Nat Reviews Clin Oncol. 2021;18:170–86.

    Article  Google Scholar 

  6. van Solinge TS, Nieland L, Chiocca EA, Broekman MLD. Advances in local therapy for Glioblastoma - taking the fight to the tumour. Nat Rev Neurol. 2022;18:221–36.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Quail DF, Joyce JA. The Microenvironmental Landscape of Brain tumors. Cancer Cell. 2017;31:326–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Wu Y, Ma J, Yang X, Nan F, Zhang T, Ji S, Rao D, Feng H, Gao K, Gu X, et al. Neutrophil profiling illuminates anti-tumor antigen-presenting potency. Cell. 2024;187:1422–e14391424.

    Article  CAS  PubMed  Google Scholar 

  9. Klemm F, Maas RR, Bowman RL, Kornete M, Soukup K, Nassiri S, Brouland JP, Iacobuzio-Donahue CA, Brennan C, Tabar V, et al. Interrogation of the Microenvironmental Landscape in Brain Tumors reveals Disease-specific alterations of Immune cells. Cell. 2020;181:1643–.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Friedmann-Morvinski D, Hambardzumyan D. Monocyte-neutrophil entanglement in glioblastoma. J Clin Invest 2023, 133.

  11. Giese MA, Hind LE, Huttenlocher A. Neutrophil plasticity in the tumor microenvironment. Blood. 2019;133:2159–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Massara M, Persico P, Bonavita O, Mollica Poeta V, Locati M, Simonelli M, Bonecchi R. Neutrophils in Gliomas. Front Immunol. 2017;8:1349.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Masucci MT, Minopoli M, Carriero MV. Tumor Associated neutrophils. Their role in Tumorigenesis, Metastasis, Prognosis and Therapy. Front Oncol. 2019;9:1146.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Fridlender ZG, Albelda SM. Tumor-associated neutrophils: friend or foe? Carcinogenesis 2012, 33:949–55.

  15. Gregory AD, Houghton AM. Tumor-associated neutrophils: new targets for cancer therapy. Cancer Res. 2011;71:2411–6.

    Article  CAS  PubMed  Google Scholar 

  16. Coffelt SB, Wellenstein MD, de Visser KE. Neutrophils in cancer: neutral no more. Nat Rev Cancer. 2016;16:431–46.

    Article  CAS  PubMed  Google Scholar 

  17. Amankulor NM, Kim Y, Arora S, Kargl J, Szulzewsky F, Hanke M, Margineantu DH, Rao A, Bolouri H, Delrow J, et al. Mutant IDH1 regulates the tumor-associated immune system in gliomas. Genes Dev. 2017;31:774–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. De Palma M, Biziato D, Petrova TV. Microenvironmental regulation of tumour angiogenesis. Nat Rev Cancer. 2017;17:457–74.

    Article  PubMed  Google Scholar 

  19. Bui TM, Yalom LK, Sumagin R. Tumor-associated neutrophils: orchestrating cancer pathobiology and therapeutic resistance. Expert Opin Ther Targets. 2021;25:573–83.

    Article  CAS  PubMed  Google Scholar 

  20. Lad BM, Beniwal AS, Jain S, Shukla P, Jung J, Shah SS, Yagnik G, Babikir H, Nguyen AT, Gill S et al. Glioblastoma induces the recruitment and differentiation of hybrid neutrophils from skull bone marrow. bioRxiv 2023.

  21. Singhal S, Bhojnagarwala PS, O’Brien S, Moon EK, Garfall AL, Rao AS, Quatromoni JG, Stephen TL, Litzky L, Deshpande C, et al. Origin and role of a subset of Tumor-Associated neutrophils with Antigen-presenting cell features in early-stage human lung Cancer. Cancer Cell. 2016;30:120–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Kolaczkowska E, Kubes P. Neutrophil recruitment and function in health and inflammation. Nat Rev Immunol. 2013;13:159–75.

    Article  CAS  PubMed  Google Scholar 

  23. Buonacera A, Stancanelli B, Colaci M, Malatino L. Neutrophil to lymphocyte ratio: an emerging marker of the relationships between the Immune System and diseases. Int J Mol Sci 2022, 23.

  24. Gomes Dos Santos A, de Carvalho RF, de Morais ANLR, Silva TM, Baylão VMR, Azevedo M, de Oliveira AJM. Role of neutrophil-lymphocyte ratio as a predictive factor of glioma t umor grade: A systematic review. Critical reviews in oncology/hematology 2021, 163:103372.

  25. Stadtmann A, Zarbock A. CXCR2: from bench to Bedside. Front Immunol. 2012;3:263.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Wang G, Wang J, Niu C, Zhao Y, Wu P. Neutrophils: new critical regulators of Glioma. Front Immunol. 2022;13:927233.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Yang L, Liu Z, Wu R, Yao Q, Gu Z, Liu M. Correlation of C-X-C chemokine receptor 2 upregulation with poor prognosis and recurrence in human glioma. Onco Targets Ther. 2015;8:3203–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Chen N, Alieva M, van der Most T, Klazen JAZ, Vollmann-Zwerenz A, Hau P, Vrisekoop N. Neutrophils promote Glioblastoma Tumor Cell Migration after Biopsy. Cells 2022, 11.

  29. Köhler A, De Filippo K, Hasenberg M, van den Brandt C, Nye E, Hosking MP, Lane TE, Männ L, Ransohoff RM, Hauser AE, et al. G-CSF-mediated thrombopoietin release triggers neutrophil motility and mobilization from bone marrow via induction of Cxcr2 ligands. Blood. 2011;117:4349–57.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Kasahara T, Mukaida N, Yamashita K, Yagisawa H, Akahoshi T, Matsushima K. IL-1 and TNF-alpha induction of IL-8 and monocyte chemotactic and activating factor (MCAF) mRNA expression in a human astrocytoma cell line. Immunology. 1991;74:60–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Griffin BD, Moynagh PN. Persistent interleukin-1beta signaling causes long term activation of NFkappaB in a promoter-specific manner in human glial cells. J Biol Chem. 2006;281:10316–26.

    Article  CAS  PubMed  Google Scholar 

  32. Tarassishin L, Lim J, Weatherly DB, Angeletti RH, Lee SC. Interleukin-1-induced changes in the glioblastoma secretome suggest its role in tumor progression. J Proteom. 2014;99:152–68.

    Article  CAS  Google Scholar 

  33. Hor WS, Huang WL, Lin YS, Yang BC. Cross-talk between tumor cells and neutrophils through the Fas (APO-1, CD95)/FasL system: human glioma cells enhance cell viability and stimulate cytokine production in neutrophils. J Leukoc Biol. 2003;73:363–8.

    Article  CAS  PubMed  Google Scholar 

  34. Zha C, Meng X, Li L, Mi S, Qian D, Li Z, Wu P, Hu S, Zhao S, Cai J, Liu Y. Neutrophil extracellular traps mediate the crosstalk between glioma progression and the tumor microenvironment via the HMGB1/RAGE/IL-8 axis. Cancer Biol Med. 2020;17:154–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Powell D, Lou M, Barros Becker F, Huttenlocher A. Cxcr1 mediates recruitment of neutrophils and supports proliferation of tumor-initiating astrocytes in vivo. Sci Rep. 2018;8:13285.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Wang T, Cao L, Dong X, Wu F, De W, Huang L, Wan Q. LINC01116 promotes tumor proliferation and neutrophil recruitment via DDX5-mediated regulation of IL-1β in glioma cell. Cell Death Dis 2020, 11.

  37. Lee SY, Kim JK, Jeon HY, Ham SW, Kim H. CD133 regulates IL-1beta signaling and Neutrophil Recruitment in Glioblastoma. Mol Cells. 2017;40:515–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Fridlender ZG, Sun J, Kim S, Kapoor V, Cheng G, Ling L, Worthen GS, Albelda SM. Polarization of tumor-associated neutrophil phenotype by TGF-beta: N1 versus N2 TAN. Cancer Cell. 2009;16:183–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Zhu S, Yi M, Wu Y, Dong B, Wu K. Roles of tumor-associated macrophages in tumor progression: implicatio ns on therapeutic strategies. Experimental Hematol Oncol. 2021;10:60.

    Article  CAS  Google Scholar 

  40. Kim J, Bae JS. Tumor-Associated Macrophages and Neutrophils in Tumor Microenvironment. Mediators Inflamm 2016, 2016:6058147.

  41. Mishalian I, Bayuh R, Levy L, Zolotarov L, Michaeli J, Fridlender ZG. Tumor-associated neutrophils (TAN) develop pro-tumorigenic properties during tumor progression. Cancer Immunol Immunother. 2013;62:1745–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Ng MSF, Kwok I, Tan L, Shi C, Cerezo-Wallis D, Tan Y, Leong K, Calvo GF, Yang K, Zhang Y, et al. Deterministic reprogramming of neutrophils within tumors. Science. 2024;383:eadf6493.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Ng MSF, Tan L, Wang Q, Mackay CR, Ng LG. Neutrophils in cancer-unresolved questions. Sci China Life Sci. 2021;64:1829–41.

    Article  CAS  PubMed  Google Scholar 

  44. Sathornsumetee S, Rich JN. Chap. 18 - Molecularly targeted therapy in neuro-oncology. In Handbook of Clinical Neurology. Volume 104. Edited by Aminoff MJ, Boller F, Swaab DF: Elsevier; 2012: 255–278.

  45. Chen R, Smith-Cohn M, Cohen AL, Colman H. Glioma subclassifications and their clinical significance. Neurotherapeutics. 2017;14:284–97.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Han S, Liu Y, Cai SJ, Qian M, Ding J, Larion M, Gilbert MR, Yang C. IDH mutation in glioma: molecular mechanisms and potential therapeutic targets. Br J Cancer. 2020;122:1580–9.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Andzinski L, Kasnitz N, Stahnke S, Wu CF, Gereke M, von Kockritz-Blickwede M, Schilling B, Brandau S, Weiss S, Jablonska J. Type I IFNs induce anti-tumor polarization of tumor associated neutrophils in mice and human. Int J Cancer. 2016;138:1982–93.

    Article  CAS  PubMed  Google Scholar 

  48. Demkow U. Neutrophil Extracellular traps (NETs) in Cancer Invasion, Evasion and Metastasis. Cancers (Basel) 2021, 13.

  49. Cristinziano L, Modestino L, Antonelli A, Marone G, Simon HU, Varricchi G, Galdiero MR. Neutrophil extracellular traps in cancer. Semin Cancer Biol. 2022;79:91–104.

    Article  CAS  PubMed  Google Scholar 

  50. Albrengues J, Shields MA, Ng D, Park CG, Ambrico A, Poindexter ME, Upadhyay P, Uyeminami DL, Pommier A, Kuttner V et al. Neutrophil extracellular traps produced during inflammation awaken dormant cancer cells in mice. Science 2018, 361.

  51. Iwatsuki K, Kumara E, Yoshimine T, Nakagawa H, Sato M, Hayakawa T. Elastase expression by infiltrating neutrophils in gliomas. Neurol Res. 2000;22:465–8.

    Article  CAS  PubMed  Google Scholar 

  52. Crocker J, Jenkins R, Burnett D. Immunohistochemical demonstration of leucocyte elastase in human tissues. J Clin Pathol. 1984;37:1114–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Xue Q, Cao L, Chen XY, Zhao J, Gao L, Li SZ, Fei Z. High expression of MMP9 in glioma affects cell proliferation and is associated with patient survival rates. Oncol Lett. 2017;13:1325–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Rubenich DS, de Souza PO, Omizzollo N, Aubin MR, Basso PJ, Silva LM, da Silva EM, Teixeira FC, Gentil GFS, Domagalski JL, et al. Tumor-neutrophil crosstalk promotes in vitro and in vivo glioblastoma progression. Front Immunol. 2023;14:1183465.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Houghton AM, Rzymkiewicz DM, Ji H, Gregory AD, Egea EE, Metz HE, Stolz DB, Land SR, Marconcini LA, Kliment CR, et al. Neutrophil elastase-mediated degradation of IRS-1 accelerates lung tumor growth. Nat Med. 2010;16:219–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Hattar K, Franz K, Ludwig M, Sibelius U, Wilhelm J, Lohmeyer J, Savai R, Subtil FS, Dahlem G, Eul B, et al. Interactions between neutrophils and non-small cell lung cancer cells: enhancement of tumor proliferation and inflammatory mediator synthesis. Cancer Immunol Immunother. 2014;63:1297–306.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Zhou SL, Yin D, Hu ZQ, Luo CB, Zhou ZJ, Xin HY, Yang XR, Shi YH, Wang Z, Huang XW, et al. A positive Feedback Loop between Cancer Stem-Like cells and Tumor-Associated neutrophils Controls Hepatocellular Carcinoma Progression. Hepatology. 2019;70:1214–30.

    Article  CAS  PubMed  Google Scholar 

  58. Yee PP, Wei Y, Kim SY, Lu T, Chih SY, Lawson C, Tang M, Liu Z, Anderson B, Thamburaj K, et al. Neutrophil-induced ferroptosis promotes tumor necrosis in glioblastoma progression. Nat Commun. 2020;11:5424.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Yao L, Sheng X, Dong X, Zhou W, Li Y, Ma X, Song Y, Dai H, Du Y. Neutrophil extracellular traps mediate TLR9/Merlin axis to resist ferroptosis and promote triple negative breast cancer progression. Apoptosis. 2023;28:1484–95.

    Article  CAS  PubMed  Google Scholar 

  60. Onishi M, Ichikawa T, Kurozumi K, Date I. Angiogenesis and invasion in glioma. Brain Tumor Pathol. 2011;28:13–24.

    Article  CAS  PubMed  Google Scholar 

  61. Tazzyman S, Lewis CE, Murdoch C. Neutrophils: key mediators of tumour angiogenesis. Int J Exp Pathol. 2009;90:222–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Simons M, Gordon E, Claesson-Welsh L. Mechanisms and regulation of endothelial VEGF receptor signalling. Nat Rev Mol Cell Biol. 2016;17:611–25.

    Article  CAS  PubMed  Google Scholar 

  63. Ardi VC, Kupriyanova TA, Deryugina EI, Quigley JP. Human neutrophils uniquely release TIMP-free MMP-9 to provide a potent catalytic stimulator of angiogenesis. Proc Natl Acad Sci U S A. 2007;104:20262–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Masure S, Proost P, Van Damme J, Opdenakker G. Purification and identification of 91-kDa neutrophil gelatinase. Release by the activating peptide interleukin-8. Eur J Biochem. 1991;198:391–8.

    Article  CAS  PubMed  Google Scholar 

  65. Van den Steen PE, Proost P, Wuyts A, Van Damme J, Opdenakker G. Neutrophil gelatinase B potentiates interleukin-8 tenfold by aminoterminal processing, whereas it degrades CTAP-III, PF-4, and GRO-alpha and leaves RANTES and MCP-2 intact. Blood. 2000;96:2673–81.

    Article  PubMed  Google Scholar 

  66. Cassatella MA. Neutrophil-derived proteins: selling cytokines by the pound. Adv Immunol. 1999;73:369–509.

    Article  CAS  PubMed  Google Scholar 

  67. Shojaei F, Wu X, Zhong C, Yu L, Liang X-H, Yao J, Blanchard D, Bais C, Peale FV, van Bruggen N, et al. Bv8 regulates myeloid-cell-dependent tumour angiogenesis. Nature. 2007;450:825–31.

    Article  CAS  PubMed  Google Scholar 

  68. LeCouter J, Ferrara N. EG-VEGF and Bv8. A novel family of tissue-selective mediators of angiogenesis, endothelial phenotype, and function. Trends Cardiovasc Med. 2003;13:276–82.

    Article  CAS  PubMed  Google Scholar 

  69. Xin H, Lu R, Lee H, Zhang W, Zhang C, Deng J, Liu Y, Shen S, Wagner K-U, Forman S, et al. G-protein-coupled receptor agonist BV8/prokineticin-2 and STAT3 protei n form a feed-forward loop in both normal and malignant myeloid cells. J Biol Chem. 2013;288:13842–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Qu X, Zhuang G, Yu L, Meng G, Ferrara N. Induction of Bv8 expression by granulocyte colony-stimulating factor i n CD11b + Gr1 + cells: key role of Stat3 signaling. J Biol Chem. 2012;287:19574–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Curtis VF, Wang H, Yang P, McLendon RE, Li X, Zhou QY, Wang XF. A PK2/Bv8/PROK2 antagonist suppresses tumorigenic processes by inhibiting angiogenesis in glioma and blocking myeloid cell infiltration in pancreatic cancer. PLoS ONE. 2013;8:e54916.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Bui TM, Yalom LK, Ning E, Urbanczyk JM, Ren X, Herrnreiter CJ, Disario JA, Wray B, Schipma MJ, Velichko YS et al. Tissue-specific reprogramming leads to angiogenic neutrophil specialization and tumor vascularization in colorectal cancer. J Clin Invest 2024, 134.

  73. Dai J, Peng L, Fan K, Wang H, Wei R, Ji G, Cai J, Lu B, Li B, Zhang D, et al. Osteopontin induces angiogenesis through activation of PI3K/AKT and ERK1/2 in endothelial cells. Oncogene. 2009;28:3412–22.

    Article  CAS  PubMed  Google Scholar 

  74. Han KY, Chang JH, Lee H, Azar DT. Proangiogenic interactions of vascular endothelial MMP14 with VEGF receptor 1 in VEGFA-Mediated corneal angiogenesis. Invest Ophthalmol Vis Sci. 2016;57:3313–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Blaheta RA, Powerski M, Hudak L, Juengel E, Jonas D, von Knethen A, Doerr HW, Cinatl J Jr. Tumor-endothelium cross talk blocks recruitment of neutrophils to endothelial cells: a novel mechanism of endothelial cell anergy. Neoplasia. 2009;11:1054–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Lee E, Yong RL, Paddison P, Zhu J. Comparison of glioblastoma (GBM) molecular classification methods. Semin Cancer Biol. 2018;53:201–11.

    Article  CAS  PubMed  Google Scholar 

  77. Lai Y, Lu X, Liao Y, Ouyang P, Wang H, Zhang X, Huang G, Qi S, Li Y. Crosstalk between glioblastoma and tumor microenvironment drives proneural-mesenchymal transition through ligand-receptor interactions. Genes Dis. 2023;11:874–89.

    Article  PubMed  PubMed Central  Google Scholar 

  78. Verhaak RG, Hoadley KA, Purdom E, Wang V, Qi Y, Wilkerson MD, Miller CR, Ding L, Golub T, Mesirov JP, et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell. 2010;17:98–110.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Fedele M, Cerchia L, Pegoraro S, Sgarra R, Manfioletti G. Proneural-mesenchymal transition: phenotypic plasticity to acquire Multitherapy Resistance in Glioblastoma. Int J Mol Sci 2019, 20.

  80. Hara T, Chanoch-Myers R, Mathewson ND, Myskiw C, Atta L, Bussema L, Eichhorn SW, Greenwald AC, Kinker GS, Rodman C, et al. Interactions between cancer cells and immune cells drive transitions to mesenchymal-like states in glioblastoma. Cancer Cell. 2021;39:779–e792711.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Bresnick AR, Weber DJ, Zimmer DB. S100 proteins in cancer. Nat Rev Cancer. 2015;15:96–109.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Fei F, Qu J, Zhang M, Li Y, Zhang S. S100A4 in cancer progression and metastasis: a systematic review. Oncotarget. 2017;8:73219–39.

    Article  PubMed  PubMed Central  Google Scholar 

  83. Liang J, Piao Y, Holmes L, Fuller GN, Henry V, Tiao N, de Groot JF. Neutrophils promote the malignant glioma phenotype through S100A4. Clin Cancer Res. 2014;20:187–98.

    Article  CAS  PubMed  Google Scholar 

  84. Chow KH, Park HJ, George J, Yamamoto K, Gallup AD, Graber JH, Chen Y, Jiang W, Steindler DA, Neilson EG, et al. S100A4 is a Biomarker and Regulator of Glioma Stem cells that is critical for mesenchymal transition in Glioblastoma. Cancer Res. 2017;77:5360–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Chen Z, Soni N, Pinero G, Giotti B, Eddins DJ, Lindblad KE, Ross JL, Puigdelloses Vallcorba M, Joshi T, Angione A, et al. Monocyte depletion enhances neutrophil influx and proneural to mesenchymal transition in glioblastoma. Nat Commun. 2023;14:1839.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. SenGupta S, Subramanian BC, Parent CA. Getting TANned: how the tumor microenvironment drives neutrophil recruitment. J Leukoc Biol. 2019;105:449–62.

    Article  CAS  PubMed  Google Scholar 

  87. Kaur B, Khwaja FW, Severson EA, Matheny SL, Brat DJ, Van Meir EG. Hypoxia and the hypoxia-inducible-factor pathway in glioma growth and angiogenesis. Neuro Oncol. 2005;7:134–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Liu D, Zhu HT, Cheng LD, Li R, Ma XY, Wang J, Wang JW, Zhang SJ, Li YJ, Shu K et al. Hypoxia-induced galectin-8 maintains stemness in glioma stem cells via autophagy regulation. Neuro-Oncology 2023.

  89. Görlach A, Bonello S. The cross-talk between NF-kappaB and HIF-1: further evidence for a sig nificant liaison. Biochem J. 2008;412:e17–19.

    Article  PubMed  Google Scholar 

  90. Jin MZ, Jin WL. The updated landscape of tumor microenvironment and drug repurposing. Signal Transduct Target Ther. 2020;5:166.

    Article  PubMed  PubMed Central  Google Scholar 

  91. Eales KL, Hollinshead KER, Tennant DA. Hypoxia and metabolic adaptation of cancer cells. Oncogenesis. 2016;5:e190.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Xu L, Xie K, Mukaida N, Matsushima K, Fidler IJ. Hypoxia-induced elevation in interleukin-8 expression by human ovarian carcinoma cells. Cancer Res. 1999;59:5822–9.

    CAS  PubMed  Google Scholar 

  93. Beaumont JEJ, Beelen NA, Wieten L, Rouschop KMA. The Immunomodulatory Role of Hypoxic Tumor-Derived Extracellular vesicles. Cancers (Basel) 2022, 14.

  94. Baggiolini M, Walz A, Kunkel SL. Neutrophil-activating peptide-1/interleukin 8, a novel cytokine that activates neutrophils. J Clin Invest. 1989;84:1045–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Murata K, Yoshitomi H, Furu M, Ishikawa M, Shibuya H, Ito H, Matsuda S. MicroRNA-451 down-regulates neutrophil chemotaxis via p38 MAPK. Arthritis Rheumatol. 2014;66:549–59.

    Article  CAS  PubMed  Google Scholar 

  96. Triner D, Xue X, Schwartz AJ, Jung I, Colacino JA, Shah YM. Epithelial hypoxia-inducible factor 2α facilitates the Progression of Colon Tumors through recruiting neutrophils. Mol Cell Biol. 2017;37:e00481–00416.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Tsai HC, Tong ZJ, Hwang TL, Wei KC, Chen PY, Huang CY, Chen KT, Lin YJ, Cheng HW, Wang HT. Acrolein produced by glioma cells under hypoxia inhibits neutrophil AKT activity and suppresses anti-tumoral activities. Free Radic Biol Med. 2023;207:17–28.

    Article  CAS  PubMed  Google Scholar 

  98. Broekman ML, Maas SLN, Abels ER, Mempel TR, Krichevsky AM, Breakefield XO. Multidimensional communication in the microenvirons of glioblastoma. Nat Rev Neurol. 2018;14:482–95.

    Article  PubMed  PubMed Central  Google Scholar 

  99. Grabowski MM, Sankey EW, Ryan KJ, Chongsathidkiet P, Lorrey SJ, Wilkinson DS, Fecci PE. Immune suppression in gliomas. J Neurooncol. 2021;151:3–12.

    Article  PubMed  Google Scholar 

  100. Alberghina C, Torrisi F, D’Aprile S, Longhitano L, Giallongo S, Scandura G, Mannino G, Mele S, Sabini MG, Cammarata FP, et al. Microglia and glioblastoma heterocellular interplay sustains tumour gr owth and proliferation as an off-target effect of radiotherapy. Cell Prolif. 2024;57:e13606.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Khan F, Pang L, Dunterman M, Lesniak MS, Heimberger AB, Chen P. Macrophages and microglia in glioblastoma: heterogeneity, plasticity, and therapy. J Clin Invest 2023, 133.

  102. Zhou Z, Wang P, Sun R, Li J, Hu Z, Xin H, Luo C, Zhou J, Fan J, Zhou S. Tumor-associated neutrophils and macrophages interaction contributes to intrahepatic cholangiocarcinoma progression by activating STAT3. J Immunother Cancer 2021, 9.

  103. Hu J, Zhang L, Xia H, Yan Y, Zhu X, Sun F, Sun L, Li S, Li D, Wang J, et al. Tumor microenvironment remodeling after neoadjuvant immunotherapy in non-small cell lung cancer revealed by single-cell RNA sequencing. Genome Med. 2023;15:14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Ponzetta A, Carriero R, Carnevale S, Barbagallo M, Molgora M, Perucchini C, Magrini E, Gianni F, Kunderfranco P, Polentarutti N, et al. Neutrophils driving unconventional T cells mediate resistance against murine sarcomas and selected human tumors. Cell. 2019;178:346–e360324.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Basheer AS, Abas F, Othman I, Naidu R. Role of Inflammatory mediators, macrophages, and neutrophils in Glioma Maintenance and progression: mechanistic understanding and potential therapeutic applications. Cancers (Basel) 2021, 13.

  106. Locarno CV, Simonelli M, Carenza C, Capucetti A, Stanzani E, Lorenzi E, Persico P, Della Bella S, Passoni L, Mavilio D, et al. Role of myeloid cells in the immunosuppressive microenvironment in gliomas. Immunobiology. 2020;225:151853.

    Article  CAS  PubMed  Google Scholar 

  107. Jaillon S, Ponzetta A, Di Mitri D, Santoni A, Bonecchi R, Mantovani A. Neutrophil diversity and plasticity in tumour progression and therapy. Nat Rev Cancer. 2020;20:485–503.

    Article  CAS  PubMed  Google Scholar 

  108. Sippel TR, White J, Nag K, Tsvankin V, Klaassen M, Kleinschmidt-DeMasters BK, Waziri A. Neutrophil degranulation and immunosuppression in patients with GBM: restoration of cellular immune function by targeting arginase I. Clin Cancer Res. 2011;17:6992–7002.

    Article  CAS  PubMed  Google Scholar 

  109. Han Y, Liu D, Li L. PD-1/PD-L1 pathway: current researches in cancer. Am J Cancer Res. 2020;10:727–42.

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Wang X, Guo G, Guan H, Yu Y, Lu J, Yu J. Challenges and potential of PD-1/PD-L1 checkpoint blockade immunothera py for glioblastoma. J Experimental Clin cancer Research: CR. 2019;38:87.

    Article  CAS  PubMed Central  Google Scholar 

  111. Maas RR, Soukup K, Fournier N, Massara M, Galland S, Kornete M, Wischnewski V, Lourenco J, Croci D, Álvarez-Prado ÁF, et al. The local microenvironment drives activation of neutrophils in human b rain tumors. Cell. 2023;186:4546–e45664527.

    Article  CAS  PubMed  Google Scholar 

  112. Dubinski D, Wolfer J, Hasselblatt M, Schneider-Hohendorf T, Bogdahn U, Stummer W, Wiendl H, Grauer OM. CD4 + T effector memory cell dysfunction is associated with the accumulation of granulocytic myeloid-derived suppressor cells in glioblastoma patients. Neuro Oncol. 2016;18:807–18.

    Article  CAS  PubMed  Google Scholar 

  113. Fujita M, Scheurer ME, Decker SA, McDonald HA, Kohanbash G, Kastenhuber ER, Kato H, Bondy ML, Ohlfest JR, Okada H. Role of type 1 IFNs in antiglioma immunosurveillance–using mouse studies to guide examination of novel prognostic markers in humans. Clin Cancer Res. 2010;16:3409–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Kim R, Hashimoto A, Markosyan N, Tyurin VA, Tyurina YY, Kar G, Fu S, Sehgal M, Garcia-Gerique L, Kossenkov A, et al. Ferroptosis of tumour neutrophils causes immune suppression in cancer. Nature. 2022;612:338–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Khan S, Mittal S, McGee K, Alfaro-Munoz KD, Majd N, Balasubramaniyan V, de Groot JF. Role of neutrophils and myeloid-derived suppressor cells in Glioma Progression and Treatment Resistance. Int J Mol Sci 2020, 21.

  116. Jeon HY, Ham SW, Kim JK, Jin X, Lee SY, Shin YJ, Choi CY, Sa JK, Kim SH, Chun T, et al. Ly6G(+) inflammatory cells enable the conversion of cancer cells to cancer stem cells in an irradiated glioblastoma model. Cell Death Differ. 2019;26:2139–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Ancey PB, Contat C, Boivin G, Sabatino S, Pascual J, Zangger N, Perentes JY, Peters S, Abel ED, Kirsch DG, et al. GLUT1 expression in Tumor-Associated neutrophils promotes Lung Cancer Growth and Resistance to Radiotherapy. Cancer Res. 2021;81:2345–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Nakada M, Kita D, Watanabe T, Hayashi Y, Hamada J. Mechanism of chemoresistance against tyrosine kinase inhibitors in malignant glioma. Brain Tumor Pathol. 2014;31:198–207.

    Article  CAS  PubMed  Google Scholar 

  119. Nozawa H, Chiu C, Hanahan D. Infiltrating neutrophils mediate the initial angiogenic switch in a mo use model of multistage carcinogenesis. Proceedings of the National Academy of Sciences of the United States o f America 2006, 103:12493–12498.

  120. Finke J, Ko J, Rini B, Rayman P, Ireland J, Cohen P. MDSC as a mechanism of tumor escape from sunitinib mediated anti-angio genic therapy. Int Immunopharmacol. 2011;11:856–61.

    Article  CAS  PubMed  Google Scholar 

  121. Itatani Y, Yamamoto T, Zhong C, Molinolo AA, Ruppel J, Hegde P, Taketo MM, Ferrara N. Suppressing neutrophil-dependent angiogenesis abrogates resistance to anti-VEGF antibody in a genetic model of colorectal cancer. Proceedings of the National Academy of Sciences of the United States o f America 2020, 117:21598–21608.

  122. Piao Y, Liang J, Holmes L, Henry V, Sulman E, de Groot JF. Acquired resistance to anti-VEGF therapy in glioblastoma is associated with a mesenchymal transition. Clin cancer Research: Official J Am Associa Tion Cancer Res. 2013;19:4392–403.

    Article  CAS  Google Scholar 

  123. Arrieta VA, Dmello C, McGrail DJ, Brat DJ, Lee-Chang C, Heimberger AB, Chand D, Stupp R, Sonabend AM. Immune checkpoint blockade in glioblastoma: from tumor heterogeneity to personalized treatment. J Clin Invest 2023, 133.

  124. Torrisi F, D’Aprile S, Denaro S, Pavone AM, Alberghina C, Zappalà A, Giuffrida R, Salvatorelli L, Broggi G, Magro GG et al. Epigenetics and metabolism reprogramming interplay into Glioblastoma: Novel insights on immunosuppressive mechanisms. Antioxid (Basel) 2023, 12.

  125. Morad G, Helmink BA, Sharma P, Wargo JA. Hallmarks of response, resistance, and toxicity to immune checkpoint blockade. Cell. 2021;184:5309–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Lim M, Weller M, Idbaih A, Steinbach J, Finocchiaro G, Raval RR, Ansstas G, Baehring J, Taylor JW, Honnorat J et al. Phase III trial of chemoradiotherapy with temozolomide plus nivolumab or placebo for newly diagnosed glioblastoma with methylated MGMT promo ter. Neuro-oncology 2022, 24:1935–1949.

  127. Chinot OL, Wick W, Mason W, Henriksson R, Saran F, Nishikawa R, Carpentier AF, Hoang-Xuan K, Kavan P, Cernea D, et al. Bevacizumab plus radiotherapy-temozolomide for newly diagnosed Gliobla Stoma. N Engl J Med. 2014;370:709–22.

    Article  CAS  PubMed  Google Scholar 

  128. Gilbert MR, Dignam JJ, Armstrong TS, Wefel JS, Blumenthal DT, Vogelbaum MA, Colman H, Chakravarti A, Pugh S, Won M, et al. A randomized trial of bevacizumab for newly diagnosed glioblastoma. N Engl J Med. 2014;370:699–708.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Lombardi G, De Salvo GL, Brandes AA, Eoli M, Rudà R, Faedi M, Lolli I, Pace A, Daniele B, Pasqualetti F, et al. Regorafenib compared with lomustine in patients with relapsed glioblas toma (REGOMA): a multicentre, open-label, randomised, controlled, phas e 2 trial. Lancet Oncol. 2019;20:110–9.

    Article  CAS  PubMed  Google Scholar 

  130. Reardon DA, Brandes AA, Omuro A, Mulholland P, Lim M, Wick A, Baehring J, Ahluwalia MS, Roth P, Bähr O, et al. Effect of Nivolumab vs Bevacizumab in patients with recurrent Glioblas toma: the CheckMate 143 phase 3 Randomized Clinical Trial. JAMA Oncol. 2020;6:1003–10.

    Article  PubMed  Google Scholar 

  131. Reardon DA, Kim TM, Frenel J-S, Simonelli M, Lopez J, Subramaniam DS, Siu LL, Wang H, Krishnan S, Stein K, Massard C. Treatment with pembrolizumab in programmed death ligand 1-positive rec urrent glioblastoma: results from the multicohort phase 1 KEYNOTE-028 trial. Cancer. 2021;127:1620–9.

    Article  CAS  PubMed  Google Scholar 

  132. Nayak L, Standifer N, Dietrich J, Clarke JL, Dunn GP, Lim M, Cloughesy T, Gan HK, Flagg E, George E, et al. Circulating Immune Cell and Outcome Analysis from the phase II study o f PD-L1 blockade with Durvalumab for newly diagnosed and recurrent gli oblastoma. Clin cancer Research: Official J Am Associa Tion Cancer Res. 2022;28:2567–78.

    Article  CAS  Google Scholar 

  133. Cloughesy TF, Mochizuki AY, Orpilla JR, Hugo W, Lee AH, Davidson TB, Wang AC, Ellingson BM, Rytlewski JA, Sanders CM, et al. Neoadjuvant anti-PD-1 immunotherapy promotes a survival benefit with i ntratumoral and systemic immune responses in recurrent glioblastoma. Nat Med. 2019;25:477–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Schalper KA, Rodriguez-Ruiz ME, Diez-Valle R, López-Janeiro A, Porciuncula A, Idoate MA, Inogés S, de Andrea C, López-Diaz, de Cerio A, Tejada S et al. Neoadjuvant nivolumab modifies the tumor immune microenvironment in re sectable glioblastoma. Nature medicine 2019, 25:470–476.

  135. Nassiri F, Patil V, Yefet LS, Singh O, Liu J, Dang RMA, Yamaguchi TN, Daras M, Cloughesy TF, Colman H, et al. Oncolytic DNX-2401 virotherapy plus pembrolizumab in recurrent gliobla stoma: a phase 1/2 trial. Nat Med. 2023;29:1370–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Duerinck J, Schwarze JK, Awada G, Tijtgat J, Vaeyens F, Bertels C, Geens W, Klein S, Seynaeve L, Cras L, et al. Intracerebral administration of CTLA-4 and PD-1 immune checkpoint bloc king monoclonal antibodies in patients with recurrent glioblastoma: a phase I clinical trial. J Immunother Cancer. 2021;9:e002296.

    Article  PubMed  PubMed Central  Google Scholar 

  137. Dominguez C, McCampbell KK, David JM, Palena C. Neutralization of IL-8 decreases tumor PMN-MDSCs and reduces mesenchymalization of claudin-low triple-negative breast cancer. JCI Insight 2017, 2.

  138. Bilusic M, Heery CR, Collins JM, Donahue RN, Palena C, Madan RA, Karzai F, Marté JL, Strauss J, Gatti-Mays ME, et al. Phase I trial of HuMax-IL8 (BMS-986253), an anti-IL-8 monoclonal antibody, in patients with metastatic or unresectable solid tumors. J Immunother Cancer. 2019;7:240.

    Article  PubMed  PubMed Central  Google Scholar 

  139. Rennard SI, Dale DC, Donohue JF, Kanniess F, Magnussen H, Sutherland ER, Watz H, Lu S, Stryszak P, Rosenberg E, Staudinger H. CXCR2 antagonist MK-7123. A phase 2 proof-of-Concept Trial for Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med. 2015;191:1001–11.

    Article  CAS  PubMed  Google Scholar 

  140. Schott AF, Goldstein LJ, Cristofanilli M, Ruffini PA, McCanna S, Reuben JM, Perez RP, Kato G, Wicha M. Phase ib pilot study to Evaluate Reparixin in Combination with Weekly Paclitaxel in patients with HER-2-Negative metastatic breast Cancer. Clin Cancer Res. 2017;23:5358–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Acker G, Zollfrank J, Jelgersma C, Nieminen-Kelhä M, Kremenetskaia I, Mueller S, Ghori A, Vajkoczy P, Brandenburg S. The CXCR2/CXCL2 signalling pathway - an alternative therapeutic approach in high-grade glioma. Eur J Cancer. 2020;126:106–15.

    Article  CAS  PubMed  Google Scholar 

  142. Herbertz S, Sawyer JS, Stauber AJ, Gueorguieva I, Driscoll KE, Estrem ST, Cleverly AL, Desaiah D, Guba SC, Benhadji KA, et al. Clinical development of galunisertib (LY2157299 monohydrate), a small molecule inhibitor of transforming growth factor-beta signaling pathway. Drug Des Devel Ther. 2015;9:4479–99.

    CAS  PubMed  PubMed Central  Google Scholar 

  143. Wick A, Desjardins A, Suarez C, Forsyth P, Gueorguieva I, Burkholder T, Cleverly AL, Estrem ST, Wang S, Lahn MM, et al. Phase 1b/2a study of galunisertib, a small molecule inhibitor of transforming growth factor-beta receptor I, in combination with standard temozolomide-based radiochemotherapy in patients with newly diagnosed malignant glioma. Invest New Drugs. 2020;38:1570–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Sawyer JS, Anderson BD, Beight DW, Campbell RM, Jones ML, Herron DK, Lampe JW, McCowan JR, McMillen WT, Mort N, et al. Synthesis and activity of new aryl- and heteroaryl-substituted pyrazole inhibitors of the transforming growth factor-beta type I receptor kinase domain. J Med Chem. 2003;46:3953–6.

    Article  CAS  PubMed  Google Scholar 

  145. Brandes AA, Carpentier AF, Kesari S, Sepulveda-Sanchez JM, Wheeler HR, Chinot O, Cher L, Steinbach JP, Capper D, Specenier P, et al. A phase II randomized study of galunisertib monotherapy or galunisertib plus lomustine compared with lomustine monotherapy in patients with recurrent glioblastoma. Neuro Oncol. 2016;18:1146–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Abdelfattah N, Kumar P, Wang C, Leu JS, Flynn WF, Gao R, Baskin DS, Pichumani K, Ijare OB, Wood SL, et al. Single-cell analysis of human glioma and immune cells identifies S100A4 as an immunotherapy target. Nat Commun. 2022;13:767.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Feng YH, Lim SW, Lin HY, Wang SA, Hsu SP, Kao TJ, Ko CY, Hsu TI. Allopregnanolone suppresses glioblastoma survival through decreasing DPYSL3 and S100A11 expression. J Steroid Biochem Mol Biol. 2022;219:106067.

    Article  CAS  PubMed  Google Scholar 

  148. Lu C, Liu J, Yao M, Li L, Li G. Downregulation of S100 calcium binding protein A12 inhibits the growth of glioma cells. BMC Cancer. 2020;20:261.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Zhang S, Guo M, Liu Q, Liu J, Cui Y. Neutrophil extracellular traps induce a hypercoagulable state in glioma. Immun Inflamm Dis. 2021;9:1383–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Shafqat A, Noor Eddin A, Adi G, Al-Rimawi M, Abdul Rab S, Abu-Shaar M, Adi K, Alkattan K, Yaqinuddin A. Neutrophil extracellular traps in central nervous system pathologies: a mini review. Front Med (Lausanne). 2023;10:1083242.

    Article  PubMed  Google Scholar 

  151. Manda-Handzlik A, Demkow U. The Brain Entangled: The Contribution of Neutrophil Extracellular Traps to the Diseases of the Central Nervous System. Cells 2019, 8.

  152. Ocana A, Nieto-Jimenez C, Pandiella A, Templeton AJ. Neutrophils in cancer: prognostic role and therapeutic strategies. Mol Cancer. 2017;16:137.

    Article  PubMed  PubMed Central  Google Scholar 

  153. Chu D, Dong X, Shi X, Zhang C, Wang Z. Neutrophil-based drug Delivery systems. Adv Mater. 2018;30:e1706245.

    Article  PubMed  PubMed Central  Google Scholar 

  154. Wang J, Tang W, Yang M, Yin Y, Li H, Hu F, Tang L, Ma X, Zhang Y, Wang Y. Inflammatory tumor microenvironment responsive neutrophil exosomes-based drug delivery system for targeted glioma therapy. Biomaterials. 2021;273:120784.

    Article  CAS  PubMed  Google Scholar 

  155. Zhang H, Li Z, Gao C, Fan X, Pang Y, Li T, Wu Z, Xie H, He Q. Dual-responsive biohybrid neutrobots for active target delivery. Sci Robot 2021, 6.

  156. Chang Y, Cai X, Syahirah R, Yao Y, Xu Y, Jin G, Bhute VJ, Torregrosa-Allen S, Elzey BD, Won YY, et al. CAR-neutrophil mediated delivery of tumor-microenvironment responsive nanodrugs for glioblastoma chemo-immunotherapy. Nat Commun. 2023;14:2266.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Jayaraml MA, Phillips JJ. Role of the Microenvironment in Glioma Pathogenesis. Annual Rev Pathology-Mechanisms Disease. 2024;19:181–201.

    Article  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by grants from the National Key R&D Program of China, MOST (2023YFC2510000) to Kai Shu, the National Natural Science Foundation of China (82403476) to Hongtao Zhu, the Hubei Association for Science and Technology Young Talents Support Projects (2024) to Hongtao Zhu, the China Postdoctoral Science Foundation (2022M711253) to Hongtao Zhu, Hubei Natural Science Foundation (2023AFB135) to Hongtao Zhu, and the postdoctoral innovation research position funding of Hubei province (2022) to Hongtao Zhu.

Author information

Authors and Affiliations

Authors

Contributions

Jiayi Wen: writing-original manuscript preparation, figure visualization. Dan Liu: writing-original manuscript preparation, reviewing and editing. Hongtao Zhu: writing-reviewing and editing, supervision, funding acquisition. Kai Shu: writing-reviewing and editing, supervision, validation, funding acquisition.

Corresponding authors

Correspondence to Hongtao Zhu or Kai Shu.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

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-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, 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 you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. 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-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wen, J., Liu, D., Zhu, H. et al. Microenvironmental regulation of tumor-associated neutrophils in malignant glioma: from mechanism to therapy. J Neuroinflammation 21, 226 (2024). https://doi.org/10.1186/s12974-024-03222-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12974-024-03222-4

Keywords