Open Access

Tumor‑associated neutrophils: Critical regulators in cancer progression and therapeutic resistance (Review)

  • Authors:
    • Rui Hou
    • Xi Wu
    • Cenzhu Wang
    • Hanfang Fan
    • Yuhan Zhang
    • Hanchi Wu
    • Huiyu Wang
    • Junli Ding
    • Huning Jiang
    • Junying Xu
  • View Affiliations

  • Published online on: February 25, 2025     https://doi.org/10.3892/ijo.2025.5734
  • Article Number: 28
  • Copyright: © Hou et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cancer is the second leading cause of death among humans worldwide. Despite remarkable improvements in cancer therapies, drug resistance remains a significant challenge. The tumor microenvironment (TME) is intimately associated with therapeutic resistance. Tumor‑associated neutrophils (TANs) are a crucial component of the TME, which, along with other immune cells, play a role in tumorigenesis, development and metastasis. In the current review, the roles of TANs in the TME, as well as the mechanisms of neutrophil‑mediated resistance to cancer therapy, including immunotherapy, chemotherapy, radiotherapy and targeted therapy, were summarized. Furthermore, strategies for neutrophil therapy were discussed and TANs were explored as potential targets for cancer treatment. In conclusion, the need to explore the precise roles, recruitment pathways and mechanisms of action of TANs was highlighted for the purpose of developing therapies that precisely target TANs and reverse drug resistance.

Introduction

Cancer is the second leading cause of human mortality worldwide, with >190,000 new cases diagnosed and 100,000 associated deaths occurring each year, accounting for one in six global deaths and representing a significant public health concern (1,2). The current standard of care for cancer involves a multimodal approach that includes surgical resection, radiotherapy, chemotherapy, targeted therapy and immunotherapy (3-5). Advances in surgical techniques and the development of targeted therapeutic agents have led to significant progress in cancer treatment (6). However, emerging drug resistance and metastasis of malignant tumors remain the primary causes of mortality in patients with cancer, representing a significant challenge in the field of oncology (7). >90% of cancer-related deaths are attributed to drug resistance (8,9). Therefore, exploring drug resistance during cancer treatment and identifying effective low-toxicity therapeutic targets that can reverse drug resistance are crucial for reducing adverse effects and improving overall prognosis.

Although the exact mechanisms underlying drug resistance in cancer cells remain elusive, current findings indicate the involvement of numerous genes associated with drug efflux, DNA repair, apoptosis and diverse cellular signaling pathways (10,11). In addition to research asserting that mutations are responsible for tumor development and drug resistance, recent studies have revealed the tumor microenvironment (TME) as an integral part of tumorigenesis. The TME, which refers to the microenvironment surrounding tumor cells, including blood vessels, immune cells, fibroblasts and extracellular matrix (12), is strongly associated with the development of therapeutic resistance in tumor cells through complex signaling pathways (7,13).

Neutrophils were originally considered as first responders of the innate immune system against extracellular pathogens (14). Increasing evidence suggests that neutrophils also play an important role in the TME (15-17). Tumor-associated neutrophils (TANs) are immune cells that infiltrate the TME (18) and act directly or indirectly on tumor cells by releasing a variety of pro-inflammatory factors, immunomodulatory factors and angiogenic factors, which either promote or inhibit tumor occurrence, progression and metastasis (19). In the current article, the role of TANs in tumorigenesis and cancer progression in the TME was summarized, their contribution to therapeutic resistance was explored and existing TAN-targeted therapeutic strategies were reviewed.

TANs and cancer progression

Formation and plasticity of TANs

The migration of neutrophils from bone marrow to the tumor site involves three stages (Fig. 1): Maturation of neutrophils in the bone marrow, circulation in the blood and chemotaxis to the tumor site (20). Increasing research on the body's immune functions have shown that TAN regulation is highly reprogrammable. Neutrophils can acquire different phenotypes based on environmental signals (21); these phenotypic differences determine functional differences among neutrophils and govern whether the cells are involved in pro- and/or anti-tumor responses.

Fridlender et al (22) described a dichotomy in TANs, whereby neutrophils reaching the vicinity of tumors in mice can develop into either anti-tumor N1 cells or pro-tumor N2 cells (23). This choice seems to depend on the production of transforming growth factor β (TGF-β) in the TME, which promotes the generation of pro-tumorigenic N2 cells (22). This differentiation may reflect plasticity rather than the true subtype of neutrophils.

Single-cell RNA sequencing (scRNA-seq) provides an unprecedented view of cellular heterogeneity in the TME. However, owing to the relatively low RNA levels and high RNase levels of neutrophils, neutrophil research is challenging. With the development of scRNA-seq, multiple phenotypes of TANs have been identified in a variety of cancers (19,24,25). Furthermore, pseudotime analysis has revealed differentiation trajectories along neutrophil states (26). In non-small cell lung cancer, Salcher et al (24) identified four TAN subsets (TAN-1 to TAN-4) and showed that the overall TAN phenotype was characterized by high expression of oxidized low-density lipoprotein receptor 1, vascular endothelial growth factor A, CD83, intercellular adhesion molecule 1 and C-X-C motif ligand receptor (CXCR)4 but low expression of CXCR1, CXCR2, prostaglandin-endoperoxide synthase 2, selectin L (CD62L), colony-stimulating factor (CSF)3R and Fc gamma receptor IIIb (CD16B). In pancreatic cancer (27), TANs were divided into five heterogeneous subgroups: A terminally differentiated pro-tumor subpopulation (TAN-1) associated with poor prognosis, an inflammatory subpopulation (TAN-2), a transitional population recently migrated to the TME (TAN-3) and a subpopulation preferentially expressing interferon-stimulated genes (TAN-4). Xue et al (19) stratified patients with liver cancer into five tumor immune microenvironment subtypes, including immune activation, immune suppression mediated by myeloid or stromal cells, immune exclusion and immune residence phenotypes, where differences in the tumor immune microenvironment steer the development of at least six different types of TAN in liver cancer. Furthermore, Wu et al (26) generated and integrated single-cell neutrophil transcriptomes from 17 cancer types and identified 10 distinct states, including inflammation, angiogenesis and antigen presentation. Notably, non-TANs could differentiate into TANs (24,25), indicating that the TME can induce plasticity.

Overall, existing research demonstrates the remarkable plasticity and re-editable nature of TANs, which is an important factor to consider when designing anti-tumor therapies. To date, scRNA-seq has provided reliable support for improved disease outcome prediction and targeted therapy for specific differentiation pathways. However, as different clusters of neutrophils cannot currently be sorted, their functions cannot be verified. Therefore, isolating these cells will be an important step forward in neutrophil research.

Role of TANs in the TME

TANs play a dual role in the TME that depends on the neutrophil phenotype, the timing and the tumor type. TANs participate in pro-tumor inflammation by promoting tumor growth, metastasis and angiogenesis, as well as in remodeling of the extracellular matrix (15). Conversely, TANs can also mediate anti-tumor responses by directly killing tumor cells and participating in cellular networks that mediate anti-tumor resistance (28,29).

Pro-tumor effects

TANs regulate tumor growth and progression. TANs secrete a variety of molecules that can stimulate tumor growth, such as TGF-β, neutrophil elastase (NE), interleukin (IL)-17a, C-C motif chemokine ligand (CCL)2 and IL-8 (20,30-32). Blocking TGF-β shifts TAN polarization from the N2 to N1 phenotype in the TME, increasing tumor cell apoptosis and suppressing tumor cell migration (33,34). In pancreatic cancer, a pro-inflammatory microenvironment can be generated by recruiting TANs and activating NE release, which contributes to the progression of pancreatic neoplasms (35). In gastric cancer, TANs induce the epithelial-to-mesenchymal transition (EMT) of gastric cancer cells by secreting IL-17a and activating Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signaling in gastric cancer cells (36). Furthermore, N2-polarized TANs reportedly promote gastric cancer metastasis through the exosomal miR-4745-5p/3911-mediated inhibition of slit guidance ligand 2 expression (37). In a mouse model of lung adenocarcinoma, TANs exhibit widespread survival and increased expression of the anti-apoptotic protein Bcl-xL to promote tumor growth via granulocyte-macrophage (GM)-CSF-induced JAK/ STAT signaling (38). Another study found that TANs govern tumor progression in lung cancer through an IL-10/STAT3/ programmed death-ligand 1 (PD-L1) feedback signaling loop (39).

Neutrophils are responsible for producing pro-angiogenic factors, including Bv8 (also known as prokineticin 2), matrix metalloproteinase 9 (MMP-9) and VEGF, which play important roles in promoting tumor angiogenesis (40-42). Bv8 is a mediator of myeloid cell-dependent tumor angiogenesis and plays a role in the angiogenic switch by affecting the neoplastic vasculature and infiltration of Gr1+ cells (43). MMP-9 is one of the most important mediators of tumor angiogenesis, with TANs thought to be a major source of MMP-9 (44,45). VEGF produced during the release of MMP-9 specifically stimulates the proliferation of vascular endothelial cells, inhibits the apoptosis of endothelial cells, promotes angiogenesis and increases vascular permeability (46).

Neutrophils can also induce neutrophil extracellular traps (NETs), which are highly expressed in a variety of cancers and promote tumor proliferation, invasion and metastasis (47). Unlike the cell death programs of necrosis and apoptosis, NETs include MMP-9, NE, myeloperoxidase and cytoskeletal proteins (48). NETs contribute to the disruption of normal connections between endothelial cells, enabling tumor cell extravasation and metastasis (49-51). Furthermore, NETs encapsulate tumor cells, protecting them from the cytotoxic effects of surrounding immune cells (52). Although NETs promote tumor recurrence and metastasis in a number of ways, tumor cells, in turn, promote NETs via chemokines (53). In hepatocellular carcinoma (HCC), acetyl-CoA accumulation induces the H3 acetylation-dependent upregulation of CXCL1 gene expression, which can lead to TAN recruitment, NET formation and the promotion of HCC metastasis (54).

Anti-tumor effects

In addition to pro-tumorigenic properties, neutrophils also exhibit anti-tumor responses. However, few studies have reported the anti-tumor effects of neutrophils.

Neutrophils can exert anti-proliferative effects. Direct cell contacts between Fas ligand on human neutrophils and Fas on tumor cells suppresses tumor cells by causing cell cycle arrest in vitro (55). In a mouse model of phosphatase and tensin homolog-deficient uterine cancer, neutrophils impeded early-stage tumor growth and retarded malignant progression by inducing tumor cell detachment from the basement membrane (56).

Neutrophils can kill tumor cells by releasing a series of effector molecules. For instance, reactive oxygen species (ROS) are secreted through neutrophil degranulation, mediating cell killing by opening the transient receptor potential ion channel M2, which leads to an influx of calcium ions into the target cells (57,58). In addition to ROS, TANs also release nitric oxide and tumor necrosis factor (TNF), which are further involved in tumor suppression (59). Neutrophil-derived TNF-related apoptosis-inducing ligand reportedly induces apoptosis of leukemic T cells (60). Furthermore, Cui et al (61) reported that neutrophils release catalytically active neutrophil elastase (ELANE) to kill numerous types of cancer cell both in vitro and in vivo. ELANE proteolytically liberates the CD95 death domain, which interacts with histone H1 isoforms to selectively eradicate cancer cells. ELANE can also attenuate primary tumor growth and produce a CD8+T cell-mediated abscopal effect to attack distant metastases. In melanomas, specific neutrophil subpopulations play an important role in preventing the immune escape of tumor cells, either through the release of inducible NO synthase or the direct phagocytosis of antigen-deficient melanoma cells (62).

Interaction of TANs with other immune cells in the TME

TANs engage in complex bidirectional interactions with macrophages and lymphocytes in the TME through the expression of multiple cytokines, as well as immunosuppressive and stimulatory molecules (63,64).

Macrophages

TANs and tumor-associated macrophages (TAMs) are functional partners in the inflammatory process, hypothesized to synergize and interact in the TME to promote tumor progression through similar molecular forms (65). When macrophages take up TAN-derived factors, they stimulate TAMs toward M1 polarization (66). NETs induce mononuclear macrophages to secrete interleukins, such as IL-1β and IL-6, and recruit progenitor cells of TANs, with activated neutrophils further releasing IL-8, among other cytokines, to recruit macrophages (67). In addition, neutrophils secrete myeloperoxidase, which binds to the mannose receptor and dominates the secretion of GM-CSF in chronic inflammatory environments (68). GM-CSF is an important factor mediating the recruitment and development of TAMs and critical for TME macrophage recruitment and polarization (69,70). In early luminal breast cancers, TAN density correlates with CD163+ M2-like TAM density. Furthermore, TANs are a negative prognostic factor in tumors with an elevated M1/M2 TAM ratio, whereas this impact on patient outcome is lost in tumors with a low M1/M2 ratio (71). Thus, the recruitment and function of TANs and TAMs are inextricably linked. In intrahepatic cholangiocarcinoma (ICC), the interaction between TANs and TAMs produces higher levels of oncostatin M and IL-11, respectively, which then activate STAT3 signaling in ICC cells. STAT3 knockdown attenuates the pro-tumorigenic effects of TANs and TAMs in ICC (63).

Lymphocytes

TANs can lead to the suppression and depletion of T-cell function in several ways; this phenomenon will be elaborated on further below. In addition, activated neutrophils can recruit type 1 T-helper (Th1) and Th17 cells by releasing multiple chemokines (72-74). CCL17 released by TANs may also support tumor growth by promoting the recruitment of regulatory T (Treg) cells to tumors and inhibiting anti-tumor immune activity (75). In pancreatic ductal adenocarcinoma (PDAC), polarized TANs upregulate CCL5 secretion, which promotes cancer cell migration and invasion and enhances Treg cell infiltration in the tumor (76). Furthermore, TANs reduce the cytotoxic and infiltration capacity of natural killer (NK) cells and regulate the expression of programmed cell death protein-1 (PD-1) and PD-L1 through the G-CSF/STAT3 and IL-18 signaling pathways, thereby inhibiting the anti-tumor immune activity of NK cells (77). G-CSF-mobilized neutrophils inhibit NK-cell activation (78-80). By contrast, NK cells control the tumor-promoting and angiogenic functions of neutrophils in an interferon γ-dependent manner by inhibiting VEGF expression (81).

Others

Other components of the TME, such as cancer-associated fibroblasts (CAFs), reportedly secrete IL-8, which further recruit neutrophils into the TME. The infiltrated neutrophils upregulate Serglycin (SRGN) expression in gastric cancer cells via the regenerating family member 4. SRGN secreted by tumor cells then activates the CD44/c-Myc pathway to upregulate Lysine[K]-specific demethylase 5B expression, thereby promoting IL-8 production in CAFs. Thus, the SRGN-IL-8-TANs-SRGN loop facilitates gastric cancer progression (82). In the TME of HCC, CAF-derived cardiotrophin-like cytokine factor 1 increases CXCL6 and TGF-β secretion in tumor cells, which subsequently promotes tumor cell stemness in an autocrine manner, as well as TAN infiltration and polarization in a paracrine manner (83). Furthermore, fibroblast growth factor 19 secreted by tumor cells induces the formation of inflammatory CAFs via the fibroblast growth factor receptor-JAK2-STAT3 pathway, and the release of C5α and IL-1β from inflammatory CAFs promotes the formation of NETs, leading to liver metastases in colorectal cancer (CRC) (84). Other studies have shown that mast cell-derived TNF promotes the extravasation of neutrophils (85).

In summary, TANs exhibit dual pro-tumor and anti-tumor effects. Next, the molecular mechanisms underlying TAN-mediated resistance to cancer therapy will be explored, including resistance to immunotherapy, chemotherapy, targeted therapy and radiotherapy.

TANs and therapeutic resistance

Anti-tumor drugs mainly include chemotherapeutic drugs, molecularly targeted drugs and immune checkpoint inhibitors, with clinical treatment plans based on a combination of drug regimens (86). As TANs can induce resistance to specific drugs through various different mechanisms, the specific mechanisms of TAN-mediated drug resistance have been studied in the context of anti-tumor therapy.

Resistance to immunotherapy

Immunotherapy, which predominantly includes immune checkpoint inhibitors and chimeric antigen receptor (CAR)-T cell therapy, utilizes the immune system of the patient to attack the tumor (87). Neutrophils play an important role in immunotherapy and have a significant impact on the outcome of tumor treatment.

Resistance to anti-PD-1/PD-L1 therapy

Anti-PD-1/ PD-L1 therapies have become an important part of numerous cancer treatments and showed remarkable success. These therapies effectively inhibit tumor growth and metastasis by inhibiting the immune checkpoint molecules PD-1/PD-L1 and restoring the T cell-mediated immune response to the tumor (88-90). However, T-cell depletion, with which TANs are closely associated, is a major impediment to immunotherapy (91-93). Numerous studies have revealed that TANs influence anti-PD-1/PD-L1 therapy primarily by promoting an immunosuppressive TME (94,95). Tumor-secreted nicotinamide phosphoribosyl transferase reprograms CD10+ alkaline phosphatase, biomineralization associated+ neutrophils via neurotrophic tropomyosin receptor kinase 1, maintaining their immaturity and inhibiting their maturation and activation, which induces apparent 'irreversible' exhaustion of T cells in terms of their cell number, frequency and gene profile (96). In HCC, overexpression of CT10 regulator of kinase-like (CRKL) shapes the immunosuppressive TME by recruiting TANs through the upregulation of CRKL/β-catenin/VEGFα and CXCL1 axes. A decrease in the proportion of activated CD8+ T cells was accompanied by an increase of depleted CD8+ T cells in the CRKL overexpression group. PD-L1+ TANs, a potential subset of TANs regulated by CRKL, are significantly upregulated in CRKL-overexpressing tumor tissues, exerting an immune-suppressive effect and resulting in poor patient prognosis. Studies in mice have verified that lymphocyte antigen-6 complex, locus G (Ly6G) restores the efficacy of anti-PD-1 treatment following CRKL overexpression-induced anti-PD-1 resistance, revealing that CRKL regulates PD-1 resistance by mediating the infiltration of TANs in HCC (97). Michaeli et al (98) isolated TANs from different mouse tumor models, which induced apoptosis of CD8+ T cells in the TME via TNF-α and NO contact-dependent mechanisms. Wang et al (99) found that tumors isolated from patients with gastric cancer were infiltrated with CD54+ TANs, expressing high levels of PD-L1. Furthermore, GM-CSF secreted by tumor cells activates TANs and induces PD-L1 expression on TANs through the JAK-STAT3 signaling pathways (99). These activated neutrophils contribute to immunosuppression and cancer progression by inhibiting T-cell immunity in a PD-L1-PD-1-dependent manner. The multiple mechanisms of T-cell exhaustion induced by neutrophils in the TME are shown in Fig. 2.

The immunosuppressive potential of NETs is highlighted by the recent discovery of PD-L1 in NETs. PD-L1 is a ligand that affects adaptive anti-cancer immune responses and metastasis combined with hepatic ischemia or reperfusion by inducing T-cell depletion and dysfunction within the TME in a murine liver model. In a mouse study, treatment with DNase I to digest NETs attenuated tumor growth and increased functional T-cell levels (100). Furthermore, treatment with an adeno-associated virus gene therapy vector expressing DNase I in the liver inhibited liver metastases of CRC by inhibiting neutrophil infiltration and NET formation, as well as restoring local immune responses at the tumor site by increasing the percentage of CD8+ T cells (101). NETs have also recently emerged as powerful modulators of immunotherapy outcomes. In a CRC xenograft model, although the digestion of NETs with DNase I and treatment with PD-1 reduced tumor growth, combination of the two strategies had a synergistic effect. Mechanistically, inhibition of NETs with DNase I reverses resistance to anti-PD-1 blockade by increasing CD8+ T-cell infiltration and cytotoxicity (102).

Resistance to CAR-T therapy

CAR-T therapy has changed the therapeutic landscape for hematological malignancies. Current challenges of CAR-T cell therapy are mainly related to side effects, toxicity, T-cell depletion and a malignant TME (103-105). The most significant difference between hematological and solid tumors is the presence of the TME in solid tumors, with the immunosuppressive nature of the TME being the likely reason why CAR-T cellular immunotherapy has not been successful in solid tumors (106,107). The TME includes highly infiltrating mesenchymal stromal cells, and immunosuppressive cells such as TANs, myeloid-derived suppressor cells (MDSCs), TAMs, mast cells and regulatory T cells (108). All of these cellular components contribute to establishing an immune-suppressive TME capable of interfering with the efficacy of CAR-T cell therapy (109,110). However, the exact mechanism requires further investigation.

Resistance to chemotherapy

Chemotherapy is currently the primary clinical option for tumors, but its efficacy is often limited by drug resistance. This phenomenon results in the failure of chemotherapeutic drugs as well as the development of multidrug resistance, which is the main cause of tumor recurrence, metastasis and death in most patients (111,112). Specifically, TANs can interact with other immune cells or modulate the TME to alter the efficacy of chemotherapy.

NETs

During chemotherapy, neutrophils respond to chemotherapy by infiltrating the TME and releasing NETs, thereby promoting chemoresistance (113,114). Mousset et al (113) reported that upregulation of the CXCL1/5-CXCR2 axis following chemotherapy is involved in the recruitment of neutrophils to metastatic lungs. Eliminating NETs through peptidylarginine deiminase 4 inhibitors or DNase I significantly improved the response to chemotherapy, suggesting a direct causal relationship between NETs and chemoresistance. In terms of the mechanism, NETs promote EMT and chemotherapy resistance by binding and activating TGF-β (a classical inducer of EMT) (113). In addition, neutrophils can induce EMT by releasing NETs (115-118). Specifically, neutrophils induce epithelial stabilization and transcription of zinc finger e-box-binding homeobox 1 by releasing NETs, thus promoting EMT and chemotherapy resistance. Furthermore, lung cancer with breast metastases treated with chemotherapy released CXCL1/5 and IL-1β, which promoted neutrophil recruitment and NET formation, respectively, leading to chemoresistance (119). Another study highlighted the role of neutrophils and NETs in chemotherapy resistance in multiple myeloma, demonstrating for the first time that neutrophils promote cancer cell survival by secreting soluble factors in patients treated with doxorubicin and melphalan (120). Moreover, NETs effectively trap and inhibit the spread of adriamycin in a two-compartment system, which may attenuate its ability to induce apoptosis in ovarian cancer cells (121). The clinical relevance of NETs in chemotherapy resistance has also been demonstrated. Plasma NET levels are significantly higher in patients with metastatic breast cancer exhibiting progressive disease 15 days after chemotherapy (113). The mechanisms through which neutrophils induce chemotherapy resistance by infiltrating the TME and releasing NETs are shown in Fig. 3.

In conclusion, chemotherapy-induced activation of the NET pathway is a major mechanism of tumor chemoresistance.

Chemokines

Chemokines represent a subset of chemoattractant cytokines that control the directed migration of immune cells and play a multifaceted role in tumor cell proliferation, tumor heterogeneity, stemness, senescence, angiogenesis and tumor metastasis (122-124). Increasing evidence has revealed that neutrophil-related chemokines exert a crucial impact on tumor progression and chemotherapy resistance. In the TME, cancer cells regulate neutrophil recruitment to tumor sites through the expression of various chemokine ligands (CXCL1, CXCL2, CXCL5, CXCL6 and CXCL8) for neutrophil receptors CXCR1 and CXCR2 (125,126). Host CXCR2 inhibition by genetic ablation prevents neutrophil accumulation in pancreatic tumors and leads to T cell-dependent suppression of tumor growth (127,128). In a zebrafish model of glioblastoma, CXCR1 mediates the recruitment of neutrophils and supports the proliferation of tumor-initiating astrocytes (129,130); in melanoma-bearing mice, CXCL1 and CXCL2 chemokines enhance neutrophil recruitment and induce angiogenesis (131); in HCC, overexpression of CXCL5 mediates neutrophil infiltration and indicates poor prognosis (132-134), whereas CXCL6 secretion in tumor cells promotes TAN infiltration and polarization to accelerate HCC progression (83); and in chemotaxis assays and mouse models of thyroid cancer, elevated concentrations of CXCL8 promote TAN recruitment and cancer progression (135).

Chemokines play essential roles in TME changes induced by chemotherapy. Apoptotic CRC cells induced by chemotherapy release abundant neutrophil-attracting chemokines, notably CXCL8, thereby attracting neutrophils into the tumor, where their interaction with neighboring macrophages can promote an immunologically unfavorable TME (136). CXCL1 and CXCL2 secretion by metastatic tumor cells recruited neutrophils to the metastatic liver in mouse models of PDAC. These recruited neutrophils expressed growth arrest specific 6 (Gas6), which led to AXL receptor activation on tumor cells, enabling their regrowth. Furthermore, disruption of neutrophil infiltration or inhibition of the Gas6/AXL signaling axis in combination with chemotherapy inhibited metastatic growth (137). To date, growing evidence has revealed that targeting the chemokine/chemokine receptor axis is a promising approach to reverse chemoresistance and improve efficacy. In an orthotopic PDAC model, CXCR2 blockade prevented neutrophil mobilization from the circulation and augmented chemotherapeutic efficacy. Targeting both CXCR2+ TANs and CCR2+ macrophages disrupted myeloid recruitment and improved the response to FOLFIRINOX chemotherapy in PDAC (138). Overexpression of CXCL1/2 in breast cancer led to metastasis and resistance to chemotherapy in a paracrine manner involving the TME. However, CXCR2 blockade inhibited this vicious cycle, increasing the efficacy of chemotherapy against breast cancer (139). Reparixin, a small-molecule inhibitor of the CXCL8-CXCR1/2 axis, offers the possibility of chemotherapy-induced synergy in breast cancer. For example, a combination of reparixin with paclitaxel reduced brain metastasis as well as the population of cancer stem-like cells (140). Sequential treatment with first-line and second-line chemotherapy and reparixin inhibited tumor growth, reduced toxicity and prolonged survival in mouse models of gastric cancer (141). In addition, a CXCR2-specific small-molecule inhibitor, SB225002, decreased neutrophil infiltration and reduced tumor growth in lung cancer (142).

Resistance to targeted therapy

In recent years, advances in molecular biology and genetics research have shown that malignant tumors exhibit complex and specific biological defects, including oncogenes, oncogene mutations and chromatin modifications (143). Targeted therapy utilizes the specific structural molecules of tumor tissues or cells and drugs that specifically bind to them to precisely kill tumor cells (144). However, primary or acquired resistance limits their clinical use. In this chapter, an overview of the mechanisms of resistance associated with TANs in common molecular targeted therapies is presented.

VEGF

The occurrence and development of tumors depend on tumor angiogenesis, which provides oxygen and nutrients to tumor cells, removes metabolic waste and also enables tumor cells to metastasize (145). Angiogenic vessels contain irregular branching and form intermittent arteriovenous shunts, leading to discontinuous perfusion and disturbed blood flow patterns, resulting in an environment with abnormally high interstitial fluid pressure (IFP) (146).

VEGF has been identified as a key cytokine involved in tumor angiogenesis and metastasis (147). TANs release NE and MMP-9 to degrade the extracellular matrix and later activate VEGF, thus promoting angiogenesis in the TME (15,20,46). MMP-9 reportedly promotes the release of VEGF or inhibits the action of anti-angiogenic factors, exerting a key role in angiogenesis in breast cancer (23,148). In addition to MMP-9, G-CSF released by tumor and stromal cells upregulated the angiogenic peptide Bv8 on neutrophils, which promoted angiogenesis by mediating endothelial cell proliferation (149,150). TANs residing in hypoxic scars may contribute to distorted blood flow and a high IFP environment through MMP-9, VEGF and Bv8 (151). High IFP can be a barrier to both the effective delivery of anti-cancer drugs toward the TME or drug accumulation within the tumor area, thus promoting tumor resistance to therapy (152) (Fig. 4).

TANs can also promote angiogenesis through the direct secretion of IL-17 or indirect activation of the nucleotide-binding domain, leucine-rich repeat and pyrin domain-containing protein 3 (NLRP3), leading to increased IL-1β secretion (153). In mouse tumor models, treatment with gemcitabine plus 5-fluorouracil induced the release of proteinase B from TANs and MDSCs, which in turn led to an IL-1β-dependent increase in IL-17 production and angiogenic blood vessel formation through the action of NLRP3 (154). Furthermore, neutrophil-derived Bv8 is associated with resistance to anti-VEGF therapy, whereas inhibition of G-CSF may increase the efficacy of anti-VEGF therapy (149,153).

Overall, these studies support the role of neutrophils in the initial angiogenic switch during tumorigenesis and reveal the pathways through which TANs may reduce the efficacy of anti-angiogenic therapy.

Epidermal growth factor receptor (EGFR)

EGFR-targeting drugs typically interfere with activation of the EGFR signaling pathway (155). When EGFR binds to its ligands, it activates numerous downstream signaling pathways, including Ras-Raf-MEK-Erk, PI3K-AKT-mTOR and STAT, thus promoting cell proliferation, growth, angiogenesis and metastasis (156-158). Experiments on A549 cells using an in vitro co-culture system have shown that elastase secreted by neutrophils stimulates proteinase-activated receptor 2 and induces EGFR trans-activation to promote drug resistance (159).

Human EGFR 2 (HER-2)

HER-2 is associated with poor prognosis in numerous cancers but predominantly breast cancer. Drugs targeting HER-2 mainly fall into one of three categories: Monoclonal antibodies, including trastuzumab and pertuzumab; tyrosine kinase inhibitors, including lapatinib and eratinib; and antibody-drug conjugates (160) such as T-DM1. The results of an autocrine model showed that NE splits cell surface EGF or TGF-α from the cell membrane to activate signal transduction (161). TGF-α not only suppresses HER-2 downregulation by disrupting endocytosis and lysosome function, but also recruits HER-2 on the cell surface (162) and may affect the therapeutic effect of targeting HER-2. Furthermore, combining the CXCR1/2 inhibitor SCH563705 with lapatinib reduced cancer stem-like cell activity compared with either treatment alone in HER2-positive breast cancer via a novel SRC and EGFR/ HER2-dependent mechanism (163).

Kirsten rat sarcoma viral oncogene (KRAS)

KRAS mutations have been implicated in ~40% of CRC cases, as well as in numerous other types of human cancers, such as lung cancer, breast cancer and prostate cancer (164-167). Exosomal KRAS mutants exert stimulatory effects on IL-8 production and NET formation to promote the growth of CRC cells (168). Furthermore, when combined with anti-VEGF, neutralizing G-CSF activity and G-CSF-induced CD11b+Ly6G+ neutrophils is effective in reducing tumor growth and increasing survival in KRAS-driven PDAC, as indicated in a mouse model with knockout of pancreatic epithelial-specific TGFβ receptor type II and activated KRAS (169).

Resistance to radiotherapy

Radiotherapy is one of the most effective approaches for achieving tumor control (170). Nearly two-thirds of patients with cancer are treated with radiotherapy, often with the intent to achieve complete and permanent tumor regression (local control) (171). However, innate or acquired radiotherapy resistance remains a significant challenge that markedly limits the therapeutic effects, leading to cancer relapse and poor prognosis (172,173). Several crucial aspects contribute to radiotherapy resistance, including radiation-induced DNA damage repair, apoptosis escape, cell-cycle arrest, abundance of cancer stem cells, modification of cancer cells and their microenvironment, metabolic reprogramming, presence of exosomal and non-coding RNA and ferroptosis (174-177).

Studies have shown that high neutrophil infiltration is associated with poor response to radiotherapy (20,171,178). Neutrophils promote radiotherapy resistance in various malignant tumors. For instance, in an irradiated glioblastoma model, Ly6G+ inflammatory cells promoted the conversion of glioblastoma cells to glioblastoma stem cells through the NOS2-NO-ID4 regulatory axis. Treatment with Ly6G-neutralizing antibodies reduced the number of glioblastoma stem cells and prolonged survival in tumor-bearing mice after radiotherapy (179). In a model of local-regional failure for breast cancer after irradiation, high expression of ectonucleotide pyrophosphatase/phosphodiesterase 1 in circulating tumor cells enhanced the expression of haptoglobin, resulting in neutrophil infiltration, NET formation and tumor relapse (180). Ancey et al (181) showed that glucose transporter 1 (GLUT1) expression in TANs promotes lung cancer growth and resistance to radiotherapy in a mouse model of lung adenocarcinoma. Loss of GLUT1 accelerates neutrophil turnover in tumors and reduces a subset of TANs expressing sialic acid-binding immunoglobulin-like lectin F. In the absence of GLUT1 expression by TANs, tumor growth is diminished and the efficacy of radiotherapy is augmented (181). Nolan et al (170) found that off-target exposure to radiation promotes the formation of a premetastatic niche by neutrophils in a mouse model of breast cancer lung metastasis. By preventing neutrophil-dependent Notch activation by blocking degranulation, radiation-enhanced metastases are significantly offset (170). In bladder cancer, radiation induces cancer cells to release high mobility group box 1 (HMGB1), which promotes the formation of NETs through Toll-like receptor 4 signaling; subsequent inhibition of HMGB1 and NETs improved the overall radiotherapy response in mouse models (182). Furthermore, in an autochthonous mouse model of soft tissue sarcoma, neutrophil depletion prior to image-guided focal irradiation improved tumor response to radiotherapy. According to scRNA-seq, tumor radiosensitization by neutrophil depletion after radiotherapy is associated with the downregulation of oncogenic transcriptional programs (171).

Furthermore, Li et al (183) proposed an innovative approach that integrated peroxynitrite (ONOO)-mediated radiosensitization with TAN polarization (reprogramming of TANs from N2 to N1 phenotypes) to reverse an immunosuppressive TME, markedly amplifying the potency of radiotherapy in metastatic CRC. In conclusion, targeting neutrophils represents a potential therapeutic strategy for modulating the efficacy of radiotherapy.

TAN-targeted therapeutic strategies

Given the impact of TANs on tumors, targeting and regulating TANs in the TME represents a promising new therapeutic approach.

Targeted neutrophil therapy

Targeted neutrophil therapy focuses on inhibiting tumor formation, metastasis and angiogenesis by inhibiting the polarization and recruitment of neutrophils in the TME, preventing the formation and aggregation of TANs (18,20,184). Several clinical trials of neutrophil-targeted tumor therapy are currently underway (Table I). In another study, antibody-dependent cell-mediated cytotoxicity (ADCC) antibody therapy with monoclonal antibodies (mAbs) was applied to enhance the ADCC potential of TANs. The results showed that, for two different tumor targets, EGFR and HER-2, a combination of IgG and IgA mAbs is more cytotoxic than the antibodies alone (185-187). In addition, as reported by Kumbhojkar et al (188), micropatch-loaded neutrophils provide a potent, scalable and drug-free approach to neutrophil-based cancer immunotherapy.

Table I

Clinical trials based on neutrophil-targeted tumor therapy.

Table I

Clinical trials based on neutrophil-targeted tumor therapy.

Target/drugCancer applicationsNCT no.
CXCR1/CXCR2 inhibitors
 ReparixinBreast cancerNCT02370238
Breast cancerNCT02001974
Breast cancerNCT01861054
 AZD5069Metastatic castration-resistant prostate cancerNCT03177187
TGF-β pathway inhibitors
 GalunisertibBreast cancerNCT02672475
GlioblastomaNCT01582269
GliomaNCT01682187
CD47-SIRPα inhibitors
 TTI-621Hematologic cancersNCT02663518
 IBI188Advanced malignanciesNCT03717103
 CC-90002Solid and hematologic cancersNCT02367196
 Hu5F9-G4Solid tumorsNCT02216409
TRAIL receptor 2 agonists
 TigatuzumabBreast cancerNCT01307891
 CS-1008Metastatic colorectal cancerNCT01220999

[i] TGF-β, transforming growth factor-β; SIRPα, signal-regulatory protein α; TRAIL, tumor necrosis factor-related apoptosis-inducing ligand; NCT, national clinical trials registry.

Biomarkers for potential tumor therapies

High infiltration of TANs is associated with poor prognosis in most human tumors. Neutrophils infiltrate to varying degrees, as assessed by routine immunohistochemical staining of neutrophil markers (CD66b in humans and Ly6G in mice) and neutrophil transcriptional profiles of solid tumors (20). Although TANs are a poor prognostic indicator of survival in a number of malignant tumors, such as HCC, cholangiocarcinoma, head and neck cancer and renal cancer (27,189,190), certain studies have found that TANs can improve the survival rate of patients with colon cancer (191,192).

The neutrophil-to-lymphocyte ratio (NLR) is valuable for determining the prognosis of patients with cancer, with a high NLR associated with poor prognosis in patients with colorectal, pancreatic, gastric and breast cancers (193-195). A large-scale meta-analysis of 8,500 patients with breast cancer found that a high NLR (1.9 to 5.0) was strongly associated with poor overall survival and reduced disease-free survival (196). In patients with CRC and liver metastases who underwent hepatic lobectomy, an elevated NLR was the only positive predictor of postoperative recurrence and was positively correlated with tumor recurrence but negatively correlated with postoperative survival in patients who underwent in situ liver transplantation for primary liver cancer (197,198). Furthermore, an elevated NLR during the postoperative follow-up period was an independent risk factor for shorter survival in a large number of patients with gastric cancer who underwent gastric resection (199,200).

Neutrophils have been reported to express ligand-activated immune checkpoints on T cells. For instance, PD-L1-expressing neutrophils have prognostic significance in both HCC and gastric cancer (99). Zhou et al (134) also demonstrated that intra-tumor neutrophils express high levels of CCL2 and CCL17, which correlate with disease progression and prognosis. The number of CCL2+ and CCL17+ TANs is also positively associated with tumor size, microvascular invasion, level of tumor differentiation and stage (201). In summary, TANs are potential biomarkers for tumor therapy.

Conclusions

TANs regulate tumorigenesis and progression by i) regulating the function of other immune cells, controlling NET formation and affecting the polarization state, and ii) mediating resistance to tumor therapy in various ways. However, the existence and interaction of different immune cell subpopulations in the TME, the different immune characteristics of the TME in different cancer types and individual patients, and the depletion of TANs, which can lead to a reduction of organism immunity, are urgent clinical problems that must be addressed. Combining existing effective tumor therapies with neutrophil-targeted therapies may represent a safer and more effective way to overcome tumor drug resistance. Future research should further investigate the exact roles, recruitment pathways and mechanisms of action of TANs to develop therapies that precisely target TANs and counter drug resistance.

Availability of data and materials

Not applicable.

Authors' contributions

RH, HJ, HYW, JD and JX conceived the study. RH and HJ prepared the original draft of the manuscript and drew the figures. XW, CW, HF, YZ and HCW revised the manuscript. JX and HJ supervised and approved the final manuscript. Data authentication is not applicable. All the authors have read and agreed to the published version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Abbreviations:

TME

tumor microenvironment

TANs

tumor-associated neutrophils

TGF-β

transforming growth factor β

scRNA-seq

single-cell RNA sequencing

NE

neutrophil elastase

IL

interleukin

EMT

epithelial-mesenchymal transition

JAK2/STAT3

janus kinase 2/signal transducer and activator of transcription 3

GM-CSF

granulocyte-macrophage colony-stimulating factor

PD-L1

programmed death-ligand 1

MMP-9

matrix metalloproteinase 9

VEGF

vascular endothelial growth factor

NETs

neutrophil extracellular traps

HCC

hepatocellular carcinoma

ROS

reactive oxygen species

TNF

tumor necrosis factor

ELANE

neutrophils released catalytically active neutrophil elastase

TAMs

tumor-associated macrophages

ICC

intrahepatic cholangiocarcinoma

PDAC

pancreatic ductal adenocarcinoma

PD-1

programmed cell death protein-1

CAFs

cancer-associated fibroblasts

SRGN

serglycin

CRKL

CT10 regulator of kinase-like

Ly6G

lymphocyte antigen-6 complex, locus G

CRC

colorectal cancer

MDSCs

myeloid-derived suppressor cells

Gas6

expressed growth arrest specific 6

IFP

interstitial fluid pressure

NLRP3

pyrin domain-containing protein 3

EGFR

epidermal growth factor receptor

HER-2

human EGFR 2

TGF-α

transforming growth factor-α

KRAS

Kirsten rat sarcoma viral oncogene

GLUT1

glucose transporter 1

HMGB1

high-mobility group box 1

ADCC

antibody-dependent cell-mediated cytotoxicity

NLR

neutrophil-to-lymphocyte ratio

Acknowledgments

Not applicable.

Funding

This work was supported by grants from the Cohort and Clinical Research Program of Wuxi Medical Center, Nanjing Medical University (grant no. WXKY202302013), 'Taihu Light' Technology Research Program of Wuxi Science and Technology Bureau (grant no. K20241014), the General Program of Wuxi Medical Center, Nanjing Medical University (grant nos. WMCG202319, WMCG202352, WMCG202353 and WMCG202354), the Doctoral Talent Fund of the Affiliated Wuxi People's Hospital of Nanjing Medical University (grant nos. BSRC202207, BSRC202303 and BSRC202309) and the Scientific Research Program of Wuxi Health Commission (grant nos. BJ2023022 and Q202451).

References

1 

Jassim A, Rahrmann EP, Simons BD and Gilbertson RJ: Cancers make their own luck: Theories of cancer origins. Nat Rev Cancer. 23:710–724. 2023. View Article : Google Scholar : PubMed/NCBI

2 

Mattiuzzi C and Lippi G: Cancer statistics: A comparison between World Health Organization (WHO) and Global Burden of Disease (GBD). Eur J Public Health. 30:1026–1027. 2020. View Article : Google Scholar

3 

Wang J, Yang J, Narang A, He J, Wolfgang C, Li K and Zheng L: Consensus, debate, and prospective on pancreatic cancer treatments. J Hematol Oncol. 17:922024. View Article : Google Scholar : PubMed/NCBI

4 

Long GV, Swetter SM, Menzies AM, Gershenwald JE and Scolyer RA: Cutaneous melanoma. Lancet. 402:485–502. 2023. View Article : Google Scholar : PubMed/NCBI

5 

Joshi SS and Badgwell BD: Current treatment and recent progress in gastric cancer. CA Cancer J Clin. 71:264–279. 2021. View Article : Google Scholar : PubMed/NCBI

6 

de Visser KE and Joyce JA: The evolving tumor microenvironment: From cancer initiation to metastatic outgrowth. Cancer Cell. 41:374–403. 2023. View Article : Google Scholar : PubMed/NCBI

7 

Gonçalves AC, Richiardone E, Jorge J, Polónia B, Xavier CPR, Salaroglio IC, Riganti C, Vasconcelos MH, Corbet C and Sarmento-Ribeiro AB: Impact of cancer metabolism on therapy resistance-clinical implications. Drug Resist Updat. 59:1007972021. View Article : Google Scholar

8 

Kalli M, Poskus MD, Stylianopoulos T and Zervantonakis IK: Beyond matrix stiffness: Targeting force-induced cancer drug resistance. Trends Cancer. 9:937–954. 2023. View Article : Google Scholar : PubMed/NCBI

9 

Chen D, Gu X, Nurzat Y, Xu L, Li X, Wu L, Jiao H, Gao P, Zhu X, Yan D, et al: Writers, readers, and erasers RNA modifications and drug resistance in cancer. Mol Cancer. 23:1782024. View Article : Google Scholar : PubMed/NCBI

10 

He J, Qiu Z, Fan J, Xie X, Sheng Q and Sui X: Drug tolerant persister cell plasticity in cancer: A revolutionary strategy for more effective anticancer therapies. Signal Transduct Target Ther. 9:2092024. View Article : Google Scholar : PubMed/NCBI

11 

Nussinov R, Tsai C-J and Jang H: Anticancer drug resistance: An update and perspective. Drug Resist Updat. 59:1007962021. View Article : Google Scholar : PubMed/NCBI

12 

Polak R, Zhang ET and Kuo CJ: Cancer organoids 2.0: Modelling the complexity of the tumour immune microenvironment. Nat Rev Cancer. 24:523–539. 2024. View Article : Google Scholar : PubMed/NCBI

13 

Hessmann E, Buchholz SM, Demir IE, Singh SK, Gress TM, Ellenrieder V and Neesse A: Microenvironmental determinants of pancreatic cancer. Physiol Rev. 100:1707–1751. 2020. View Article : Google Scholar : PubMed/NCBI

14 

Koenderman L and Vrisekoop N: Neutrophils in cancer: From biology to therapy. Cell Mol Immunol. 22:4–23. 2025. View Article : Google Scholar :

15 

Liu S, Wu W, Du Y, Yin H, Chen Q, Yu W, Wang W, Yu J, Liu L, Lou W and Pu N: The evolution and heterogeneity of neutrophils in cancers: Origins, subsets, functions, orchestrations and clinical applications. Mol Cancer. 22:1482023. View Article : Google Scholar : PubMed/NCBI

16 

Zhang J, Gu J, Wang X, Ji C, Yu D, Wang M, Pan J, Santos HA, Zhang H and Zhang X: Engineering and targeting neutrophils for cancer therapy. Adv Mater. 36:e23103182024. View Article : Google Scholar : PubMed/NCBI

17 

van Vlerken-Ysla L, Tyurina YY, Kagan VE and Gabrilovich DI: Functional states of myeloid cells in cancer. Cancer Cell. 41:490–504. 2023. View Article : Google Scholar : PubMed/NCBI

18 

Que H, Fu Q, Lan T, Tian X and Wei X: Tumor-associated neutrophils and neutrophil-targeted cancer therapies. Biochim Biophys Acta Rev Cancer. 1877:1887622022. View Article : Google Scholar : PubMed/NCBI

19 

Xue R, Zhang Q, Cao Q, Kong R, Xiang X, Liu H, Feng M, Wang F, Cheng J, Li Z, et al: Liver tumour immune microenvironment subtypes and neutrophil heterogeneity. Nature. 612:141–147. 2022. View Article : Google Scholar : PubMed/NCBI

20 

Jaillon S, Ponzetta A, Di Mitri D, Santoni A, Bonecchi R and Mantovani A: Neutrophil diversity and plasticity in tumour progression and therapy. Nat Rev Cancer. 20:485–503. 2020. View Article : Google Scholar : PubMed/NCBI

21 

Mantovani A, Cassatella MA, Costantini C and Jaillon S: Neutrophils in the activation and regulation of innate and adaptive immunity. Nat Rev Immunol. 11:519–531. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Fridlender ZG, Sun J, Kim S, Kapoor V, Cheng G, Ling L, Worthen GS and Albelda SM: Polarization of tumor-associated neutrophil phenotype by TGF-beta: 'N1' versus 'N2' TAN. Cancer Cell. 16:183–194. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Shaul ME and Fridlender ZG: Tumour-associated neutrophils in patients with cancer. Nat Rev Clin Oncol. 16:601–620. 2019. View Article : Google Scholar : PubMed/NCBI

24 

Salcher S, Sturm G, Horvath L, Untergasser G, Kuempers C, Fotakis G, Panizzolo E, Martowicz A, Trebo M, Pall G, et al: High-resolution single-cell atlas reveals diversity and plasticity of tissue-resident neutrophils in non-small cell lung cancer. Cancer Cell. 40:1503–1520.e8. 2022. View Article : Google Scholar : PubMed/NCBI

25 

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. 383:eadf64932024. View Article : Google Scholar : PubMed/NCBI

26 

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. 187:1422–1439.e24. 2024. View Article : Google Scholar : PubMed/NCBI

27 

Wang L, Liu Y, Dai Y, Tang X, Yin T, Wang C, Wang T, Dong L, Shi M, Qin J, et al: Single-cell RNA-seq analysis reveals BHLHE40-driven pro-tumour neutrophils with hyperactivated glycolysis in pancreatic tumour microenvironment. Gut. 72:958–971. 2023. View Article : Google Scholar

28 

Xia L, Oyang L, Lin J, Tan S, Han Y, Wu N, Yi P, Tang L, Pan Q, Rao S, et al: The cancer metabolic reprogramming and immune response. Mol Cancer. 20:282021. View Article : Google Scholar : PubMed/NCBI

29 

Tian S, Chu Y, Hu J, Ding X, Liu Z, Fu D, Yuan Y, Deng Y, Wang G, Wang L and Wang Z: Tumour-associated neutrophils secrete AGR2 to promote colorectal cancer metastasis via its receptor CD98hc-xCT. Gut. 71:2489–2501. 2022. View Article : Google Scholar : PubMed/NCBI

30 

Tie Y, Tang F, Wei YQ and Wei XW: Immunosuppressive cells in cancer: Mechanisms and potential therapeutic targets. J Hematol Oncol. 15:612022. View Article : Google Scholar : PubMed/NCBI

31 

Lianyuan T, Gang L, Ming T, Dianrong X, Chunhui Y, Zhaolai M and Bin J: Tumor associated neutrophils promote the metastasis of pancreatic ductal adenocarcinoma. Cancer Biol Ther. 21:937–945. 2020. View Article : Google Scholar : PubMed/NCBI

32 

Amorim C, Docasar CL, Guimarães-Bastos D, Frony AC, Barja-Fidalgo C, Renovato-Martins M and Moraes JA: Extracellular vesicles derived from MDA-MB-231 cells trigger neutrophils to a pro-tumor profile. Cells. 11:18752022. View Article : Google Scholar : PubMed/NCBI

33 

Qin F, Liu X, Chen J, Huang S, Wei W, Zou Y, Liu X, Deng K, Mo S, Chen J, et al: Anti-TGF-β attenuates tumor growth via polarization of tumor associated neutrophils towards an anti-tumor phenotype in colorectal cancer. J Cancer. 11:2580–2592. 2020. View Article : Google Scholar :

34 

Peng H, Shen J, Long X, Zhou X, Zhang J, Xu X, Huang T, Xu H, Sun S, Li C, et al: Local release of TGF-β inhibitor modulates tumor-associated neutrophils and enhances pancreatic cancer response to combined irreversible electroporation and immunotherapy. Adv Sci (Weinh). 9:e21052402022. View Article : Google Scholar

35 

Tan Q, Ma X, Yang B, Liu Y, Xie Y, Wang X, Yuan W and Ma J: Periodontitis pathogen Porphyromonas gingivalis promotes pancreatic tumorigenesis via neutrophil elastase from tumor-associated neutrophils. Gut Microbes. 14:20737852022. View Article : Google Scholar : PubMed/NCBI

36 

Li S, Cong X, Gao H, Lan X, Li Z, Wang W, Song S, Wang Y, Li C, Zhang H, et al: Tumor-associated neutrophils induce EMT by IL-17a to promote migration and invasion in gastric cancer cells. J Exp Clin Cancer Res. 38:62019. View Article : Google Scholar : PubMed/NCBI

37 

Zhang J, Yu D, Ji C, Wang M, Fu M, Qian Y and Zhang X, Ji R, Li C, Gu J and Zhang X: Exosomal miR-4745-5p/3911 from N2-polarized tumor-associated neutrophils promotes gastric cancer metastasis by regulating SLIT2. Mol Cancer. 23:1982024. View Article : Google Scholar : PubMed/NCBI

38 

Bodac A, Mayet A, Rana S, Pascual J, Bowler AD, Roh V, Fournier N, Craciun L, Demetter P, Radtke F and Meylan E: Bcl-xL targeting eliminates ageing tumor-promoting neutrophils and inhibits lung tumor growth. EMBO Mol Med. 16:158–184. 2024. View Article : Google Scholar : PubMed/NCBI

39 

Zhang S, Sun L, Zuo J and Feng D: Tumor associated neutrophils governs tumor progression through an IL-10/STAT3/PD-L1 feedback signaling loop in lung cancer. Transl Oncol. 40:1018662024. View Article : Google Scholar

40 

Huang X, Nepovimova E, Adam V, Sivak L, Heger Z, Valko M, Wu Q and Kuca K: Neutrophils in cancer immunotherapy: Friends or foes? Mol Cancer. 23:1072024. View Article : Google Scholar : PubMed/NCBI

41 

Bird L: Neutrophils become pro-angiogenic in tumours. Nat Rev Immunol. 24:1572024. View Article : Google Scholar : PubMed/NCBI

42 

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 brain tumors. Cell. 186:4546–4566.e27. 2023. View Article : Google Scholar : PubMed/NCBI

43 

Qu X, Zhuang G, Yu L, Meng G and Ferrara N: Induction of Bv8 expression by granulocyte colony-stimulating factor in CD11b+Gr1+ cells: Key role of Stat3 signaling. J Biol Chem. 287:19574–19584. 2012. View Article : Google Scholar : PubMed/NCBI

44 

Fetz AE, Radic MZ and Bowlin GL: Neutrophils in biomaterial-guided tissue regeneration: Matrix reprogramming for angiogenesis. Tissue Eng Part B Rev. 27:95–106. 2021. View Article : Google Scholar

45 

Vannitamby A, Seow HJ, Anderson G, Vlahos R, Thompson M, Steinfort D, Irving LB and Bozinovski S: Tumour-associated neutrophils and loss of epithelial PTEN can promote corticosteroid-insensitive MMP-9 expression in the chronically inflamed lung microenvironment. Thorax. 72:1140–1143. 2017. View Article : Google Scholar : PubMed/NCBI

46 

Mizuno R, Kawada K, Itatani Y, Ogawa R, Kiyasu Y and Sakai Y: The role of tumor-associated neutrophils in colorectal cancer. Int J Mol Sci. 20:5292019. View Article : Google Scholar : PubMed/NCBI

47 

Wang Y, Liu F, Chen L, Fang C, Li S, Yuan S, Qian X, Yin Y, Yu B, Fu B, et al: Neutrophil extracellular traps (NETs) promote non-small cell lung cancer metastasis by suppressing lncRNA MIR503HG to activate the NF-κB/NLRP3 inflammasome pathway. Front Immunol. 13:8675162022. View Article : Google Scholar

48 

Adrover JM, McDowell SAC, He XY, Quail DF and Egeblad M: NETworking with cancer: The bidirectional interplay between cancer and neutrophil extracellular traps. Cancer Cell. 41:505–526. 2023. View Article : Google Scholar : PubMed/NCBI

49 

Chu C, Wang X, Yang C, Chen F, Shi L, Xu W, Wang K, Liu B, Wang C, Sun D and Ding W: Neutrophil extracellular traps drive intestinal microvascular endothelial ferroptosis by impairing Fundc1-dependent mitophagy. Redox Biol. 67:1029062023. View Article : Google Scholar : PubMed/NCBI

50 

Zheng F, Ma L, Li X, Wang Z, Gao R, Peng C, Kang B, Wang Y, Luo T, Wu J, et al: Neutrophil extracellular traps induce glomerular endothelial cell dysfunction and pyroptosis in diabetic kidney disease. Diabetes. 71:2739–2750. 2022. View Article : Google Scholar : PubMed/NCBI

51 

Ngo AT, Skidmore A, Oberg J, Yarovoi I, Sarkar A, Levine N, Bochenek V, Zhao G, Rauova L, Kowalska MA, et al: Platelet factor 4 limits neutrophil extracellular trap- and cell-free DNA-induced thrombogenicity and endothelial injury. JCI Insight. 8:e1710542023. View Article : Google Scholar : PubMed/NCBI

52 

Teijeira Á, Garasa S, Gato M, Alfaro C, Migueliz I, Cirella A, de Andrea C, Ochoa MC, Otano I, Etxeberria I, et al: CXCR1 and CXCR2 chemokine receptor agonists produced by tumors induce neutrophil extracellular traps that interfere with immune cytotoxicity. Immunity. 52:856–871.e8. 2020. View Article : Google Scholar : PubMed/NCBI

53 

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

54 

Pan JJ, Xie SZ, Zheng X, Xu JF, Xu H, Yin RQ, Luo YL, Shen L, Chen ZR, Chen YR, et al: Acetyl-CoA metabolic accumulation promotes hepatocellular carcinoma metastasis via enhancing CXCL1-dependent infiltration of tumor-associated neutrophils. Cancer Lett. 592:2169032024. View Article : Google Scholar : PubMed/NCBI

55 

Sun B, Qin W, Song M, Liu L, Yu Y, Qi X and Sun H: neutrophil suppresses tumor cell proliferation via fas/fas ligand pathway mediated cell cycle arrested. Int J Biol Sci. 14:2103–2113. 2018. View Article : Google Scholar :

56 

Blaisdell A, Crequer A, Columbus D, Daikoku T, Mittal K, Dey SK and Erlebacher A: Neutrophils oppose uterine epithelial carcinogenesis via debridement of hypoxic tumor cells. Cancer Cell. 28:785–799. 2015. View Article : Google Scholar : PubMed/NCBI

57 

Gershkovitz M, Caspi Y, Fainsod-Levi T, Katz B, Michaeli J, Khawaled S, Lev S, Polyansky L, Shaul ME, Sionov RV, et al: TRPM2 mediates neutrophil killing of disseminated tumor cells. Cancer Res. 78:2680–2690. 2018. View Article : Google Scholar : PubMed/NCBI

58 

Li Y, Wu S, Zhao Y, Dinh T, Jiang D, Selfridge JE, Myers G, Wang Y, Zhao X, Tomchuck S, et al: Neutrophil extracellular traps induced by chemotherapy inhibit tumor growth in murine models of colorectal cancer. J Clin Invest. 134:e1750312024. View Article : Google Scholar : PubMed/NCBI

59 

Antuamwine BB, Bosnjakovic R, Hofmann-Vega F, Wang X, Theodosiou T, Iliopoulos I and Brandau S: N1 versus N2 and PMN-MDSC: A critical appraisal of current concepts on tumor-associated neutrophils and new directions for human oncology. Immunol Rev. 314:250–279. 2023. View Article : Google Scholar

60 

Koga Y, Matsuzaki A, Suminoe A, Hattori H and Hara T: Neutrophil-derived TNF-related apoptosis-inducing ligand (TRAIL): A novel mechanism of antitumor effect by neutrophils. Cancer Res. 64:1037–1043. 2004. View Article : Google Scholar : PubMed/NCBI

61 

Cui C, Chakraborty K, Tang XA, Zhou G, Schoenfelt KQ, Becker KM, Hoffman A, Chang YF, Blank A, Reardon CA, et al: Neutrophil elastase selectively kills cancer cells and attenuates tumorigenesis. Cell. 184:3163–3177.e21. 2021. View Article : Google Scholar : PubMed/NCBI

62 

Hirschhorn D, Budhu S, Kraehenbuehl L, Gigoux M, Schröder D, Chow A, Ricca JM, Gasmi B, De Henau O, Mangarin LMB, et al: T cell immunotherapies engage neutrophils to eliminate tumor antigen escape variants. Cell. 186:1432–1447.e17. 2023. View Article : Google Scholar : PubMed/NCBI

63 

Zhou Z, Wang P, Sun R, Li J, Hu Z, Xin H, Luo C, Zhou J, Fan J and Zhou S: Tumor-associated neutrophils and macrophages interaction contributes to intrahepatic cholangiocarcinoma progression by activating STAT3. J Immunother Cancer. 9:e0019462021. View Article : Google Scholar : PubMed/NCBI

64 

Singhal S, Rao AS, Stadanlick J, Bruns K, Sullivan NT, Bermudez A, Honig-Frand A, Krouse R, Arambepola S, Guo E, et al: Human tumor-associated macrophages and neutrophils regulate antitumor antibody efficacy through lethal and sublethal trogocytosis. Cancer Res. 84:1029–1047. 2024. View Article : Google Scholar : PubMed/NCBI

65 

Wu L and Zhang XH: Tumor-associated neutrophils and macrophages-heterogenous but not chaotic. Front Immunol. 11:5539672020. View Article : Google Scholar : PubMed/NCBI

66 

Haider P, Kral-Pointner JB, Mayer J, Richter M, Kaun C, Brostjan C, Eilenberg W, Fischer MB, Speidl WS, Hengstenberg C, et al: Neutrophil extracellular trap degradation by differently polarized macrophage subsets. Arterioscler Thromb Vasc Biol. 40:2265–2278. 2020. View Article : Google Scholar : PubMed/NCBI

67 

Prame Kumar K, Nicholls AJ and Wong CHY: Partners in crime: Neutrophils and monocytes/macrophages in inflammation and disease. Cell Tissue Res. 371:551–565. 2018. View Article : Google Scholar : PubMed/NCBI

68 

Borella R, De Biasi S, Paolini A, Boraldi F, Lo Tartaro D, Mattioli M, Fidanza L, Neroni A, Caro-Maldonado A, Meschiari M, et al: Metabolic reprograming shapes neutrophil functions in severe COVID-19. Eur J Immunol. 52:484–502. 2022. View Article : Google Scholar

69 

Cannarile MA, Weisser M, Jacob W, Jegg AM, Ries CH and Rüttinger D: Colony-stimulating factor 1 receptor (CSF1R) inhibitors in cancer therapy. J Immunother Cancer. 5:532017. View Article : Google Scholar : PubMed/NCBI

70 

Cho H, Seo Y, Loke KM, Kim SW, Oh SM, Kim JH, Soh J, Kim HS, Lee H, Kim J, et al: Cancer-stimulated CAFs enhance monocyte differentiation and protumoral TAM Activation via IL6 and GM-CSF Secretion. Clin Cancer Res. 24:5407–5421. 2018. View Article : Google Scholar : PubMed/NCBI

71 

Schmidt E, Distel L, Erber R, Büttner-Herold M, Rosahl MC, Ott OJ, Strnad V, Hack CC, Hartmann A, Hecht M, et al: Tumor-associated neutrophils are a negative prognostic factor in early luminal breast cancers lacking immunosuppressive macrophage recruitment. Cancers (Basel). 16:31602024. View Article : Google Scholar : PubMed/NCBI

72 

Puerta-Arias JD, Mejía SP and González Á: The role of the interleukin-17 axis and neutrophils in the pathogenesis of endemic and systemic mycoses. Front Cell Infect Microbiol. 10:5953012020. View Article : Google Scholar

73 

Murata K, Murao A, Aziz M and Wang P: Extracellular CIRP induces novel Nectin-2+ (CD112+) neutrophils to promote Th1 differentiation in sepsis. J Immunol. 210:310–321. 2023. View Article : Google Scholar

74 

Parackova Z, Bloomfield M, Klocperk A and Sediva A: Neutrophils mediate Th17 promotion in COVID-19 patients. J Leukoc Biol. 109:73–76. 2021. View Article : Google Scholar

75 

Mishalian I, Bayuh R, Eruslanov E, Michaeli J, Levy L, Zolotarov L, Singhal S, Albelda SM, Granot Z and Fridlender ZG: Neutrophils recruit regulatory T-cells into tumors via secretion of CCL17-a new mechanism of impaired antitumor immunity. Int J Cancer. 135:1178–1186. 2014. View Article : Google Scholar : PubMed/NCBI

76 

Luo H, Ikenaga N, Nakata K, Higashijima N, Zhong P, Kubo A, Wu C, Tsutsumi C, Shimada Y, Hayashi M, et al: Tumor-associated neutrophils upregulate Nectin2 expression, creating the immunosuppressive microenvironment in pancreatic ductal adenocarcinoma. J Exp Clin Cancer Res. 43:2582024. View Article : Google Scholar : PubMed/NCBI

77 

Sun R, Xiong Y, Liu H, Gao C, Su L, Weng J, Yuan X, Zhang D and Feng J: Tumor-associated neutrophils suppress antitumor immunity of NK cells through the PD-L1/PD-1 axis. Transl Oncol. 13:1008252020. View Article : Google Scholar : PubMed/NCBI

78 

Tumino N, Besi F, Di Pace AL, Mariotti FR, Merli P, Li Pira G, Galaverna F, Pitisci A, Ingegnere T, Pelosi A, et al: PMN-MDSC are a new target to rescue graft-versus-leukemia activity of NK cells in haplo-HSC transplantation. Leukemia. 34:932–937. 2020. View Article : Google Scholar :

79 

Pelosi A, Besi F, Tumino N, Merli P, Quatrini L, Li Pira G, Algeri M, Moretta L and Vacca P: NK Cells and PMN-MDSCs in the graft from G-CSF mobilized haploidentical donors display distinct gene expression profiles from those of the non-mobilized counterpart. Front Immunol. 12:6573292021. View Article : Google Scholar : PubMed/NCBI

80 

Mouchemore KA and Anderson RL: Immunomodulatory effects of G-CSF in cancer: Therapeutic implications. Semin Immunol. 54:1015122021. View Article : Google Scholar : PubMed/NCBI

81 

Ogura K, Sato-Matsushita M, Yamamoto S, Hori T, Sasahara M, Iwakura Y, Saiki I, Tahara H and Hayakawa Y: NK cells control tumor-promoting function of neutrophils in mice. Cancer Immunol Res. 6:348–357. 2018. View Article : Google Scholar : PubMed/NCBI

82 

Li X, Xie G, Chen J, Wang Y, Zhai J and Shen L: Tumour cell-derived serglycin promotes IL-8 secretion of CAFs in gastric cancer. Br J Cancer. 131:271–282. 2024. View Article : Google Scholar : PubMed/NCBI

83 

Song M, He J, Pan QZ, Yang J, Zhao J, Zhang YJ, Huang Y, Tang Y, Wang Q, He J, et al: Cancer-associated fibroblast-mediated cellular crosstalk supports hepatocellular carcinoma progression. Hepatology. 73:1717–1735. 2021. View Article : Google Scholar : PubMed/NCBI

84 

Li C, Chen T, Liu J, Wang Y, Zhang C, Guo L, Shi D, Zhang T, Wang X and Li J: FGF19-Induced inflammatory CAF promoted neutrophil extracellular trap formation in the liver metastasis of colorectal cancer. Adv Sci (Weinh). 10:e23026132023. View Article : Google Scholar : PubMed/NCBI

85 

Dudeck J, Kotrba J, Immler R, Hoffmann A, Voss M, Alexaki VI, Morton L, Jahn SR, Katsoulis-Dimitriou K, Winzer S, et al: Directional mast cell degranulation of tumor necrosis factor into blood vessels primes neutrophil extravasation. Immunity. 54:468–483.e5. 2021. View Article : Google Scholar : PubMed/NCBI

86 

Li JY, Chen YP, Li YQ, Liu N and Ma J: Chemotherapeutic and targeted agents can modulate the tumor microenvironment and increase the efficacy of immune checkpoint blockades. Mol Cancer. 20:272021. View Article : Google Scholar : PubMed/NCBI

87 

Oliveira G and Wu CJ: Dynamics and specificities of T cells in cancer immunotherapy. Nat Rev Cancer. 23:295–316. 2023. View Article : Google Scholar : PubMed/NCBI

88 

Yi M, Zheng X, Niu M, Zhu S, Ge H and Wu K: Combination strategies with PD-1/PD-L1 blockade: Current advances and future directions. Mol Cancer. 21:282022. View Article : Google Scholar : PubMed/NCBI

89 

Chu X, Tian W, Wang Z, Zhang J and Zhou R: Co-inhibition of TIGIT and PD-1/PD-L1 in cancer immunotherapy: Mechanisms and clinical trials. Mol Cancer. 22:932023. View Article : Google Scholar : PubMed/NCBI

90 

Wu M, Huang Q, Xie Y, Wu X, Ma H, Zhang Y and Xia Y: Improvement of the anticancer efficacy of PD-1/PD-L1 blockade via combination therapy and PD-L1 regulation. J Hematol Oncol. 15:242022. View Article : Google Scholar : PubMed/NCBI

91 

Gjuka D, Adib E, Garrison K, Chen J, Zhang Y, Li W, Boutz D, Lamb C, Tanno Y, Nassar A, et al: Enzyme-mediated depletion of methylthioadenosine restores T cell function in MTAP-deficient tumors and reverses immunotherapy resistance. Cancer Cell. 41:1774–1787.e9. 2023. View Article : Google Scholar

92 

Niederlova V, Tsyklauri O, Kovar M and Stepanek O: IL-2-driven CD8+ T cell phenotypes: Implications for immunotherapy. Trends Immunol. 44:890–901. 2023. View Article : Google Scholar : PubMed/NCBI

93 

Si J, Shi X, Sun S, Zou B, Li Y, An D, Lin X, Gao Y, Long F, Pang B, et al: Hematopoietic progenitor kinase1 (HPK1) mediates T cell dysfunction and is a druggable target for T cell-based immunotherapies. Cancer Cell. 38:551–566.e11. 2020. View Article : Google Scholar : PubMed/NCBI

94 

Miao S, Rodriguez BL and Gibbons DL: The multifaceted role of neutrophils in NSCLC in the era of immune checkpoint inhibitors. Cancers (Basel). 16:25072024. View Article : Google Scholar : PubMed/NCBI

95 

Xu P, Zhang X, Chen K, Zhu M, Jia R, Zhou Q, Yang J, Dai J, Jin Y and Shi K: Tumor cell-derived microparticles induced by methotrexate augment T-cell antitumor responses by downregulating expression of PD-1 in neutrophils. Cancer Immunol Res. 11:501–514. 2023. View Article : Google Scholar : PubMed/NCBI

96 

Meng Y, Ye F, Nie P, Zhao Q, An L, Wang W, Qu S, Shen Z, Cao Z, Zhang X, et al: Immunosuppressive CD10+ALPL+ neutrophils promote resistance to anti-PD-1 therapy in HCC by mediating irreversible exhaustion of T cells. J Hepatol. 79:1435–1449. 2023. View Article : Google Scholar : PubMed/NCBI

97 

Xie P, Yu M, Zhang B, Yu Q, Zhao Y, Wu M, Jin L, Yan J, Zhou B, Liu S, et al: CRKL dictates anti-PD-1 resistance by mediating tumor-associated neutrophil infiltration in hepatocellular carcinoma. J Hepatol. 81:93–107. 2024. View Article : Google Scholar : PubMed/NCBI

98 

Michaeli J, Shaul ME, Mishalian I, Hovav AH, Levy L, Zolotriov L, Granot Z and Fridlender ZG: Tumor-associated neutrophils induce apoptosis of non-activated CD8 T-cells in a TNFα and NO-dependent mechanism, promoting a tumor-supportive environment. Oncoimmunology. 6:e13569652017. View Article : Google Scholar

99 

Wang TT, Zhao YL, Peng LS, Chen N, Chen W, Lv YP, Mao FY, Zhang JY, Cheng P, Teng YS, et al: Tumour-activated neutrophils in gastric cancer foster immune suppression and disease progression through GM-CSF-PD-L1 pathway. Gut. 66:1900–1911. 2017. View Article : Google Scholar : PubMed/NCBI

100 

Kaltenmeier C, Yazdani HO, Morder K, Geller DA, Simmons RL and Tohme S: Neutrophil extracellular traps promote T cell exhaustion in the tumor microenvironment. Front Immunol. 12:7852222021. View Article : Google Scholar : PubMed/NCBI

101 

Xia Y, He J, Zhang H, Wang H, Tetz G, Maguire CA, Wang Y, Onuma A, Genkin D, Tetz V, et al: AAV-mediated gene transfer of DNase I in the liver of mice with colorectal cancer reduces liver metastasis and restores local innate and adaptive immune response. Mol Oncol. 14:2920–2935. 2020. View Article : Google Scholar : PubMed/NCBI

102 

Zhang H, Wang Y, Onuma A, He J, Wang H, Xia Y, Lal R, Cheng X, Kasumova G, Hu Z, et al: Neutrophils extracellular traps inhibition improves PD-1 blockade immunotherapy in colorectal cancer. Cancers (Basel). 13:53332021. View Article : Google Scholar : PubMed/NCBI

103 

Peng JJ, Wang L, Li Z, Ku CL and Ho PC: Metabolic challenges and interventions in CAR T cell therapy. Sci Immunol. 8:eabq30162023. View Article : Google Scholar : PubMed/NCBI

104 

Albelda SM: CAR T cell therapy for patients with solid tumours: Key lessons to learn and unlearn. Nat Rev Clin Oncol. 21:47–66. 2024. View Article : Google Scholar

105 

Bulliard Y, Andersson BS, Baysal MA, Damiano J and Tsimberidou AM: Reprogramming T cell differentiation and exhaustion in CAR-T cell therapy. J Hematol Oncol. 16:1082023. View Article : Google Scholar : PubMed/NCBI

106 

Pan K, Farrukh H, Chittepu VCSR, Xu H, Pan CX and Zhu Z: CAR race to cancer immunotherapy: from CAR T, CAR NK to CAR macrophage therapy. J Exp Clin Cancer Res. 41:1192022. View Article : Google Scholar : PubMed/NCBI

107 

Hong M, Clubb JD and Chen YY: Engineering CAR-T cells for next-generation cancer therapy. Cancer Cell. 38:473–488. 2020. View Article : Google Scholar : PubMed/NCBI

108 

Zhang H, Yu P, Tomar VS, Chen X, Atherton MJ, Lu Z, Zhang HG, Li S, Ortiz A, Gui J, et al: Targeting PARP11 to avert immunosuppression and improve CAR T therapy in solid tumors. Nat Cancer. 3:808–820. 2022. View Article : Google Scholar : PubMed/NCBI

109 

The Lancet Oncology: CAR T-cell therapy for solid tumours. Lancet Oncol. 22:8932021. View Article : Google Scholar : PubMed/NCBI

110 

Li X, Zhu T, Wang R, Chen J, Tang L, Huo W, Huang X and Cao Q: Genetically programmable vesicles for enhancing CAR-T therapy against solid tumors. Adv Mater. 35:e22111382023. View Article : Google Scholar : PubMed/NCBI

111 

Krishnan SR and Bebawy M: Circulating biosignatures in multiple myeloma and their role in multidrug resistance. Mol Cancer. 22:792023. View Article : Google Scholar : PubMed/NCBI

112 

Wen X, Huang Z, Yang X, He X, Li L, Chen H, Wang K, Guo Q and Liu J: Development of an aptamer capable of multidrug resistance reversal for tumor combination chemotherapy. Proc Natl Acad Sci USA. 121:e23211161212024. View Article : Google Scholar : PubMed/NCBI

113 

Mousset A, Lecorgne E, Bourget I, Lopez P, Jenovai K, Cherfils-Vicini J, Dominici C, Rios G, Girard-Riboulleau C, Liu B, et al: Neutrophil extracellular traps formed during chemotherapy confer treatment resistance via TGF-β activation. Cancer Cell. 41:757–775.e10. 2023. View Article : Google Scholar

114 

Saw PE, Chen J and Song E: ChemoNETosis: A road to tumor therapeutic resistance. Cancer Cell. 41:655–657. 2023. View Article : Google Scholar : PubMed/NCBI

115 

Yang Y, Yu S, Lv C and Tian Y: NETosis in tumour microenvironment of liver: From primary to metastatic hepatic carcinoma. Ageing Res Rev. 97:1022972024. View Article : Google Scholar : PubMed/NCBI

116 

Kong X, Zhang Y, Xiang L, You Y, Duan Y, Zhao Y, Li S, Wu R, Zhang J, Zhou L and Duan L: Fusobacterium nucleatum-triggered neutrophil extracellular traps facilitate colorectal carcinoma progression. J Exp Clin Cancer Res. 42:2362023. View Article : Google Scholar : PubMed/NCBI

117 

Zhang Y, Yang Y, Hu X, Wang Z, Li L and Chen P: PADs in cancer: Current and future. Biochim Biophys Acta Rev Cancer. 1875:1884922021. View Article : Google Scholar

118 

Zhan X, Wu R, Kong XH, You Y, He K, Sun XY, Huang Y, Chen WX and Duan L: Elevated neutrophil extracellular traps by HBV-mediated S100A9-TLR4/RAGE-ROS cascade facilitate the growth and metastasis of hepatocellular carcinoma. Cancer Commun (Lond). 43:225–245. 2023. View Article : Google Scholar

119 

Mousset A, Bellone L, Gaggioli C and Albrengues J: NETscape or NEThance: Tailoring anti-cancer therapy. Trends Cancer. 10:655–667. 2024. View Article : Google Scholar : PubMed/NCBI

120 

Ramachandran IR, Condamine T, Lin C, Herlihy SE, Garfall A, Vogl DT, Gabrilovich DI and Nefedova Y: Bone marrow PMN-MDSCs and neutrophils are functionally similar in protection of multiple myeloma from chemotherapy. Cancer Lett. 371:117–124. 2016. View Article : Google Scholar :

121 

Tamura K, Miyato H, Kanamaru R, Sadatomo A, Takahashi K, Ohzawa H, Koyanagi T, Saga Y, Takei Y, Fujiwara H, et al: Neutrophil extracellular traps (NETs) reduce the diffusion of doxorubicin which may attenuate its ability to induce apoptosis of ovarian cancer cells. Heliyon. 8:e097302022. View Article : Google Scholar : PubMed/NCBI

122 

Goenka A, Khan F, Verma B, Sinha P, Dmello CC, Jogalekar MP, Gangadaran P and Ahn BC: Tumor microenvironment signaling and therapeutics in cancer progression. Cancer Commun (Lond). 43:525–561. 2023. View Article : Google Scholar : PubMed/NCBI

123 

Zhang R, Dong M, Tu J, Li F, Deng Q, Xu J, He X, Ding J, Xia J, Sheng D, et al: PMN-MDSCs modulated by CCL20 from cancer cells promoted breast cancer cell stemness through CXCL2-CXCR2 pathway. Signal Transduct Target Ther. 8:972023. View Article : Google Scholar : PubMed/NCBI

124 

Kang J, La Manna F, Bonollo F, Sampson N, Alberts IL, Mingels C, Afshar-Oromieh A, Thalmann GN and Karkampouna S: Tumor microenvironment mechanisms and bone metastatic disease progression of prostate cancer. Cancer Lett. 530:156–169. 2022. View Article : Google Scholar : PubMed/NCBI

125 

Capucetti A, Albano F and Bonecchi R: Multiple roles for chemokines in neutrophil biology. Front Immunol. 11:12592020. View Article : Google Scholar : PubMed/NCBI

126 

Rajarathnam K, Schnoor M, Richardson RM and Rajagopal S: How do chemokines navigate neutrophils to the target site: Dissecting the structural mechanisms and signaling pathways. Cell Signal. 54:69–80. 2019. View Article : Google Scholar :

127 

Bianchi A, De Castro Silva I, Deshpande NU, Singh S, Mehra S, Garrido VT, Guo X, Nivelo LA, Kolonias DS, Saigh SJ, et al: Cell-Autonomous Cxcl1 Sustains Tolerogenic Circuitries and Stromal Inflammation via Neutrophil-Derived TNF in Pancreatic Cancer. Cancer Discov. 13:1428–1453. 2023. View Article : Google Scholar : PubMed/NCBI

128 

Chao T, Furth EE and Vonderheide RH: CXCR2-Dependent accumulation of tumor-associated neutrophils regulates T-cell immunity in pancreatic ductal adenocarcinoma. Cancer Immunol Res. 4:968–982. 2016. View Article : Google Scholar : PubMed/NCBI

129 

Corsaro A, Tremonti B, Bajetto A, Barbieri F, Thellung S and Florio T: Chemokine signaling in tumors: potential role of CXC chemokines and their receptors as glioblastoma therapeutic targets. Expert Opin Ther Targets. 28:937–952. 2024. View Article : Google Scholar : PubMed/NCBI

130 

Powell D, Lou M, Barros Becker F and Huttenlocher A: Cxcr1 mediates recruitment of neutrophils and supports proliferation of tumor-initiating astrocytes in vivo. Sci Rep. 8:132852018. View Article : Google Scholar : PubMed/NCBI

131 

Jablonska J, Wu CF, Andzinski L, Leschner S and Weiss S: CXCR2-mediated tumor-associated neutrophil recruitment is regulated by IFN-β. Int J Cancer. 134:1346–1358. 2014. View Article : Google Scholar

132 

Haider C, Hnat J, Wagner R, Huber H, Timelthaler G, Grubinger M, Coulouarn C, Schreiner W, Schlangen K, Sieghart W, et al: Transforming growth factor-β and Axl induce CXCL5 and neutrophil recruitment in hepatocellular carcinoma. Hepatology. 69:222–236. 2019. View Article : Google Scholar

133 

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. 70:1214–1230. 2019. View Article : Google Scholar : PubMed/NCBI

134 

Zhou SL, Zhou ZJ, Hu ZQ, Huang XW, Wang Z, Chen EB, Fan J, Cao Y, Dai Z and Zhou J: Tumor-associated neutrophils recruit macrophages and T-regulatory cells to promote progression of hepatocellular carcinoma and resistance to sorafenib. Gastroenterology. 150:1646–1658.e17. 2016. View Article : Google Scholar : PubMed/NCBI

135 

He J, Zhou M, Yin J, Wan J, Chu J, Jia J, Sheng J, Wang C, Yin H and He F: METTL3 restrains papillary thyroid cancer progression via m6A/c-Rel/IL-8-mediated neutrophil infiltration. Mol Ther. 29:1821–1837. 2021. View Article : Google Scholar : PubMed/NCBI

136 

Schimek V, Strasser K, Beer A, Göber S, Walterskirchen N, Brostjan C, Müller C, Bachleitner-Hofmann T, Bergmann M, Dolznig H and Oehler R: Tumour cell apoptosis modulates the colorectal cancer immune microenvironment via interleukin-8-dependent neutrophil recruitment. Cell Death Dis. 13:1132022. View Article : Google Scholar : PubMed/NCBI

137 

Bellomo G, Rainer C, Quaranta V, Astuti Y, Raymant M, Boyd E, Stafferton R, Campbell F, Ghaneh P, Halloran CM, et al: Chemotherapy-induced infiltration of neutrophils promotes pancreatic cancer metastasis via Gas6/AXL signalling axis. Gut. 71:2284–2299. 2022. View Article : Google Scholar : PubMed/NCBI

138 

Nywening TM, Belt BA, Cullinan DR, Panni RZ, Han BJ, Sanford DE, Jacobs RC, Ye J, Patel AA, Gillanders WE, et al: Targeting both tumour-associated CXCR2+ neutrophils and CCR2+ macrophages disrupts myeloid recruitment and improves chemotherapeutic responses in pancreatic ductal adenocarcinoma. Gut. 67:1112–1123. 2018. View Article : Google Scholar

139 

Cheng Y, Ma XL, Wei YQ and Wei XW: Potential roles and targeted therapy of the CXCLs/CXCR2 axis in cancer and inflammatory diseases. Biochim Biophys Acta Rev Cancer. 1871:289–312. 2019. View Article : Google Scholar : PubMed/NCBI

140 

Schott AF, Goldstein LJ, Cristofanilli M, Ruffini PA, McCanna S, Reuben JM, Perez RP, Kato G and 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. 23:5358–5365. 2017. View Article : Google Scholar : PubMed/NCBI

141 

Jiang H, Cui J, Chu H, Xu T, Xie M, Jing X, Xu J, Zhou J and Shu Y: Targeting IL8 as a sequential therapy strategy to overcome chemotherapy resistance in advanced gastric cancer. Cell Death Discov. 8:2352022. View Article : Google Scholar : PubMed/NCBI

142 

Cheng Y, Mo F, Li Q, Han X, Shi H, Chen S, Wei Y and Wei X: Targeting CXCR2 inhibits the progression of lung cancer and promotes therapeutic effect of cisplatin. Mol Cancer. 20:622021. View Article : Google Scholar : PubMed/NCBI

143 

Kiri S and Ryba T: Cancer, metastasis, and the epigenome. Mol Cancer. 23:1542024. View Article : Google Scholar : PubMed/NCBI

144 

Fang Y, Wang S, Han S, Zhao Y, Yu C, Liu H and Li N: Targeted protein degrader development for cancer: Advances, challenges, and opportunities. Trends Pharmacol Sci. 44:303–317. 2023. View Article : Google Scholar : PubMed/NCBI

145 

Liu ZL, Chen HH, Zheng LL, Sun LP and Shi L: Angiogenic signaling pathways and anti-angiogenic therapy for cancer. Signal Transduct Target Ther. 8:1982023. View Article : Google Scholar : PubMed/NCBI

146 

Zhang T, Jia Y, Yu Y, Zhang B, Xu F and Guo H: Targeting the tumor biophysical microenvironment to reduce resistance to immunotherapy. Adv Drug Deliv Rev. 186:1143192022. View Article : Google Scholar : PubMed/NCBI

147 

Yang M, Mu Y, Yu X, Gao D, Zhang W, Li Y, Liu J, Sun C and Zhuang J: Survival strategies: How tumor hypoxia microenvironment orchestrates angiogenesis. Biomed Pharmacother. 176:1167832024. View Article : Google Scholar : PubMed/NCBI

148 

Li H, Qiu Z, Li F and Wang C: The relationship between MMP-2 and MMP-9 expression levels with breast cancer incidence and prognosis. Oncol Lett. 14:5865–5870. 2017.PubMed/NCBI

149 

Negri L and Ferrara N: The prokineticins: Neuromodulators and mediators of inflammation and myeloid cell-dependent angiogenesis. Physiol Rev. 98:1055–1082. 2018. View Article : Google Scholar : PubMed/NCBI

150 

Shojaei F and Ferrara N: Refractoriness to antivascular endothelial growth factor treatment: Role of myeloid cells. Cancer Res. 68:5501–5504. 2008. View Article : Google Scholar : PubMed/NCBI

151 

Shojaei F, Wu X, Zhong C, Yu L, Liang XH, Yao J, Blanchard D, Bais C, Peale FV, van Bruggen N, et al: Bv8 regulates myeloid-cell-dependent tumour angiogenesis. Nature. 450:825–831. 2007. View Article : Google Scholar : PubMed/NCBI

152 

Majidpoor J and Mortezaee K: Angiogenesis as a hallmark of solid tumors-clinical perspectives. Cell Oncol (Dordr). 44:715–737. 2021. View Article : Google Scholar : PubMed/NCBI

153 

Chung AS, Wu X, Zhuang G, Ngu H, Kasman I, Zhang J, Vernes JM, Jiang Z, Meng YG, Peale FV, et al: An interleukin-17-mediated paracrine network promotes tumor resistance to anti-angiogenic therapy. Nat Med. 19:1114–1123. 2013. View Article : Google Scholar : PubMed/NCBI

154 

Li TJ, Jiang YM, Hu YF, Huang L, Yu J, Zhao LY, Deng HJ, Mou TY, Liu H, Yang Y, et al: Interleukin-17-producing neutrophils link inflammatory stimuli to disease progression by promoting angiogenesis in gastric cancer. Clin Cancer Res. 23:1575–1585. 2017. View Article : Google Scholar

155 

Lee JM, McNamee CJ, Toloza E, Negrao MV, Lin J, Shum E, Cummings AL, Kris MG, Sepesi B, Bara I, et al: Neoadjuvant targeted therapy in resectable NSCLC: Current and future perspectives. J Thorac Oncol. 18:1458–1477. 2023. View Article : Google Scholar : PubMed/NCBI

156 

Napolitano S, Martini G, Ciardiello D, Del Tufo S, Martinelli E, Troiani T and Ciardiello F: Targeting the EGFR signalling pathway in metastatic colorectal cancer. Lancet Gastroenterol Hepatol. 9:664–676. 2024. View Article : Google Scholar : PubMed/NCBI

157 

Damare R, Engle K and Kumar G: Targeting epidermal growth factor receptor and its downstream signaling pathways by natural products: A mechanistic insight. Phytother Res. 38:2406–2447. 2024. View Article : Google Scholar : PubMed/NCBI

158 

Wang X, Jiang W, Du Y, Zhu D, Zhang J, Fang C, Yan F and Chen ZS: Targeting feedback activation of signaling transduction pathways to overcome drug resistance in cancer. Drug Resist Updat. 65:1008842022. View Article : Google Scholar : PubMed/NCBI

159 

Kim GT, Hahn KW, Yoon SY, Sohn KY and Kim JW: PLAG exerts anti-metastatic effects by interfering with neutrophil elastase/PAR2/EGFR signaling in A549 lung cancer orthotopic model. Cancers (Basel). 12:5602020. View Article : Google Scholar : PubMed/NCBI

160 

Swain SM, Shastry M and Hamilton E: Targeting HER2-positive breast cancer: Advances and future directions. Nat Rev Drug Discov. 22:101–126. 2023. View Article : Google Scholar

161 

Sato T, Takahashi S, Mizumoto T, Harao M, Akizuki M, Takasugi M, Fukutomi T and Yamashita J: Neutrophil elastase and cancer. Surg Oncol. 15:217–222. 2006. View Article : Google Scholar

162 

Schlessinger J: Common and distinct elements in cellular signaling via EGF and FGF receptors. Science. 306:1506–1507. 2004. View Article : Google Scholar : PubMed/NCBI

163 

Singh JK, Farnie G, Bundred NJ, Simões BM, Shergill A, Landberg G, Howell SJ and Clarke RB: Targeting CXCR1/2 significantly reduces breast cancer stem cell activity and increases the efficacy of inhibiting HER2 via HER2-dependent and -independent mechanisms. Clin Cancer Res. 19:643–656. 2013. View Article : Google Scholar

164 

Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A, Fleming T, Eiermann W, Wolter J, Pegram M, et al: Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med. 344:783–792. 2001. View Article : Google Scholar : PubMed/NCBI

165 

Singhal A, Li BT and O'Reilly EM: Targeting KRAS in cancer. Nat Med. 30:969–983. 2024. View Article : Google Scholar : PubMed/NCBI

166 

Biller LH and Schrag D: Diagnosis and treatment of metastatic colorectal cancer: A review. JAMA. 325:669–685. 2021. View Article : Google Scholar : PubMed/NCBI

167 

Zhu G, Pei L, Xia H, Tang Q and Bi F: Role of oncogenic KRAS in the prognosis, diagnosis and treatment of colorectal cancer. Mol Cancer. 20:1432021. View Article : Google Scholar : PubMed/NCBI

168 

Shang A, Gu C, Zhou C, Yang Y, Chen C, Zeng B, Wu J, Lu W, Wang W, Sun Z and Li D: Exosomal KRAS mutation promotes the formation of tumor-associated neutrophil extracellular traps and causes deterioration of colorectal cancer by inducing IL-8 expression. Cell Commun Signal. 18:522020. View Article : Google Scholar : PubMed/NCBI

169 

Pickup MW, Owens P, Gorska AE, Chytil A, Ye F, Shi C, Weaver VM, Kalluri R, Moses HL and Novitskiy SV: Development of aggressive pancreatic ductal adenocarcinomas depends on granulocyte colony stimulating factor secretion in carcinoma cells. Cancer Immunol Res. 5:718–729. 2017. View Article : Google Scholar : PubMed/NCBI

170 

Nolan E, Bridgeman VL, Ombrato L, Karoutas A, Rabas N, Sewnath CAN, Vasquez M, Rodrigues FS, Horswell S, Faull P, et al: Radiation exposure elicits a neutrophil-driven response in healthy lung tissue that enhances metastatic colonization. Nat Cancer. 3:173–187. 2022. View Article : Google Scholar : PubMed/NCBI

171 

Wisdom AJ, Hong CS, Lin AJ, Xiang Y, Cooper DE, Zhang J, Xu ES, Kuo HC, Mowery YM, Carpenter DJ, et al: Neutrophils promote tumor resistance to radiation therapy. Proc Natl Acad Sci USA. 116:18584–18589. 2019. View Article : Google Scholar : PubMed/NCBI

172 

Zheng X, Song X, Zhu G, Pan D, Li H, Hu J, Xiao K, Gong Q, Gu Z, Luo K and Li W: Nanomedicine combats drug resistance in lung cancer. Adv Mater. 36:e23089772024. View Article : Google Scholar

173 

Xu K, Guo H, Xia A, Wang Z, Wang S and Wang Q: Non-coding RNAs in radiotherapy resistance: Roles and therapeutic implications in gastrointestinal cancer. Biomed Pharmacother. 161:1144852023. View Article : Google Scholar : PubMed/NCBI

174 

Wu Y, Song Y, Wang R and Wang T: Molecular mechanisms of tumor resistance to radiotherapy. Mol Cancer. 22:962023. View Article : Google Scholar : PubMed/NCBI

175 

An L, Li M and Jia Q: Mechanisms of radiotherapy resistance and radiosensitization strategies for esophageal squamous cell carcinoma. Mol Cancer. 22:1402023. View Article : Google Scholar : PubMed/NCBI

176 

Peng J, Yin X, Yun W, Meng X and Huang Z: Radiotherapyinduced tumor physical microenvironment remodeling to overcome immunotherapy resistance. Cancer Lett. 559:2161082023. View Article : Google Scholar

177 

Beckers C, Pruschy M and Vetrugno I: Tumor hypoxia and radiotherapy: A major driver of resistance even for novel radiotherapy modalities. Semin Cancer Biol. 98:19–30. 2024. View Article : Google Scholar

178 

Wang X, Li X, Wu Y, Hong J and Zhang M: The prognostic significance of tumor-associated neutrophils and circulating neutrophils in glioblastoma (WHO CNS5 classification). BMC Cancer. 23:202023. View Article : Google Scholar : PubMed/NCBI

179 

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. 26:2139–2156. 2019. View Article : Google Scholar : PubMed/NCBI

180 

Ruiz-Fernández de Córdoba B, Moreno H, Valencia K, Perurena N, Ruedas P, Walle T, Pezonaga-Torres A, Hinojosa J, Guruceaga E, Pineda-Lucena A, et al: Tumor ENPP1 (CD203a)/ haptoglobin axis exploits myeloid-derived suppressor cells to promote post-radiotherapy local recurrence in breast cancer. Cancer Discov. 12:1356–1377. 2022. View Article : Google Scholar

181 

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. 81:2345–2357. 2021. View Article : Google Scholar : PubMed/NCBI

182 

Shinde-Jadhav S, Mansure JJ, Rayes RF, Marcq G, Ayoub M, Skowronski R, Kool R, Bourdeau F, Brimo F, Spicer J and Kassouf W: Role of neutrophil extracellular traps in radiation resistance of invasive bladder cancer. Nat Commun. 12:27762021. View Article : Google Scholar : PubMed/NCBI

183 

Li H, Zeng J, You Q, Zhang M, Shi Y, Yang X, Gu W, Liu Y, Hu N, Wang Y, et al: X-ray-activated nanoscintillators integrated with tumor-associated neutrophils polarization for improved radiotherapy in metastatic colorectal cancer. Biomaterials. 316:1230312025. View Article : Google Scholar

184 

Rys RN and Calcinotto A: Senescent neutrophils: A hidden role in cancer progression. Trends Cell Biol. S0962-8924(24)00187-9. 2024.Epub ahead of print. View Article : Google Scholar : PubMed/NCBI

185 

Treffers LW, Ten Broeke T, Rösner T, Jansen JHM, van Houdt M, Kahle S, Schornagel K, Verkuijlen PJJH, Prins JM, Franke K, et al: IgA-mediated killing of tumor cells by neutrophils is enhanced by CD47-SIRPα checkpoint inhibition. Cancer Immunol Res. 8:120–130. 2020. View Article : Google Scholar

186 

Brandsma AM, Ten Broeke T, Nederend M, Meulenbroek LA, van Tetering G, Meyer S, Jansen JH, Beltrán Buitrago MA, Nagelkerke SQ, Németh I, et al: Simultaneous targeting of FcγRs and FcαRI enhances tumor cell killing. Cancer Immunol Res. 3:1316–1324. 2015. View Article : Google Scholar : PubMed/NCBI

187 

Borrok MJ, Luheshi NM, Beyaz N, Davies GC, Legg JW, Wu H, Dall'Acqua WF and Tsui P: Enhancement of antibody-dependent cell-mediated cytotoxicity by endowing IgG with FcαRI (CD89) binding. MAbs. 7:743–751. 2015. View Article : Google Scholar :

188 

Kumbhojkar N, Prakash S, Fukuta T, Adu-Berchie K, Kapate N, An R, Darko S, Chandran Suja V, Park KS, Gottlieb AP, et al: Neutrophils bearing adhesive polymer micropatches as a drug-free cancer immunotherapy. Nat Biomed Eng. 8:579–592. 2024. View Article : Google Scholar : PubMed/NCBI

189 

Quaas A, Pamuk A, Klein S, Quantius J, Rehkaemper J, Bar utcu AG, Rueschoff J, Zander T, Gebauer F, Hillmer A, et al: Sex-specific prognostic effect of CD66b-positive tumor-infiltrating neutrophils (TANs) in gastric and esophageal adenocarcinoma. Gastric Cancer. 24:1213–1226. 2021. View Article : Google Scholar : PubMed/NCBI

190 

Peng H, Wu X, Liu S, He M, Tang C, Wen Y, Xie C, Zhong R, Li C, Xiong S, et al: Cellular dynamics in tumour microenvironment along with lung cancer progression underscore spatial and evolutionary heterogeneity of neutrophil. Clin Transl Med. 13:e13402023. View Article : Google Scholar : PubMed/NCBI

191 

Zhang J, Zhang M, Lou J, Wu L, Zhang S, Liu X, Ke Y, Zhao S, Song Z, Bai X, et al: Machine learning integration with single-cell transcriptome sequencing datasets reveals the impact of tumor-associated neutrophils on the immune microenvironment and immunotherapy outcomes in gastric cancer. Int J Mol Sci. 25:127152024. View Article : Google Scholar : PubMed/NCBI

192 

Ye L, Zhang T, Kang Z, Guo G, Sun Y, Lin K, Huang Q, Shi X, Ni Z, Ding N, et al: Tumor-infiltrating immune cells act as a marker for prognosis in colorectal cancer. Front Immunol. 10:23682019. View Article : Google Scholar : PubMed/NCBI

193 

Nøst TH, Alcala K, Urbarova I, Byrne KS, Guida F, Sandanger TM and Johansson M: Systemic inflammation markers and cancer incidence in the UK Biobank. Eur J Epidemiol. 36:841–848. 2021. View Article : Google Scholar : PubMed/NCBI

194 

Mosca M, Nigro MC, Pagani R, De Giglio A and Di Federico A: Neutrophil-to-lymphocyte ratio (NLR) in NSCLC, gastrointestinal, and other solid tumors: Immunotherapy and beyond. Biomolecules. 13:18032023. View Article : Google Scholar : PubMed/NCBI

195 

Cupp MA, Cariolou M, Tzoulaki I, Aune D, Evangelou E and Berlanga-Taylor AJ: Neutrophil to lymphocyte ratio and cancer prognosis: An umbrella review of systematic reviews and meta-analyses of observational studies. BMC Med. 18:3602020. View Article : Google Scholar : PubMed/NCBI

196 

Ethier JL, Desautels D, Templeton A, Shah PS and Amir E: Prognostic role of neutrophil-to-lymphocyte ratio in breast cancer: A systematic review and meta-analysis. Breast Cancer Res. 19:22017. View Article : Google Scholar : PubMed/NCBI

197 

Pecqueux M, Brückner F, Oehme F, Hempel S, Baenke F, Riediger C, Distler M, Weitz J and Kahlert C: Preoperative IL-8 levels as prognostic indicators of overall survival: An extended follow-up in a prospective cohort with colorectal liver metastases. BMC Cancer. 24:902024. View Article : Google Scholar : PubMed/NCBI

198 

Hsu YJ, Chern YJ, Wu ZE, Yu YL, Liao CK, Tsai WS, You JF and Lee CW: The oncologic outcome and prognostic factors for solitary colorectal liver metastasis after liver resection. J Gastrointest Surg. 28:267–275. 2024. View Article : Google Scholar : PubMed/NCBI

199 

Huang W, Jiang Y, Xiong W, Sun Z, Chen C, Yuan Q, Zhou K, Han Z, Feng H, Chen H, et al: Noninvasive imaging of the tumor immune microenvironment correlates with response to immunotherapy in gastric cancer. Nat Commun. 13:50952022. View Article : Google Scholar : PubMed/NCBI

200 

Tan S, Zheng Q, Zhang W, Zhou M, Xia C and Feng W: Prognostic value of inflammatory markers NLR, PLR, and LMR in gastric cancer patients treated with immune checkpoint inhibitors: A meta-analysis and systematic review. Front Immunol. 15:14087002024. View Article : Google Scholar : PubMed/NCBI

201 

He G, Zhang H, Zhou J, Wang B, Chen Y, Kong Y, Xie X, Wang X, Fei R, Wei L, et al: Peritumoural neutrophils negatively regulate adaptive immunity via the PD-L1/PD-1 signalling pathway in hepatocellular carcinoma. J Exp Clin Cancer Res. 34:1412015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2025
Volume 66 Issue 4

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Hou R, Wu X, Wang C, Fan H, Zhang Y, Wu H, Wang H, Ding J, Jiang H, Xu J, Xu J, et al: Tumor‑associated neutrophils: Critical regulators in cancer progression and therapeutic resistance (Review). Int J Oncol 66: 28, 2025.
APA
Hou, R., Wu, X., Wang, C., Fan, H., Zhang, Y., Wu, H. ... Xu, J. (2025). Tumor‑associated neutrophils: Critical regulators in cancer progression and therapeutic resistance (Review). International Journal of Oncology, 66, 28. https://doi.org/10.3892/ijo.2025.5734
MLA
Hou, R., Wu, X., Wang, C., Fan, H., Zhang, Y., Wu, H., Wang, H., Ding, J., Jiang, H., Xu, J."Tumor‑associated neutrophils: Critical regulators in cancer progression and therapeutic resistance (Review)". International Journal of Oncology 66.4 (2025): 28.
Chicago
Hou, R., Wu, X., Wang, C., Fan, H., Zhang, Y., Wu, H., Wang, H., Ding, J., Jiang, H., Xu, J."Tumor‑associated neutrophils: Critical regulators in cancer progression and therapeutic resistance (Review)". International Journal of Oncology 66, no. 4 (2025): 28. https://doi.org/10.3892/ijo.2025.5734