Next Article in Journal
Effects of SGLT2 Inhibitors on Sleep Apnea Parameters and Cheyne–Stokes Respiration in Patients with Acute Decompensated Heart Failure: A Prospective Cohort Study
Previous Article in Journal
Applications of Surface Plasmon Resonance in Heparan Sulfate Interactome Research
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Neutrophil Spatiotemporal Regulatory Networks: Dual Roles in Tumor Growth Regulation and Metastasis

1
Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
2
Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan 430030, China
3
Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 430030, China
*
Authors to whom correspondence should be addressed.
Biomedicines 2025, 13(6), 1473; https://doi.org/10.3390/biomedicines13061473 (registering DOI)
Submission received: 30 April 2025 / Revised: 11 June 2025 / Accepted: 12 June 2025 / Published: 14 June 2025
(This article belongs to the Section Cancer Biology and Oncology)

Abstract

:
Neutrophils, accounting for 50–70% of circulating leukocytes, exhibit remarkable plasticity in tumor biology. Depending on tumor type and microenvironmental cues, they can exert either anti-tumor or pro-tumor effects. During tumor initiation, neutrophils exposed to chronic inflammation secrete cytokines and oncogenic microRNAs that promote genomic instability and malignant transformation. In tumor progression, neutrophils adopt context-dependent phenotypes and execute diverse functions, including polarization into anti-tumor (N1) or pro-tumor (N2) subsets; secretion of inflammatory and angiogenic mediators; formation of neutrophil extracellular traps (NETs); production of reactive oxygen and nitrogen species (e.g., H2O2 and nitric oxide); and modulation of immune cell infiltration and function within the tumor microenvironment. During metastasis, neutrophils facilitate cancer dissemination through three principal mechanisms: (1) promoting epithelial–mesenchymal transition (EMT) via inflammatory signaling, adhesion molecule interactions, and lipid metabolic support; (2) establishing pre-metastatic niches by remodeling distant organ stroma through NETs and matrix metalloproteinases; and (3) reactivating dormant tumor cells in response to chronic inflammation, viral infection, or stress hormones. Collectively, neutrophils function as central regulators across all stages of tumor evolution, influencing cancer growth, immune evasion, and metastatic progression. This review aims to provide a comprehensive synthesis of neutrophil-mediated mechanisms in the tumor microenvironment and highlight emerging strategies for neutrophil-targeted cancer therapy.

1. Introduction

Neutrophils, the most prevalent innate immune cells in bone marrow and peripheral blood, are widely acknowledged as a vital element of the immune system [1]. They utilize a range of strategies, such as phagocytosis, NETs formation and degranulation to efficiently eradicate pathogens [2]. Under physiological environments, neutrophils exhibit limited longevity, with their stimulation and trafficking tightly regulated to prevent potential damage to healthy tissues due to their highly cytotoxic responses [3]. Neutrophils manifest a dual nature in disease contexts, serving as both essential defenders and potential contributors to pathological processes. On one hand, they are essential in host defense by combating various pathogens [4,5,6] and facilitate tissue regeneration and angiogenesis post-injury by clearing apoptotic cell debris [7]. On the other hand, in diverse pathological conditions, such as cardiovascular disorders, autoimmune diseases, infectious diseases, allergic conditions, and vasculitides, the presence of both pro-inflammatory and anti-inflammatory neutrophil subpopulations has been observed, underscoring their functional complexity and heterogeneity [8,9,10,11,12,13,14,15,16].
The tumor microenvironment (TME) is populated by heterogeneous immune cell subsets, notably neutrophils, macrophages, and T cells, each contributing fundamentally to tumor immunobiology [17,18,19,20]. Neutrophils represent a quantitatively and functionally significant population within the immune landscape of most solid tumors [21,22,23,24]. As the predominant immune cell population, neutrophils exhibit complex and significant functions in the context of cancer. The evolving understanding of neutrophil heterogeneity and plasticity has revitalized their study in cancer pathogenesis. Emerging evidence indicates that neutrophils are far from the homogeneous population once presumed; rather, their functionality is profoundly influenced by the surrounding microenvironment, resulting in varying degrees of pro-tumorigenic or anti-tumorigenic properties [25,26]. Extensive research has revealed the pleiotropic anti-tumor capacities of neutrophils, including direct tumoricidal activity and the capacity to suppress metastasis [27,28,29,30]. Conversely, other investigations have revealed that tumor-associated neutrophils (TANs) can promote tumor progression by facilitating angiogenic switching and enhancing tumor cell motility, migration, and invasion [31,32] (Figure 1). Epidemiological studies further underscore a significant correlation between neutrophils and clinical outcomes across various tumor types [33,34,35,36,37,38].
This review synthesizes current knowledge on neutrophil ontogeny, functional plasticity, and their dichotomous roles in tumor progression. By delineating neutrophil-microenvironment crosstalk, we aim to identify actionable targets for intercepting cancer progression and metastasis.

2. Neutrophil Biology: Development, Homeostasis, and Circulation

2.1. Neutrophil Development and Functional Maturation

Under homeostatic conditions, neutrophils, as the most predominant immune effector cells in humans, constitute approximately 50–70% of the total peripheral white blood cell count. Their daily production exceeds 1011 cells [39], yet mature neutrophils lose their proliferative capacity. To sustain this high output and rapid turnover, the bone marrow dedicates about 65% of its hematopoietic space to granulocyte-monocyte lineage development under physiological conditions [40]. Notably, the conventional identification of granulocyte precursors primarily relies on histological observation via Giemsa staining after density gradient centrifugation [41]. This staging system, based on morphological features (including cell volume, nuclear-to-cytoplasmic ratio, chromatin condensation, and specific granule content), still suffers from limitations due to subjective interpretation.
Developmental studies indicate neutrophils arise from lymphoid-primed multipotent progenitors (LMPPs) during hematopoietic differentiation, with LMPPs themselves being derived from multipotent progenitors (MPPs) [42]. These progenitors undergo stepwise differentiation through the granulocyte–monocyte progenitor (GMP) stage, a process regulated by a cascade of key transcription factors [40,43,44]. Terminal differentiation follows a strict morphological progression: myeloblast → promyelocyte → myelocyte → metamyelocyte → band cell → mature neutrophil [40,45,46,47]. Importantly, the granule assembly cascade initiates with primary (azurophilic) granule formation at the promyelocyte stage, followed by robust production of secondary (specific) granules in myelocytes, culminating in tertiary (gelatinase) granule generation during terminal neutrophil maturation [40,48]. These subcellular structures compartmentalize effector molecules such as myeloperoxidase, defensins, antimicrobial peptides, as well as proteolytic enzymes like elastase and matrix metalloproteinases (MMPs), playing dual roles in antimicrobial defense and tissue homeostasis regulation [48,49] (Figure 2).
Following egress from the bone marrow, terminally differentiated neutrophils enter the circulatory compartment as mature circulating neutrophils, with species-dependent half-lives: ~7 h in humans [50,51] and 8–10 h in mice [52]. After migrating to inflammatory sites, their lifespan can extend to 24–48 h before spontaneous apoptosis [53], with certain microenvironments further delaying apoptosis [48,52,54].

2.2. Bone Marrow Reservoir and Regulatory Functions

As the most numerically dominant cell type in the bone marrow, neutrophils constitute the largest granulocyte reservoir in the body. Cross-species classical studies tracking their migration and distribution dynamics have revealed that the bone marrow-resident granulocyte pool exhibits a magnitude-order quantitative superiority over the circulating granulocyte pool [55,56,57].
Neutrophils are distinct from other immune cells as they are directly released into the peripheral circulation in a terminally differentiated mature state. The bone marrow not only regulates the homeostatic maturation of granulocytic precursors (spanning from myeloblasts to band-stage cells) but also serves as the final destination for peripheral circulating neutrophils. Approximately 30% of circulating neutrophils can re-enter the bone marrow via the CXCR4/CXCL12 axis-mediated homing mechanism, a process exhibiting pronounced 24-h circadian fluctuations [58,59].
Bone marrow-resident neutrophils exhibit unique immunomodulatory functions: under genotoxic stress, they secrete TNF-α to promote sinusoidal endothelial cell proliferation, thereby maintaining hematopoietic microenvironment homeostasis [60]. Adrenergic stimulation induces neutrophils to biosynthesize prostaglandin E2 (PGE2), which activates osteoblast function through prostaglandin E Receptor 4 (EP4) receptor-dependent mechanisms, consequently enhancing hematopoietic stem cell (HSC) homing and retention capabilities within the bone marrow microenvironment [61]. Furthermore, histidine decarboxylase-positive (HDC+) neutrophil subsets preserve quiescence and enhance regenerative potential in myeloid-biased HSCs [62].
Multiple molecular mechanisms regulate neutrophil residence in the bone marrow through a complex regulatory network: (1) Chemokine balance system: osteoblasts constitutively secrete CXCL12, anchoring mature granulocytes to the endosteal region via CXCR4, while endothelial cells and megakaryocytes produce CXCL1/2 to drive granulocyte mobilization through CXCR2 signaling [59,63,64,65,66,67]; (2) Adhesion molecule interactions: the Very Late Antigen-4-Vascular Cell Adhesion Molecule 1 (VLA-4-VCAM1) ligand-receptor pairing synergizes with proteases to reinforce neutrophil-stromal adhesion [68,69,70]; (3) Growth factor regulation: G-CSF disrupts retention equilibrium through triple mechanisms—promoting CXCR2 ligand (CXCL1 and CXCL2) expression in megakaryocytes through Thrombopoietin (THPO)-dependent pathways, suppressing CXCL12 synthesis in stromal cells, and downregulating CXCR4 receptor levels on neutrophils [67,71,72,73].

2.3. Circulatory Dynamics and Homeostatic Regulation

Under physiological homeostasis, neutrophils adhere to strict dynamic equilibrium: following release from the bone marrow, they undergo an approximately 12 h circulatory cycle (murine models indicate a half-life of ~8–10 h), with clearance ultimately achieved via tissue infiltration [58,74,75]. Notably, despite their transient residence in the bloodstream, their phenotypic alterations exhibit circadian rhythm-dependent aging characteristics, a phenomenon termed neutrophil aging [76]. Quantitative studies confirm that under steady-state conditions, the circulating pool constitutes merely 1–2% of the total systemic neutrophil population [77].
The maintenance of neutrophil homeostasis relies on an intricate multi-layered regulatory network external to the bone marrow: (1) IL-23/IL-17/G-CSF cascade regulation: Tissue-resident phagocytes (e.g., macrophages) control IL-17 production by γδ T cells, αβ T cells, and native lymphoid cells through IL-23 [78,79]. IL-17 drives bone marrow neutrophil release by inducing G-CSF production [80,81]. The timely clearance of apoptotic neutrophils (mediated by macrophages and dendritic cells) suppresses IL-23/IL-17 axis activity, forming a negative feedback loop [82,83,84,85]. (2) Gut microenvironment regulation: Gut commensal microbiota stimulate intestinal epithelial cells to secrete CXCL5, which promotes granulopoiesis through IL-17 induction [86,87]. (3) IL-1β as an inflammatory amplifier: IL-1β released during cellular damage or via inflammasome activation potently enhances IL-17 transcription, thereby amplifying G-CSF signaling [88,89].

3. Neutrophils as Key Mediators of Inflammation-to-Carcinogenesis

During the acute phase of bacterial or viral infections, neutrophils, as the vanguard of innate immunity, play a pivotal protective role. However, as the acute phase transitions to the chronic phase, neutrophils may engage in deleterious actions, promoting tumor formation and progression. The inflammation–cancer axis, first conceptualized by Virchow in 1863, now represents one of the hallmarks of cancer pathogenesis. In cellular pathology, he first described leukocyte infiltration within tumor microenvironments and pioneered the concept that chronic inflammation constitutes a critical soil for carcinogenesis [90,91]. Epidemiological studies reveal that ~25% of malignancies are directly linked to chronic inflammation, with pathological subtypes spanning multiple anatomical systems: gastrointestinal (esophagitis → esophageal cancer, colitis → colorectal cancer, pancreatitis → pancreatic cancer), hepatobiliary (hepatitis → hepatocellular carcinoma), respiratory (pneumonitis → lung cancer) [90,92,93,94,95,96,97,98]. Modern molecular pathology confirms that pathogen-associated molecular patterns (PAMPs) stimulate Toll-like receptor (TLR)-dependent NF-κB activation, establishing a pro-tumorigenic microenvironment through the continuous release of inflammatory cytokines such as IL-6 and TNF-α by neutrophils, thus providing the molecular basis for the transformation from inflammation to tumorigenesis [99]. Pathological analyses reveal that activated infiltrating neutrophils in inflammatory bowel disease (IBD) patients and experimental colon injury models secrete pro-inflammatory microvesicles enriched with oncogenic molecules (e.g., miR-23a/miR-155), which trigger genomic instability by suppressing DNA repair pathway genes [100]. Strikingly, miR-155 directly mediates neutrophil-associated DNA double-strand breaks and repair network dysregulation under acute colonic injury, driving colorectal carcinogenesis and malignant progression [101].
Reactive oxygen species (ROS), particularly those derived from neutrophils, play a pivotal role in chronic inflammatory lesions by inducing carcinogenic mutations, with colitis serving as a representative example [102,103]. Helicobacter pylori infection and IBD induce extensive neutrophil infiltration and oxidative burst, generating ROS and RNS, including superoxide (O2˙), H2O2, hypochlorous acid (HOCl), hydroxyl radicals (˙OH), and peroxynitrite, which drive gastric and colorectal carcinogenesis. Produced primarily via NADPH oxidase and mitochondrial electron transport chain, ROS/RNSs, while intended to eliminate pathogens, cause widespread host cell damage, including DNA lesions, lipid peroxidation, and protein oxidation. These insults provoke genomic instability, membrane dysfunction, and protein impairment, compromising cellular integrity. Additionally, ROSs act as second messengers, activating NF-κB, AP-1, and matrix metalloproteinase-1 (MMP1), upregulating IL-8, VEGF, and iNOS, thereby promoting inflammation, proliferation, survival, angiogenesis, and tumor invasion/metastasis. Oxidative stress further induces epigenetic silencing of tumor suppressor genes (e.g., p53, Rb, hMLH1, p16, p14) via DNA methylation. Collectively, neutrophil-driven oxidative stress significantly contributes to H. pylori-associated gastric cancer and IBD-related colorectal cancer by damaging cellular components, modulating signaling pathways, and inducing genetic/epigenetic aberrations.
Inflammatory responses act as key tumorigenic drivers by disrupting tissue homeostasis [94], with neutrophils functioning as central effector cells. Within the TME, soluble mediators and intercellular interactions form dynamic regulatory networks, where neutrophils act as hub nodes, constructing molecular bridges linking inflammatory processes to oncogenic pathways.

4. The Behavior of Neutrophils in the Tumor Progression and Tumor Metastasis

Neutrophils exhibit dual regulatory functions within the TME, actively participating in the dynamic regulation of metastatic cascades through either pro-tumorigenic or anti-tumorigenic mechanisms [104,105]. Their functional plasticity allows them to respond to a wide array of inflammatory signals and TME-derived factors, thereby exerting contrasting effects on tumor progression [106]. A clear understanding of these dual roles is essential for the development of effective neutrophil-targeted cancer therapies (see Figure 3 and Table 1). Furthermore, neutrophils play a pivotal role in promoting tumor cell migration, facilitating the formation of pre-metastatic niches, and reactivating dormant tumor cells at distant sites (see Figure 4 and Table 2).

4.1. The Dualistic Behavior of Neutrophils in the Tumor Progression

4.1.1. Pro-Tumorigenic Roles of Neutrophils

Pro-Angiogenesis

Neutrophils, particularly those polarized toward the N2 phenotype, are key contributors to tumor angiogenesis through the secretion of multiple pro-angiogenic factors, including FGF2, VEGF-A, ANGPT1, CXCL8, HGF, and MMP9 [36,104,105,107,108,109,110,111,112,113,114,115,116,117], which collectively facilitate tumor vascularization and metastatic spread. Studies have shown that metastasis-associated neutrophils exhibit markedly elevated FGF2 expression relative to their naïve counterparts, contributing to enhanced hepatic metastatic progression through increased vascular density and branching [107]. In addition, MMP9 secreted by these neutrophils further enhances tumor neoangiogenesis, thereby promoting the development of pancreatic and lung cancers [108]. Importantly, research has demonstrated that MMP9 released by bone marrow-derived neutrophils collaborates with other hematopoietic lineage cells, possibly by modulating the tumor microenvironment or extracellular matrix remodeling, to promote the initiation of squamous cell carcinoma [118].

NETs Formation

NETs promote tumor metastasis through both physical entrapment and molecular signaling mechanisms. Within their extracellular fibrous network, NETs capture circulating tumor cells (CTCs), shielding them from the cytotoxic effects of CD8+ T cells and NK cells while simultaneously facilitating metastatic adhesion [104,119,120]. Higher levels of circulating NETs have been shown to correlate with increased metastatic burden in advanced esophageal cancer, gastric cancer, and lung cancer [120]. Amyloid-beta, secreted by CAFs, promotes NETs formation and contributes to tumor progression [121]. At the mechanistic level, tumor cells utilize the CCDC25 receptor to recognize DNA associated with NETs, thereby activating the ILK-β-parvin signaling axis and enhancing the adhesion, motility, and proliferation of liver-metastatic cells [122]. This process accelerates the growth of liver metastases, providing direct evidence for the pro-metastatic role of NETs. Studies in mouse models further demonstrate that NETs infiltrating into peritumoral tissues can enhance metastatic potential [123,124]. Furthermore, inflammatory signals not directly linked to tumor initiation can also trigger neutrophil NETosis [125,126].

Immunosuppressive Microenvironment Remodeling

The suppression of T cell activity by neutrophils represents a critical mechanism underlying tumor immune escape, with distinct mechanisms observed in different tumor types: in colorectal cancer, N2-polarized neutrophils promote immune evasion by activating latent TGFβ via MMP9, thereby suppressing the function of tumor-infiltrating T cells [127]; in ovarian cancer, these TANs mediate IL-8-dependent immunosuppression (suppressing CD8+ T cells) through the Jagged2 signaling pathway [128]. Furthermore, tumor-derived small extracellular vesicles (sEVs) deliver serine protease inhibitors (serpins), which reprogram neutrophils toward a pro-tumorigenic phenotype characterized by an extended lifespan and an immunosuppressive expression pattern. These neutrophils functionally enhance tumor cell epithelial–mesenchymal transition and potently inhibit cytotoxic CD8+ T cells, thereby reducing their tumor-killing efficacy [129]. In the tumor microenvironment, IL-1β induces G-CSF production, promoting both neutrophil expansion and functional polarization. Tumor-educated neutrophils gain the capacity to suppress CD8+ T cell cytotoxicity, thereby facilitating tumor progression [130,131,132,133,134].

4.1.2. Anti-Tumorigenic Roles of Neutrophils

Oxidative Stress-Mediated Tumor Cell Killing

Neutrophils exert dual antitumor effects through a synergistic mechanism involving ROS and RNS. Mechanistic studies have demonstrated that H2O2 secreted by neutrophils activates the TRPM2 calcium channel on tumor cell membranes, leading to a rapid increase in intracellular Ca2+ levels. This elevation in Ca2+ subsequently initiates calpain-mediated activation of caspase-3, culminating in apoptotic cell death [135,136]. In addition, neutrophil-derived H2O2 has been shown to suppress pulmonary metastatic colonization [30]. Moreover, research by the Finisguerra group revealed that HGF interacts with the MET receptor on neutrophil surfaces, thereby activating the iNOS signaling axis and promoting NO production, which enhances tumor cell cytotoxicity [137]. Moreover, N1-polarized neutrophils potentiate anti-tumor immune responses and improve therapeutic outcomes via oxidative stress-mediated suppression of IL-17+ γδ T cells [138].

Protease-Dependent Selective Cytotoxicity

NE demonstrates a distinct ability to target tumors and has been shown to selectively destroy cancer cells while inhibiting tumorigenesis [139]. This targeted cytotoxic effect works alongside oxidative mechanisms, thereby enhancing the overall antitumor response.
Figure 3. The dualistic behavior of neutrophils in the tumor progression. Neutrophils exhibit functional plasticity and play a dual role in modulating tumor progression, which can be either tumor-promoting or tumor-suppressive, depending on cues from the microenvironment and epigenetic regulation. Tumor-promoting neutrophils contribute to cancer progression through multiple pathways, including the secretion of pro-angiogenic factors (e.g., VEGF, MMP9, FGF2), induction of immunosuppression (e.g., via TGFβ and NETs), and suppression of immune cell activity. In contrast, tumor-suppressive neutrophils mediate cytotoxicity through multiple mechanisms, such as the generation of ROS, RNS, and NE.
Figure 3. The dualistic behavior of neutrophils in the tumor progression. Neutrophils exhibit functional plasticity and play a dual role in modulating tumor progression, which can be either tumor-promoting or tumor-suppressive, depending on cues from the microenvironment and epigenetic regulation. Tumor-promoting neutrophils contribute to cancer progression through multiple pathways, including the secretion of pro-angiogenic factors (e.g., VEGF, MMP9, FGF2), induction of immunosuppression (e.g., via TGFβ and NETs), and suppression of immune cell activity. In contrast, tumor-suppressive neutrophils mediate cytotoxicity through multiple mechanisms, such as the generation of ROS, RNS, and NE.
Biomedicines 13 01473 g003
Table 1. Mechanisms underlying the role of neutrophils in tumor growth regulation.
Table 1. Mechanisms underlying the role of neutrophils in tumor growth regulation.
CharacteristicsMechanismsReferences
Anti-tumorNeutrophils suppress the proliferation of IL-17+ γδ T cells by secreting ROS, thereby exerting anti-tumor effects in melanoma and hepatocellular carcinoma.[138]
Neutrophils acquire anti-tumor phenotypes in breast cancer under the influence of ACE inhibitors and AGTR1 antagonists, thereby suppressing tumor growth.[140]
Neutrophils undergo N1-dominant reprogramming upon TGF-β blockade, resulting in effective tumor suppression across various cancers including breast, bile duct, colon, lung, and melanoma.[141,142,143,144,145,146,147,148]
Neutrophils secrete H2O2, which binds to the TRPM2 receptor on tumor cells, inducing Ca²+ influx and promoting tumor cell death through activation of the caspase-3 apoptosis signaling pathway in breast cancer.[135,136]
HGF/MET-dependent nitric oxide release by neutrophils promotes cancer cell killing in many tumors, including fibrosarcoma, colon cancer, lung cancer, melanoma, and hepatocellular carcinoma.[137]
Neutrophils inhibit metastatic seeding in the lungs by generating H2O2 in breast cancer, lung cancer, melanoma.[30]
Neutrophils release catalytically active NE, which hydrolyzes the CD95 death domain to selectively eliminate cancer cells in pan-cancers.[139]
Pro-tumorNeutrophils suppress tumor-infiltrating T cells in colon cancer via MMP9-mediated activation of TGFβ in colon cancer.[127]
Neutrophils contribute to skin carcinogenesis by releasing MMP9.[118]
CAFs secrete Amyloid β, which enhances the formation of NETs, thereby promoting tumor progression in melanoma, pancreatic cancer.[121]
Neutrophils promote angiogenesis through FGF2 secretion.[107]
Neutrophils promote angiogenesis through MMP9 secretion.[36,108,117]
IFN-β inhibits the production of VEGF and matrix MMP9 by neutrophils, consequently suppressing angiogenesis in melanoma.[111]
Neutrophils promote angiogenesis by releasing VEGF, HGF, and Angiopoietin-1.[112,113,114,115,116]
NETs promote tumor cell metastasis in esophageal, gastric, colon and lung cancer.[120]
Neutrophil-derived NETs engage the CCDC25 receptor on tumor cells, triggering ILK-β-parvin signaling and promoting metastasis in breast and colon cancer.[122]
NETs promote tumor cell metastasis in breast cancer.[123]
Lung inflammation promotes metastasis through neutrophil protease-mediated degradation of Tsp-1 in melanoma.[125]

4.2. The Role of Neutrophils in Tumor Metastasis

4.2.1. Facilitating Tumor Migration

Mechanisms of EMT Regulation

Neutrophils orchestrate tumor cell EMT by secreting mediators such as IL-17, TGF-β, and NE, which synergistically enhance transcriptional reprogramming [149,150,151]. Gastric cancer-derived HMGB1-enriched exosomes activate the TLR4/NF-κB pathway in neutrophils, triggering the autophagy-dependent secretion of IL-1β and OSM. This cascade markedly amplifies tumor cell migratory capacity [152].

Adhesive Support and Metastatic Niche Formation

Neutrophils facilitate metastatic colonization through both biomechanical interactions and molecular anchoring mechanisms. Clinical cohort analyses reveal a positive correlation between the proportion of circulating tumor cell–neutrophil clusters (CTC-Neu clusters) in peripheral blood and metastasis risk in breast cancer patients [153]. In murine melanoma models, IL-8-induced binding of neutrophil surface integrin β2 to tumor cell ICAM-1 stabilizes circulating tumor cells and potentiates their pulmonary dissemination [154]. Intravital microscopy further confirms that Lewis lung carcinoma cells form stable adhesions with neutrophils within hepatic microenvironments [155].

Metabolic Reprogramming and Energy Supply Networks

Neutrophils contribute to metastatic progression by remodeling lipid metabolism to provide energetic support for disseminating tumor cells [156]. In breast cancer lung metastasis models, angiopoietin-like 4 (ANGPTL4) secreted by pulmonary stromal cells impairs ATGL function in neutrophils, thereby promoting intracellular triglyceride storage. Through macropinocytosis-lysosomal pathways, single neutrophils transfer lipids to tumor cells, fulfilling their bioenergetic demands [156].

4.2.2. Promotion of Pre-Metastatic Niche Formation

Theoretical Evolution of Pre-Metastatic Niches

Stephen Paget’s 1889 “Seed and Soil” hypothesis [157] has been validated at the molecular level in modern metastasis research. This theory emphasizes that metastatic dissemination requires dynamic interactions between CTCs and distal organ microenvironments: CTCs, as biologically active “seeds,” must locate a “permissive soil”—termed the PMN—characterized by enhanced vascular permeability, infiltrated immunosuppressive cells, and extracellular matrix remodeling. Kaplan et al. first demonstrated the spatiotemporal plasticity of PMN through organ-specific recruitment experiments involving bone marrow-derived cells [158,159].

Neutrophils as Central Drivers of PMN Formation

In the MMTV-PyMT transgenic mouse model of spontaneous breast cancer metastasis, CD11b+Ly6G+ neutrophils form sentinel clusters at target sites before tumor cell arrival, with their density showing a strong positive correlation with subsequent metastatic burden [160]. Therapeutic targeting of neutrophils with anti-Ly6G antibodies markedly attenuated metastatic colonization in the lungs [160]. The nicotine-exposure further corroborates that early neutrophil infiltration into the lungs elevates the incidence of melanoma lung metastasis [161].

Exosome-Mediated Organotropic Programming

Tumor-derived exosomes (TDEs) remodel distal organ microenvironments through molecular navigation mechanisms. The priming effect of TDEs on neutrophils was first demonstrated in melanoma [162], with subsequent studies showing that exosomes from diverse metastatic tumors specifically home to their typical metastatic organs, establishing organotropic metastatic tropism [163]. Further research reveals that lung-tropic TDEs activate TLR3 in alveolar epithelial cells, inducing chemokine production and recruitment of immunosuppressive neutrophils [164]. These pro-metastatic neutrophils, recruited to the lungs, enhance metastasis in breast cancer models by releasing MMPs to modify pulmonary stromal architecture [165].

Regulatory Networks of NETs

In the 4T1 breast cancer model, mitochondria DNA-enriched NETs released by senescent neutrophils promote collagen remodeling and accelerate metastatic colonization [166]. Clinicopathological analyses of advanced ovarian cancer patients demonstrate elevated H3Cit+MPO+ NETs density in omental tissues compared to early-stage cases, correlating significantly with reduced progression-free survival [124]. Another study highlights that NETs-driven PMN formation depends on complex interactions among diverse stromal cell populations within the tumor microenvironment [167].

4.2.3. Activation of Dormant Tumor Cells

Biological Features of Tumor Cell Dormancy

Metastatic tumor cells can remain in a dormant state characterized by dynamic equilibrium between proliferation and apoptosis. This process involves impaired angiogenic mimicry and cell cycle arrest. Clinical cohort studies demonstrate that even after successful resection of primary tumors, patients may experience metastatic recurrence due to activation of dormant cells [168]. This dormant state is generally maintained until external stimuli from the microenvironment reignite proliferative activity [169].

NET-Mediated Reactivation of Dormant Cells

Chronic inflammation-induced NETs serve as critical drivers of dormant cell activation. NE and MMP9 released from NETs cleave adhesion proteins and activate integrin α3β1-dependent signaling, thereby reigniting dormant tumor cell proliferation [126]. Integrin signaling, particularly through integrin β1, has been implicated in regulating the switch from tumor dormancy to metastatic proliferation [169]. Of clinical relevance, data from breast cancer patients with COVID-19 comorbidity reveal a positive correlation between pulmonary NETs density and circulating tumor cell activation rates, suggesting that virus-associated pneumonia may accelerate tumor recurrence via NETs-mediated mechanisms [126,170,171].

The Regulatory Role of Secreted Protein

Using murine models—including Mmp9−/− mice and anti-Ly6G antibody-mediated neutrophil depletion—experimental evidence reveals that 14,15-epoxyeicosatrienoic acid (14,15-EET) initiates a pro-metastatic cascade involving neutrophil-dependent MMP9 production, which both enhances angiogenesis and disrupts tumor dormancy [172].

Stress Hormone-Driven Activation

In ovarian and pulmonary neoplasms, stress hormone signaling activates neutrophils, which in turn facilitates the transition from dormant micrometastases to clinically detectable recurrences [173]. This activation involves a tripartite cascade: S100A8/A9 protein secretion, upregulated MPO activity, and oxidized lipid accumulation. These events converge to activate dormant tumor cells via the FGF signaling axis, facilitating their transition from quiescence to proliferative states [173].
Figure 4. The role of neutrophils in tumor metastasis. Neutrophils drive tumor migration, PMN formation, and dormant tumor cell activation. They promote EMT via IL-17, IL-1β, OSM, TGF-β, and NE, enhance metastatic adhesion through CTC-Neu clusters, and support tumor energy needs via lipid transfer. Neutrophils form PMNs by remodeling stromal architecture and releasing MMPs and NETs, which also reactivate dormant cells by cleaving adhesion proteins and activating integrin signaling. Chronic inflammation, COVID-19, and 14,15-EET can also activate tumor cells. Tumor-derived exosomes and stress hormones further amplify neutrophil-driven metastasis, highlighting their critical role in cancer progression and recurrence.
Figure 4. The role of neutrophils in tumor metastasis. Neutrophils drive tumor migration, PMN formation, and dormant tumor cell activation. They promote EMT via IL-17, IL-1β, OSM, TGF-β, and NE, enhance metastatic adhesion through CTC-Neu clusters, and support tumor energy needs via lipid transfer. Neutrophils form PMNs by remodeling stromal architecture and releasing MMPs and NETs, which also reactivate dormant cells by cleaving adhesion proteins and activating integrin signaling. Chronic inflammation, COVID-19, and 14,15-EET can also activate tumor cells. Tumor-derived exosomes and stress hormones further amplify neutrophil-driven metastasis, highlighting their critical role in cancer progression and recurrence.
Biomedicines 13 01473 g004
Table 2. Mechanisms underlying the role of neutrophils in tumor metastasis.
Table 2. Mechanisms underlying the role of neutrophils in tumor metastasis.
CharacteristicsMechanismsReferences
Facilitating tumor migrationTANs produce IL-17a, which promotes EMT of GC cells through JAK2/STAT3 signaling in gastric cancer.[149]
Neutrophils secrete NE, which cleaves E-cadherin on tumor cell surfaces while inducing nuclear translocation of β-catenin and Zeb1, promoting tumor cell EMT in pancreatic ductal adenocarcinoma.[150]
GC-Ex activates neutrophils through the HMGB1/TLR4/NF-κB signaling pathway, thereby promoting tumor metastasis in gastric cancer.[152]
Clinical cohort analysis shows that the proportion of circulating tumor cell-neutrophil clusters in the peripheral blood of breast cancer patients is positively correlated with metastasis risk in breast cancer.[153]
ICAM-1 on melanoma cells and β2 integrin on neutrophils interacted, promoting anchoring to vascular endothelium in melanoma.[154]
The adhesion of lipopolysaccharide-activated neutrophils to cancer cells was mediated by neutrophil Mac-1/ICAM-1 in lung cancer.[155]
Upon contact with MC cells, neutrophils experience ATGL suppression, leading to intracellular lipid accumulation, which is transferred to tumor cells via the macropinocytosis-lysosome pathway, promoting tumor metastasis in breast cancer.[156]
Neutrophils accumulate in the pre-metastatic lung microenvironment and promote tumor cell colonization by secreting leukotrienes in breast cancer.[160]
Promotion of pre-metastatic niche formationNET formation in rendering the PMN conducive for implantation of ovarian cancer cells, while PAD4 plays a critical role in NETs formation in ovarian cancer.[124]
Chronic nicotine exposure induces neutrophil recruitment in the lung, where neutrophils release LCN2, promoting MET in tumor cells, thereby enhancing their colonization and metastatic potential in breast cancer.[161]
Lung epithelial cells are critical for initiating neutrophil recruitment and lung metastatic niche formation by sensing tumor exosomal RNAs via TLR3 in melanoma.[164]
Exosomes from highly metastatic melanoma increased the metastatic behavior of primary tumors by permanently “educating” bone marrow progenitors via the MET receptor in melanoma.[162]
Neutrophils operate to facilitate extravasation of tumor cells through the secretion of IL1β and matrix metalloproteinases in breast cancer and melanoma.[165]
Neutrophils promote breast cancer lung metastasis through the SIRT1-Naged-NETs axis in breast cancer.[166]
LMSCs promote neutrophil recruitment and NETs formation by secreting C3, thereby facilitating cancer cell metastasis to the lung in breast cancer.[167]
Dormancy activationNE and MMP9 in NETs promote ECM remodeling, activating the integrin α3β1-FAK/ERK/MLCK/YAP signaling pathway to enhance dormant tumor cell proliferation in breast cancer.[126]
14,15-EET induces G-CSF/IL-6 production in vivo, enhancing STAT3 activation in neutrophils to promote MMP-9 expression and suppress TRAIL expression, with neutrophil-derived MMP-9 being essential for inducing angiogenesis in dormant micrometastases in melanoma.[172]
Stress hormones trigger release of S100A8/A9 proteins from neutrophils, which activate MPO, leading to oxidized lipid accumulation. These lipids, upon release, upregulate the fibroblast growth factor pathway in tumor cells, promoting their exit from dormancy and formation of new tumor lesions in lung cancer.[173]

5. Single-Cell and Spatial Omics Uncover Neutrophil Heterogeneity and Therapeutic Targets

Single-cell and spatial omics have elucidated the precise localization and functional roles of neutrophils within the TME across various cancer types, highlighting their heterogeneity and interactions with other cellular components. In prostate cancer, integrated scRNA sequencing and high-resolution spatial transcriptomics revealed an immunosuppressive TME driven by myeloid cells, including neutrophils, and exhausted T cells, contributing to tumor recurrence and metastasis [174]. In non-small cell lung cancer (NSCLC), single-cell and spatial transcriptomic analyses underscored the pervasive presence of myeloid cells, including neutrophils, and their pivotal role in disease progression, providing a high-resolution molecular map of their contributions to the TME [175]. For pancreatic ductal adenocarcinoma (PDAC), multi-omics analyses have elucidated the heterogeneity of neutrophils in the PDAC tumor microenvironment at single-cell resolution, identifying four distinct neutrophil subsets and revealing their dynamic functions, metabolic reprogramming, and regulatory mechanisms [176]. Additionally, single-cell and spatial transcriptomic studies have characterized multicellular dynamics associated with neoadjuvant therapy in PDAC, providing a framework for understanding neutrophil contributions to the spatial TME, despite not directly addressing them [177]. In ovarian cancer, a microfluidic-based TIME-on-Chip model demonstrated that NETs promote collective tumor invasion, offering an in vitro platform to study neutrophil spatial functions [178]. In glioblastoma: single-cell RNA sequencing highlighted the emergence of pro-tumor myeloid-derived suppressor cells (MDSCs), including neutrophils, during disease progression [179]. In Gastric cancer, single-cell RNA sequencing post-PD-1 therapy identified increased circulating neutrophils, with the NE-1 subset exhibiting an activated phenotype (high MMP9, S100A8, S100A9, PORK2, TGF-β1 expression) and interacting with malignant epithelial cells and M2 macrophages via chemokine pathways to promote tumor progression [180]. Pan-cancer analyses across 17 cancer types and 225 samples revealed transcriptional heterogeneity among tumor-associated neutrophils, forming 10 distinct states; HLA-DR+CD74+ antigen-presenting neutrophils correlated with improved survival, while VEGFA+SPP1+ angiogenic neutrophils were associated with poorer prognosis. Interventions like leucine diets or delivery of antigen-presenting neutrophils enhanced anti-PD-1 immunotherapy in mouse models and human tumor fragment models, suggesting novel neutrophil-based therapeutic strategies [181]. Additionally, a single-cell tumor immune atlas from 217 patients across 13 cancers, integrated with the SPOTlight tool, enabled in situ immune population mapping, supporting patient stratification based on immune composition [182].
Spatial omics, integrated with single-cell omics, will advance our understanding of neutrophil heterogeneity, spatial distribution, and their complex interactions with tumor progression and therapeutic responses within the tumor microenvironment. These high-resolution molecular profiles, coupled with in vitro models simulating three-dimensional tumor microenvironments, will provide a robust foundation for developing precise neutrophil-targeted anticancer therapies.

6. Targeting Neutrophils in Cancer Therapy: Strategies and Approaches

TANs play a pivotal role in tumor progression. This section explores various strategies to target and inhibit TANs recruitment, as well as methods for monitoring and modulating neutrophil behavior to improve cancer treatment outcomes.

6.1. Strategies to Inhibit TANs Recruitment

The recruitment of TANs is a critical event in tumor progression, and targeting this process offers various strategies for cancer therapy. Chemokines and their receptors play an essential role in the migration of neutrophils to tumor sites. For example, CXCL2 promotes neutrophil recruitment and inflammatory responses via the CXCR2 axis, and inhibiting CXCR2 can alleviate myeloid inflammation and reverse treatment resistance in prostate cancer [183,184]. The ALX/FPR2 receptor regulates neutrophil infiltration based on ligands such as serum amyloid A, and targeting this receptor holds promise for controlling tumor-associated inflammation [185]. CCL11, a multifunctional chemokine, has a dual role in cancer, demonstrating potential in immune therapy [186]. CXCL5 has been also identified as a key chemokine driving the infiltration of mature pro-tumor neutrophils into lung cancer tissues [187]. Nevertheless, precisely blocking these chemokines without affecting systemic immune function remains a challenge. Additionally, sialic acid-modified nanoplatforms can interfere with the physical or molecular interactions between neutrophils and the tumor microenvironment, blocking their infiltration and enhancing the efficacy of checkpoint blockade therapies [188]. Another strategy involves the direct clearance of pro-tumor myeloid cells, such as PMN-MDSCs, using near-infrared immunotherapy with Ly6G antibodies to selectively eliminate neutrophils in the tumor bed, significantly inhibiting tumor growth and enhancing host immune responses [189,190]. Further studies have shown that pathogens in the tumor microenvironment, such as Fusobacterium nucleatum, recruit TANs by activating the IL-17/NF-κB/RelB pathway, promoting gastric cancer progression, suggesting that targeting the microbiome may indirectly modulate neutrophil behavior [191]. These strategies provide diverse pathways for precisely intervening in neutrophil recruitment.

6.2. Regulation of Neutrophil-Derived Cytokine Release

Neutrophils, as critical effector cells in inflammation and immune regulation, can synthesize and secrete various cytokines, including pro-inflammatory cytokines (TNF-α, IL-1β), chemokines (IL-8, IP-10, MIP-1α), and angiogenic factors (VEGF). These factors not only promote inflammation and immune responses within the tumor microenvironment but also accelerate tumor progression [192]. Therefore, inhibiting the secretion of these cytokines by neutrophils is considered a potential strategy for immunotherapy. TGF-β1 activates the SMAD3 and ERK1/2 signaling pathways, inducing neutrophils in the tumor microenvironment to express tumor-promoting factors, such as OSM and VEGFA mRNA, thereby converting neutrophils into a pro-tumor phenotype [193]. Targeting the TGF-β1 signaling pathway can inhibit the pro-tumor functions of neutrophils, offering a new therapeutic target for precise modulation of the tumor microenvironment.

6.3. Bispecific Antibodies Enhance Neutrophil-Mediated Antitumor Activity

The novel bispecific antibody (TrisomAb) effectively recruits neutrophils as effector cells, enhancing their cytotoxicity against tumor cells. Studies have shown that, in colorectal cancer patients, when neutrophils are able to efficiently eliminate tumor cells are exposed to anti-EGFR TrisomAb [194].

6.4. Innovative Carriers for Tumor-Targeted Nanodrug Delivery

Neutrophils, as core effector cells of innate immunity, have become ideal carriers for targeted nanodrug delivery in brain tumors due to their rapid inflammatory response, strong chemotaxis, and ability to cross the blood-brain barrier (BBB) [195,196,197]. These cells actively participate in inflammation by forming NETs and releasing cytokines, providing a new mechanism for precise drug delivery [195] (Table 3). In conditions of neuroinflammation or pathological states, the integrity of the BBB is compromised, which facilitates neutrophil migration into the central nervous system (CNS) [198,199]. Acting as “Trojan horses,” neutrophils can carry nanodrugs and use their pro-inflammatory properties to deliver anti-cancer drugs precisely to residual glioma cells after surgery, significantly inhibiting tumor recurrence and extending mouse survival [197]. Biomimetic nanocarriers, designed to mimic the surface characteristics of neutrophils, possess immune evasion and targeting capabilities, effectively overcoming the limitations of the BBB and the tumor microenvironment, thereby enhancing drug accumulation and therapeutic efficacy. In a breast cancer model, these carriers demonstrated potential in modulating the microenvironment and inhibiting metastasis, offering new insights into brain tumor treatment [200,201]. Additionally, neutrophil-derived exosomes (NEs-Exos/DOX) leverage their strong inflammatory chemotaxis and BBB penetration ability to successfully deliver doxorubicin to gliomas, significantly inhibiting tumor growth, extending survival, and reducing systemic toxicity [202]. In a breast cancer model, local inflammation induced the recruitment of neutrophils and the release of NETs, further enhancing anti-tumor effects. At the same time, neutrophil infusion helped alleviate systemic inflammation, ensuring clinical safety [203] (Table 3). These studies collectively highlight the unique advantages of neutrophils and their derived carriers in overcoming the BBB barrier and enhancing targeted brain tumor therapy, providing innovative strategies for precision treatment and suggesting substantial clinical potential.

6.5. Neutrophil-Based Combination Therapy

6.5.1. Combination with Immune Checkpoint Inhibitors

In a glioma mouse model, neutrophil depletion combined with anti-PD-1 antibody treatment significantly inhibited glioma growth and extended mouse survival [204]. In a non-small cell lung cancer (NSCLC) mouse model, CXCL5 gene knockout completely prevented neutrophil accumulation in the lung tissue, which in turn promoted the expansion and cytolytic function of tumor-specific CD8+ T cells [187]. Additionally, studies have shown that the synergistic action of T cells and neutrophils can enhance the efficacy of immunotherapy [205].

6.5.2. Combination with Chemotherapy

In pancreatic cancer patients, combining anti-Ly6G therapy to deplete neutrophils with gemcitabine/paclitaxel chemotherapy not only significantly reduced tumor burden and metastatic growth but also enhanced the functionality of tumor-infiltrating CD8+ T cells, effectively suppressed the polarization of CAF, and inhibited chemotherapy resistance via the IL-6/STAT-3 signaling pathway [206].

6.5.3. Other Combination Strategies with Neutrophil Modulators

The role of neutrophils in cancer immunotherapy has been significantly enhanced through various pharmacological and biological interventions. Pharmacological interventions, such as angiotensin-converting enzyme inhibitors (ACEIs) and angiotensin II type 1 receptor (AGTR1) antagonists, can drive neutrophil polarization toward the antitumor N1 phenotype, thereby enhancing their cytotoxic potential [140]. Clinically, increased intratumoral infiltration of N1 neutrophils has been correlated with improved patient outcomes. For example, suppression of the TGF-β signaling pathway has been shown to reprogram neutrophils into the N1 phenotype, leading to effective tumor suppression [141,142,143,144,145,146,147,148]. Computational modeling has further refined the regulation of N1/N2 neutrophil balance by TGF-β inhibitors and IFN-β, favoring N1 recruitment and significantly impeding tumor progression [207]. Moreover, melatonin has been found to enhance antitumor immunity through modulation of tumor-associated neutrophil infiltration and NETosis in pancreatic ductal adenocarcinoma [208]. Probiotics have also been implicated in exerting therapeutic benefits through the inhibition of neutrophil-mediated cancer metastasis [209].

6.6. Neutrophil-Lymphocyte Ratio (NLR): A Critical Biomarker for Cancer Prognosis Evaluation

Although clinical treatments targeting neutrophils are still in the exploratory stage, several studies in preclinical models and early clinical observations suggest that neutrophil-targeted therapies may improve patient prognosis. Research has shown that an increase in peripheral blood neutrophil count or NLR is an independent predictor of poor prognosis in various cancers. For example, in colorectal cancer patients, elevated neutrophil count is associated with a shorter overall survival (OS) [210]. In glioma patients, the presence of CTCs post-surgery is closely linked to neutrophil-mediated inflammatory immune environments and correlates with poor prognosis [211]. In NSCLC patients, persistent neutrophil elevation and a high NLR (≥5) are associated with negative prognostic value [212]. Additionally, in gastric cancer patients receiving PD-1 antibody treatment, high NLR is also linked to adverse outcomes [180]. These findings highlight the critical role of neutrophils in cancer progression and suggest their potential as therapeutic targets.
In pancreatic cancer patients, dynamic changes in NLR before and during chemotherapy are closely associated with pathological response, disease-free survival (DFS), and OS [206]. Some studies have also proposed that a TICO regimen, consisting of tadalafil, isotretinoin, colchicine, and Omega-3 fatty acids, may inhibit cancer growth and metastasis by reducing high NLR [213]. Although clinical data for this regimen still require validation through large-scale trials, its theoretical foundation and the safety of the drugs involved provide promising possibilities for future clinical applications.
Neutrophils and related indicators, such as NLR, not only serve as prognostic biomarkers but may also guide personalized treatment. Monitoring neutrophils in the tumor microenvironment is crucial for tumor treatment and prognosis assessment. Advances in molecular imaging technologies now enable non-invasive visualization of neutrophil biological behavior, offering strong support for personalized cancer diagnostics and therapeutic strategies [214].
Table 3. The role of NETs in different cancer types.
Table 3. The role of NETs in different cancer types.
Tumor TypeMechanismsReferences
Breast cancerPromote tumor cell proliferation, intravascular infiltration, and distant metastasis, PMN formation, and awakening of dormant cancer cells.[215,216,217,218,219]
NeuroblastomaPlay a key role in tumor cell proliferation, metastasis, and immune escape.[220]
Oropharyngeal squamous cell carcinomaIn the initial stages of tumor formation, it may suppress tumorigenesis by clearing bacteria; after the tumor is established, it promotes tumor progression by releasing cytokines and chemokines.[221]
Gastric cancerPromote tumor progression (e.g., IL-8-induced NETs); facilitate EMT and metastasis through the PAI-1/TGF-β axis.[222,223,224]
Colorectal cancerPromote tumor progression and liver metastasis.[225,226,227,228,229]
Esophageal squamous cell carcinomaPromote tumor cell proliferation, migration, invasion, and angiogenesis; CCDC25 is associated with poor prognosis.[230,231]
OsteosarcomaHigh levels of NETs formation in diagnostic biopsies are associated with poor response to neoadjuvant chemotherapy and worse overall survival; NETs levels are higher in metastatic sites than in primary lesions.[232,233]
Ovarian cancerPromote the formation of the pre-metastatic niche in the peritoneum.[234]
Pancreatic adenocarcinomaMelatonin has been found to enhance anti-tumor immunity by regulating TANs infiltration and NETosis.[208]

7. Conclusions and Future Perspectives

In summary, the functional heterogeneity and plasticity of neutrophils play a pivotal role in tumor initiation, progression, and metastasis. These cells exhibit a dual role: either promoting inflammatory responses and tumor metastasis, or acquiring anti-tumorigenic phenotypes under specific conditions. This functional duality positions neutrophils as critical regulators within the tumor microenvironment, highlighting their potential as targets for innovative anti-cancer therapeutic strategies.
However, current research faces several limitations and challenges: 1. Unclear Mechanisms: the intricate molecular mechanisms underlying the interactions among neutrophils, tumor cells, and other immune components, along with their evolving roles during cancer progression, are not yet fully elucidated. 2. High Variability: neutrophil behavior displays significant heterogeneity across different cancer types, individual patients, and tumor stages. This variability complicates mechanistic studies and limits the development of broadly applicable therapeutic strategies. 3. Technical Limitations: current methodologies, including single-cell sequencing and advanced imaging technologies, are often expensive, technically challenging, and may lack standardization. Accurately characterizing neutrophil functional states and identifying tumor-specific subpopulations remains a major technical barrier.
To address these limitations, future research should focus on the following areas:1. In-depth Mechanistic Studies: Utilizing cutting-edge approaches such as gene editing (e.g., CRISPR-Cas9) and multi-omics profiling to dissect the precise and context-dependent interactions between neutrophils, tumor cells, and immune cells. These efforts aim to uncover both shared and cancer-specific molecular mechanisms. 2. Clinical Translation: Building upon deeper mechanistic understanding, integrating artificial intelligence and big data analytics to identify novel diagnostic and therapeutic targets. This will facilitate the development of personalized precision medicine strategies for early cancer detection, diagnosis, and monitoring of treatment responses. 3. Technological Advancements: Advancing the development of cost-effective, user-friendly tools—such as high-throughput sequencing platforms and high-resolution imaging systems—to improve accessibility of affordable diagnostics and alleviate the clinical burden of cancer.

Author Contributions

H.L. and C.Y. provided direction and guidance throughout the preparation of this manuscript. P.L. wrote and edited the manuscript. F.F. and B.Z. reviewed and contributed to the revision of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

Open Project of the Key Laboratory of Organ Transplantation, Ministry of Education, and NHC, Grant/Award Number: No. 2023QYKF03; Hubei Provincial Natural Science Foundation, Grant/Award Number: 2015CFB462; National Natural Science Foundation of China, Grant/Award Number: 81202300, 8240035219 and 82373052.

Acknowledgments

Figures presented in this review were generated by Figdraw.

Conflicts of Interest

The authors declare that there are no conflicts of interest.

Abbreviations

14,15-EET14,15-epoxyeicosatrienoic acid
ACEIsangiotensin-converting enzyme inhibitors
AGTR1angiotensin II receptor type 1
ANGPT1Angiopoietin 1
ANGPTL4angiopoietin-like 4
CAFscancer-associated fibroblasts
CTCscirculating tumor cells
EMTepithelial-mesenchymal transition
EP4prostaglandin E Receptor 4
G-CSFGranulocyte Colony-Stimulating Factor
GC-Exgastric cancer cell-derived exosomes
GMPgranulocyte-monocyte progenitor
HDC+decarboxylase-positive
HSChematopoietic stem cell
ICAM-1intercellular adhesion molecule 1
ITGB2integrin β2
LMPPslymphoid-primed multipotent progenitors
MMP9matrix metalloproteinase 9
MMPsmatrix metalloproteinases
MPOmyeloperoxidase
mtDNAmitochondrial DNA
NEneutrophil elastase
NETsneutrophil extracellular traps
NOnitric oxide
NSAIDsnon-steroidal anti-inflammatory drugs
OSMOncostatin M
PAMPspathogen-associated molecular patterns
PGE2prostaglandin E2
PMNpre-metastatic niche
RNSreactive nitrogen species
ROSreactive oxygen species
S100A8/A9s100 calcium-binding protein A8/A9
TAMstumor-associated macrophages
TANstumor-associated neutrophils
TDEsTumor-derived exosomes
THPOThrombopoietin
TLRToll-like receptor
TMEtumor microenvironment
VCAM1Vascular cell adhesion molecule 1
VLA-4Very late antigen 4

References

  1. Coffelt, S.B.; Wellenstein, M.D.; De Visser, K.E. Neutrophils in Cancer: Neutral No More. Nat. Rev. Cancer 2016, 16, 431–446. [Google Scholar] [CrossRef] [PubMed]
  2. Mayadas, T.N.; Cullere, X.; Lowell, C.A. The Multifaceted Functions of Neutrophils. Annu. Rev. Pathol. Mech. Dis. 2014, 9, 181–218. [Google Scholar] [CrossRef] [PubMed]
  3. Yvan-Charvet, L.; Ng, L.G. Granulopoiesis and Neutrophil Homeostasis: A Metabolic, Daily Balancing Act. Trends Immunol. 2019, 40, 598–612. [Google Scholar] [CrossRef]
  4. Thanabalasuriar, A.; Scott, B.N.V.; Peiseler, M.; Willson, M.E.; Zeng, Z.; Warrener, P.; Keller, A.E.; Surewaard, B.G.J.; Dozier, E.A.; Korhonen, J.T.; et al. Neutrophil Extracellular Traps Confine Pseudomonas Aeruginosa Ocular Biofilms and Restrict Brain Invasion. Cell Host Microbe 2019, 25, 526–536.e4. [Google Scholar] [CrossRef]
  5. Liu, X.; Shin, S. Listening In: Plasmacytoid DC, Monocyte-Derived DC, and Neutrophil Crosstalk in Antifungal Defense. Cell Host Microbe 2020, 28, 9–11. [Google Scholar] [CrossRef]
  6. Iversen, M.B.; Reinert, L.S.; Thomsen, M.K.; Bagdonaite, I.; Nandakumar, R.; Cheshenko, N.; Prabakaran, T.; Vakhrushev, S.Y.; Krzyzowska, M.; Kratholm, S.K.; et al. An Innate Antiviral Pathway Acting before Interferons at Epithelial Surfaces. Nat. Immunol. 2016, 17, 150–158. [Google Scholar] [CrossRef]
  7. Castanheira, F.V.S.; Kubes, P. Neutrophils and NETs in Modulating Acute and Chronic Inflammation. Blood 2019, 133, 2178–2185. [Google Scholar] [CrossRef] [PubMed]
  8. Ma, Y.; Yabluchanskiy, A.; Iyer, R.P.; Cannon, P.L.; Flynn, E.R.; Jung, M.; Henry, J.; Cates, C.A.; Deleon-Pennell, K.Y.; Lindsey, M.L. Temporal Neutrophil Polarization Following Myocardial Infarction. Cardiovasc. Res. 2016, 110, 51–61. [Google Scholar] [CrossRef]
  9. Mistry, P.; Nakabo, S.; O’Neil, L.; Goel, R.R.; Jiang, K.; Carmona-Rivera, C.; Gupta, S.; Chan, D.W.; Carlucci, P.M.; Wang, X.; et al. Transcriptomic, Epigenetic, and Functional Analyses Implicate Neutrophil Diversity in the Pathogenesis of Systemic Lupus Erythematosus. Proc. Natl. Acad. Sci. USA 2019, 116, 25222–25228. [Google Scholar] [CrossRef]
  10. Kuley, R.; Duvvuri, B.; Hasnain, S.; Dow, E.R.; Koch, A.E.; Higgs, R.E.; Krishnan, V.; Lood, C. Neutrophil Activation Markers and Rheumatoid Arthritis Treatment Response to the JAK1/2 Inhibitor Baricitinib. Arthritis Rheumatol. 2025, 77, 395–404. [Google Scholar] [CrossRef]
  11. Zhang, H.; Wang, Y.; Qu, M.; Li, W.; Wu, D.; Cata, J.P.; Miao, C. Neutrophil, Neutrophil Extracellular Traps and Endothelial Cell Dysfunction in Sepsis. Clin. Transl. Med. 2023, 13, e1170. [Google Scholar] [CrossRef] [PubMed]
  12. Herster, F.; Bittner, Z.; Archer, N.K.; Dickhöfer, S.; Eisel, D.; Eigenbrod, T.; Knorpp, T.; Schneiderhan-Marra, N.; Löffler, M.W.; Kalbacher, H.; et al. Neutrophil Extracellular Trap-Associated RNA and LL37 Enable Self-Amplifying Inflammation in Psoriasis. Nat. Commun. 2020, 11, 105. [Google Scholar] [CrossRef]
  13. Márquez-Coello, M.; Ruiz-Sánchez, C.; Martín-Aspas, A.; Fernández Gutiérrez Del Álamo, C.; Illanes-Álvarez, F.; Cuesta-Sancho, S.; Girón-González, J.-A. Neutrophil Expression of T and B Immunomodulatory Molecules in HIV Infection. Front. Immunol. 2021, 12, 670966. [Google Scholar] [CrossRef]
  14. Jia, M.; Fu, H.; Jiang, X.; Wang, L.; Xu, J.; Barnes, P.J.; Adcock, I.M.; Liu, Y.; He, S.; Zhang, F.; et al. DEL-1, as an Anti-neutrophil Transepithelial Migration Molecule, Inhibits Airway Neutrophilic Inflammation in Asthma. Allergy 2024, 79, 1180–1194. [Google Scholar] [CrossRef]
  15. Nakazawa, D.; Masuda, S.; Tomaru, U.; Ishizu, A. Pathogenesis and Therapeutic Interventions for ANCA-Associated Vasculitis. Nat. Rev. Rheumatol. 2019, 15, 91–101. [Google Scholar] [CrossRef] [PubMed]
  16. Du, X.; Ren, B.; Li, C.; Li, Q.; Kan, S.; Wang, X.; Bai, W.; Wu, C.; Kassegne, K.; Yan, H.; et al. PRL2 Regulates Neutrophil Extracellular Trap Formation Which Contributes to Severe Malaria and Acute Lung Injury. Nat. Commun. 2024, 15, 881. [Google Scholar] [CrossRef] [PubMed]
  17. Gajewski, T.F.; Schreiber, H.; Fu, Y.-X. Innate and Adaptive Immune Cells in the Tumor Microenvironment. Nat. Immunol. 2013, 14, 1014–1022. [Google Scholar] [CrossRef]
  18. Joyce, J.A.; Fearon, D.T. T Cell Exclusion, Immune Privilege, and the Tumor Microenvironment. Science 2015, 348, 74–80. [Google Scholar] [CrossRef]
  19. Treffers, L.W.; Hiemstra, I.H.; Kuijpers, T.W.; Van Den Berg, T.K.; Matlung, H.L. Neutrophils in Cancer. Immunol. Rev. 2016, 273, 312–328. [Google Scholar] [CrossRef]
  20. Binnewies, M.; Roberts, E.W.; Kersten, K.; Chan, V.; Fearon, D.F.; Merad, M.; Coussens, L.M.; Gabrilovich, D.I.; Ostrand-Rosenberg, S.; Hedrick, C.C.; et al. Understanding the Tumor Immune Microenvironment (TIME) for Effective Therapy. Nat. Med. 2018, 24, 541–550. [Google Scholar] [CrossRef]
  21. Shen, M.; Hu, P.; Donskov, F.; Wang, G.; Liu, Q.; Du, J. Tumor-Associated Neutrophils as a New Prognostic Factor in Cancer: A Systematic Review and Meta-Analysis. PLoS ONE 2014, 9, e98259. [Google Scholar] [CrossRef]
  22. Carus, A.; Ladekarl, M.; Hager, H.; Nedergaard, B.S.; Donskov, F. Tumour-Associated CD66b+ Neutrophil Count Is an Independent Prognostic Factor for Recurrence in Localised Cervical Cancer. Br. J. Cancer 2013, 108, 2116–2122. [Google Scholar] [CrossRef]
  23. Rao, H.-L.; Chen, J.-W.; Li, M.; Xiao, Y.-B.; Fu, J.; Zeng, Y.-X.; Cai, M.-Y.; Xie, D. Increased Intratumoral Neutrophil in Colorectal Carcinomas Correlates Closely with Malignant Phenotype and Predicts Patients’ Adverse Prognosis. PLoS ONE 2012, 7, e30806. [Google Scholar] [CrossRef]
  24. Kargl, J.; Busch, S.E.; Yang, G.H.Y.; Kim, K.-H.; Hanke, M.L.; Metz, H.E.; Hubbard, J.J.; Lee, S.M.; Madtes, D.K.; McIntosh, M.W.; et al. Neutrophils Dominate the Immune Cell Composition in Non-Small Cell Lung Cancer. Nat. Commun. 2017, 8, 14381. [Google Scholar] [CrossRef]
  25. Jaillon, S.; Ponzetta, A.; Di Mitri, D.; Santoni, A.; Bonecchi, R.; Mantovani, A. Neutrophil Diversity and Plasticity in Tumour Progression and Therapy. Nat. Rev. Cancer 2020, 20, 485–503. [Google Scholar] [CrossRef] [PubMed]
  26. Mackey, J.B.G.; Coffelt, S.B.; Carlin, L.M. Neutrophil Maturity in Cancer. Front. Immunol. 2019, 10, 1912. [Google Scholar] [CrossRef] [PubMed]
  27. Townsend, M.; Galbraith, M.A.; Ellber, R. Selective Reduction of Human Tumor Cell Populations by Human Granulocytesin Vitro. Cancer Res. 1978, 38, 4534–4539. [Google Scholar] [PubMed]
  28. Gerrard, T.L.; Kaplan, A.M. Human Neutrophil-Mediated Cytotoxicity to Tumor Cells. J. Natl. Cancer Inst. 1981, 66, 483–488. [Google Scholar] [PubMed]
  29. Cameron, D.J. A Comparison of the Cytotoxic Potential in Polymorphonuclear Leukocytes Obtained from Normal Donors and Cancer Patients. Clin. Immunol. Immunopathol. 1983, 28, 115–124. [Google Scholar] [CrossRef]
  30. Granot, Z.; Henke, E.; Comen, E.A.; King, T.A.; Norton, L.; Benezra, R. Tumor Entrained Neutrophils Inhibit Seeding in the Premetastatic Lung. Cancer Cell 2011, 20, 300–314. [Google Scholar] [CrossRef]
  31. Galdiero, M.R.; Varricchi, G.; Loffredo, S.; Mantovani, A.; Marone, G. Roles of Neutrophils in Cancer Growth and Progression. J. Leukoc. Biol. 2018, 103, 457–464. [Google Scholar] [CrossRef] [PubMed]
  32. Shaul, M.E.; Fridlender, Z.G. Neutrophils as Active Regulators of the Immune System in the Tumor Microenvironment. J. Leukoc. Biol. 2017, 102, 343–349. [Google Scholar] [CrossRef] [PubMed]
  33. Donskov, F. Immunomonitoring and Prognostic Relevance of Neutrophils in Clinical Trials. Semin. Cancer Biol. 2013, 23, 200–207. [Google Scholar] [CrossRef] [PubMed]
  34. Galdiero, M.R.; Bianchi, P.; Grizzi, F.; Di Caro, G.; Basso, G.; Ponzetta, A.; Bonavita, E.; Barbagallo, M.; Tartari, S.; Polentarutti, N.; et al. Occurrence and Significance of Tumor-associated Neutrophils in Patients with Colorectal Cancer. Int. J. Cancer 2016, 139, 446–456. [Google Scholar] [CrossRef]
  35. Jensen, H.K.; Donskov, F.; Marcussen, N.; Nordsmark, M.; Lundbeck, F.; Von Der Maase, H. Presence of Intratumoral Neutrophils Is an Independent Prognostic Factor in Localized Renal Cell Carcinoma. JCO 2009, 27, 4709–4717. [Google Scholar] [CrossRef]
  36. Kuang, D.-M.; Zhao, Q.; Wu, Y.; Peng, C.; Wang, J.; Xu, Z.; Yin, X.-Y.; Zheng, L. Peritumoral Neutrophils Link Inflammatory Response to Disease Progression by Fostering Angiogenesis in Hepatocellular Carcinoma. J. Hepatol. 2011, 54, 948–955. [Google Scholar] [CrossRef]
  37. Trellakis, S.; Bruderek, K.; Dumitru, C.A.; Gholaman, H.; Gu, X.; Bankfalvi, A.; Scherag, A.; Hütte, J.; Dominas, N.; Lehnerdt, G.F.; et al. Polymorphonuclear Granulocytes in Human Head and Neck Cancer: Enhanced Inflammatory Activity, Modulation by Cancer Cells and Expansion in Advanced Disease. Int. J. Cancer 2011, 129, 2183–2193. [Google Scholar] [CrossRef]
  38. Wislez, M.; Rabbe, N.; Marchal, J.; Milleron, B.; Crestani, B.; Mayaud, C.; Antoine, M.; Soler, P.; Cadranel, J. Hepatocyte Growth Factor Production by Neutrophils Infiltrating Bronchioloalveolar Subtype Pulmonary Adenocarcinoma: Role in Tumor Progression and Death. Cancer Res. 2003, 63, 1405–1412. [Google Scholar] [PubMed]
  39. Mollinedo, F.; Borregaard, N.; Boxer, L.A. Novel Trends in Neutrophil Structure, Function and Development. Immunol. Today 1999, 20, 535–537. [Google Scholar] [CrossRef]
  40. Borregaard, N. Neutrophils, from Marrow to Microbes. Immunity 2010, 33, 657–670. [Google Scholar] [CrossRef]
  41. Bjerregaard, M.D.; Jurlander, J.; Klausen, P.; Borregaard, N.; Cowland, J.B. The in Vivo Profile of Transcription Factors during Neutrophil Differentiation in Human Bone Marrow. Blood 2003, 101, 4322–4332. [Google Scholar] [CrossRef] [PubMed]
  42. Görgens, A.; Radtke, S.; Möllmann, M.; Cross, M.; Dürig, J.; Horn, P.A.; Giebel, B. Revision of the Human Hematopoietic Tree: Granulocyte Subtypes Derive from Distinct Hematopoietic Lineages. Cell Rep. 2013, 3, 1539–1552. [Google Scholar] [CrossRef] [PubMed]
  43. Friedman, A.D. Transcriptional Control of Granulocyte and Monocyte Development. Oncogene 2007, 26, 6816–6828. [Google Scholar] [CrossRef]
  44. Fiedler, K.; Brunner, C. The Role of Transcription Factors in the Guidance of Granulopoiesis. Am. J. Blood Res. 2012, 2, 57–65. [Google Scholar] [PubMed]
  45. Rosenbauer, F.; Tenen, D.G. Transcription Factors in Myeloid Development: Balancing Differentiation with Transformation. Nat. Rev. Immunol. 2007, 7, 105–117. [Google Scholar] [CrossRef]
  46. Pillay, J.; Tak, T.; Kamp, V.M.; Koenderman, L. Immune Suppression by Neutrophils and Granulocytic Myeloid-Derived Suppressor Cells: Similarities and Differences. Cell. Mol. Life Sci. 2013, 70, 3813–3827. [Google Scholar] [CrossRef]
  47. Manz, M.G.; Boettcher, S. Emergency Granulopoiesis. Nat. Rev. Immunol. 2014, 14, 302–314. [Google Scholar] [CrossRef]
  48. Häger, M.; Cowland, J.B.; Borregaard, N. Neutrophil Granules in Health and Disease. J. Intern. Med. 2010, 268, 25–34. [Google Scholar] [CrossRef] [PubMed]
  49. Borregaard, N.; Sørensen, O.E.; Theilgaard-Mönch, K. Neutrophil Granules: A Library of Innate Immunity Proteins. Trends Immunol. 2007, 28, 340–345. [Google Scholar] [CrossRef]
  50. Dancey, J.T.; Deubelbeiss, K.A.; Harker, L.A.; Finch, C.A. Neutrophil Kinetics in Man. J. Clin. Invest. 1976, 58, 705–715. [Google Scholar] [CrossRef]
  51. Saverymuttu, S.H.; Peters, A.M.; Keshavarzian, A.; Reavy, H.J.; Lavender, J.P. The Kinetics of111 Indium Distribution Following Injection of111 Indium Labelled Autologous Granulocytes in Man. Br. J. Haematol. 1985, 61, 675–685. [Google Scholar] [CrossRef] [PubMed]
  52. Basu, S.; Hodgson, G.; Katz, M.; Dunn, A.R. Evaluation of Role of G-CSF in the Production, Survival, and Release of Neutrophils from Bone Marrow into Circulation. Blood 2002, 100, 854–861. [Google Scholar] [CrossRef]
  53. Santos-Beneit, A.M.; Mollinedo, F. Expression of Genes Involved in Initiation, Regulation, and Execution of Apoptosis in Human Neutrophils and during Neutrophil Differentiation of HL-60 Cells. J. Leukoc. Biol. 2000, 67, 712–724. [Google Scholar] [CrossRef] [PubMed]
  54. Kolaczkowska, E.; Kubes, P. Neutrophil Recruitment and Function in Health and Inflammation. Nat. Rev. Immunol. 2013, 13, 159–175. [Google Scholar] [CrossRef]
  55. Craddock, C.G.; Perry, S.; Ventzke, L.E.; Lawrence, J.S.; Baker, M.H.; Paul, G. Evaluation of Marrow Granulocytic Reserves in Normal and Disease States. Blood 1960, 15, 840–855. [Google Scholar] [CrossRef]
  56. Donohue, D.M.; Reiff, R.H.; Hanson, M.L.; Betson, Y.; Finch, C.A. Quantitative Measurement of the Erythrocytic and Granulocytic Cells of the Marrow and Blood1. J. Clin. Invest. 1958, 37, 1571–1576. [Google Scholar] [CrossRef] [PubMed]
  57. Perry, S.; Weinstein, I.M.; Craddock, C.G.; Lawrence, J.S.; Paul, G.; Baker, M.H.; Green, J.; Cianko, G. The Combined Use of Typhoid Vaccine and P32 Labeling to Assess Myelopoiesis. Blood 1957, 12, 549–558. [Google Scholar] [CrossRef]
  58. Casanova-Acebes, M.; Pitaval, C.; Weiss, L.A.; Nombela-Arrieta, C.; Chèvre, R.; A-González, N.; Kunisaki, Y.; Zhang, D.; van Rooijen, N.; Silberstein, L.E.; et al. Rhythmic Modulation of the Hematopoietic Niche through Neutrophil Clearance. Cell 2013, 153, 1025–1035. [Google Scholar] [CrossRef]
  59. Martin, C.; Burdon, P.C.E.; Bridger, G.; Gutierrez-Ramos, J.-C.; Williams, T.J.; Rankin, S.M. Chemokines Acting via CXCR2 and CXCR4 Control the Release of Neutrophils from the Bone Marrow and Their Return Following Senescence. Immunity 2003, 19, 583–593. [Google Scholar] [CrossRef]
  60. Bowers, E.; Slaughter, A.; Frenette, P.S.; Kuick, R.; Pello, O.M.; Lucas, D. Granulocyte-Derived TNFα Promotes Vascular and Hematopoietic Regeneration in the Bone Marrow. Nat. Med. 2018, 24, 95–102. [Google Scholar] [CrossRef]
  61. Kawano, Y.; Fukui, C.; Shinohara, M.; Wakahashi, K.; Ishii, S.; Suzuki, T.; Sato, M.; Asada, N.; Kawano, H.; Minagawa, K.; et al. G-CSF-Induced Sympathetic Tone Provokes Fever and Primes Antimobilizing Functions of Neutrophils via PGE2. Blood 2017, 129, 587–597. [Google Scholar] [CrossRef] [PubMed]
  62. Chen, X.; Deng, H.; Churchill, M.J.; Luchsinger, L.L.; Du, X.; Chu, T.H.; Friedman, R.A.; Middelhoff, M.; Ding, H.; Tailor, Y.H.; et al. Bone Marrow Myeloid Cells Regulate Myeloid-Biased Hematopoietic Stem Cells via a Histamine-Dependent Feedback Loop. Cell Stem Cell 2017, 21, 747–760.e7. [Google Scholar] [CrossRef]
  63. Ma, Q.; Jones, D.; Springer, T.A. The Chemokine Receptor CXCR4 Is Required for the Retention of B Lineage and Granulocytic Precursors within the Bone Marrow Microenvironment. Immunity 1999, 10, 463–471. [Google Scholar] [CrossRef] [PubMed]
  64. Suratt, B.T.; Petty, J.M.; Young, S.K.; Malcolm, K.C.; Lieber, J.G.; Nick, J.A.; Gonzalo, J.-A.; Henson, P.M.; Worthen, G.S. Role of the CXCR4/SDF-1 Chemokine Axis in Circulating Neutrophil Homeostasis. Blood 2004, 104, 565–571. [Google Scholar] [CrossRef]
  65. Eash, K.J.; Means, J.M.; White, D.W.; Link, D.C. CXCR4 Is a Key Regulator of Neutrophil Release from the Bone Marrow under Basal and Stress Granulopoiesis Conditions. Blood 2009, 113, 4711–4719. [Google Scholar] [CrossRef]
  66. Eash, K.J.; Greenbaum, A.M.; Gopalan, P.K.; Link, D.C. CXCR2 and CXCR4 Antagonistically Regulate Neutrophil Trafficking from Murine Bone Marrow. J. Clin. Invest. 2010, 120, 2423–2431. [Google Scholar] [CrossRef]
  67. Köhler, A.; De Filippo, K.; Hasenberg, M.; Van Den Brandt, C.; Nye, E.; Hosking, M.P.; Lane, T.E.; Männ, L.; Ransohoff, R.M.; Hauser, A.E.; et al. G-CSF–Mediated Thrombopoietin Release Triggers Neutrophil Motility and Mobilization from Bone Marrow via Induction of Cxcr2 Ligands. Blood 2011, 117, 4349–4357. [Google Scholar] [CrossRef]
  68. Levesque, J.-P.; Liu, F.; Simmons, P.J.; Betsuyaku, T.; Senior, R.M.; Pham, C.; Link, D.C. Characterization of Hematopoietic Progenitor Mobilization in Protease-Deficient Mice. Blood 2004, 104, 65–72. [Google Scholar] [CrossRef] [PubMed]
  69. Burdon, P.C.E.; Martin, C.; Rankin, S.M. The CXC Chemokine MIP-2 Stimulates Neutrophil Mobilization from the Rat Bone Marrow in a CD49d-Dependent Manner. Blood 2005, 105, 2543–2548. [Google Scholar] [CrossRef]
  70. Petty, J.M.; Lenox, C.C.; Weiss, D.J.; Poynter, M.E.; Suratt, B.T. Crosstalk between CXCR4/Stromal Derived Factor-1 and VLA-4/VCAM-1 Pathways Regulates Neutrophil Retention in the Bone Marrow. J. Immunol. 2009, 182, 604–612. [Google Scholar] [CrossRef]
  71. Kim, H.K.; De La Luz Sierra, M.; Williams, C.K.; Gulino, A.V.; Tosato, G. G-CSF down-Regulation of CXCR4 Expression Identified as a Mechanism for Mobilization of Myeloid Cells. Blood 2006, 108, 812–820. [Google Scholar] [CrossRef] [PubMed]
  72. Petit, I.; Szyper-Kravitz, M.; Nagler, A.; Lahav, M.; Peled, A.; Habler, L.; Ponomaryov, T.; Taichman, R.S.; Arenzana-Seisdedos, F.; Fujii, N.; et al. G-CSF Induces Stem Cell Mobilization by Decreasing Bone Marrow SDF-1 and up-Regulating CXCR4. Nat. Immunol. 2002, 3, 687–694. [Google Scholar] [CrossRef] [PubMed]
  73. Semerad, C.L.; Christopher, M.J.; Liu, F.; Short, B.; Simmons, P.J.; Winkler, I.; Levesque, J.-P.; Chappel, J.; Ross, F.P.; Link, D.C. G-CSF Potently Inhibits Osteoblast Activity and CXCL12 mRNA Expression in the Bone Marrow. Blood 2005, 106, 3020–3027. [Google Scholar] [CrossRef]
  74. Casanova-Acebes, M.; Nicolás-Ávila, J.A.; Li, J.L.; García-Silva, S.; Balachander, A.; Rubio-Ponce, A.; Weiss, L.A.; Adrover, J.M.; Burrows, K.; A-González, N.; et al. Neutrophils Instruct Homeostatic and Pathological States in Naive Tissues. J. Exp. Med. 2018, 215, 2778–2795. [Google Scholar] [CrossRef] [PubMed]
  75. Scheiermann, C.; Kunisaki, Y.; Lucas, D.; Chow, A.; Jang, J.-E.; Zhang, D.; Hashimoto, D.; Merad, M.; Frenette, P.S. Adrenergic Nerves Govern Circadian Leukocyte Recruitment to Tissues. Immunity 2012, 37, 290–301. [Google Scholar] [CrossRef]
  76. Adrover, J.M.; Nicolás-Ávila, J.A.; Hidalgo, A. Aging: A Temporal Dimension for Neutrophils. Trends Immunol. 2016, 37, 334–345. [Google Scholar] [CrossRef]
  77. Semerad, C.L.; Liu, F.; Gregory, A.D.; Stumpf, K.; Link, D.C. G-CSF Is an Essential Regulator of Neutrophil Trafficking from the Bone Marrow to the Blood. Immunity 2002, 17, 413–423. [Google Scholar] [CrossRef]
  78. Cua, D.J.; Tato, C.M. Innate IL-17-Producing Cells: The Sentinels of the Immune System. Nat. Rev. Immunol. 2010, 10, 479–489. [Google Scholar] [CrossRef]
  79. Gaffen, S.L.; Jain, R.; Garg, A.V.; Cua, D.J. The IL-23–IL-17 Immune Axis: From Mechanisms to Therapeutic Testing. Nat. Rev. Immunol. 2014, 14, 585–600. [Google Scholar] [CrossRef]
  80. Schwarzenberger, P.; Huang, W.; Ye, P.; Oliver, P.; Manuel, M.; Zhang, Z.; Bagby, G.; Nelson, S.; Kolls, J.K. Requirement of Endogenous Stem Cell Factor and Granulocyte-Colony-Stimulating Factor for IL-17-Mediated Granulopoiesis. J. Immunol. 2000, 164, 4783–4789. [Google Scholar] [CrossRef]
  81. Forlow, S.B.; Schurr, J.R.; Kolls, J.K.; Bagby, G.J.; Schwarzenberger, P.O.; Ley, K. Increased Granulopoiesis through Interleukin-17 and Granulocyte Colony-Stimulating Factor in Leukocyte Adhesion Molecule–Deficient Mice. Blood 2001, 98, 3309–3314. [Google Scholar] [CrossRef] [PubMed]
  82. Stark, M.A.; Huo, Y.; Burcin, T.L.; Morris, M.A.; Olson, T.S.; Ley, K. Phagocytosis of Apoptotic Neutrophils Regulates Granulopoiesis via IL-23 and IL-17. Immunity 2005, 22, 285–294. [Google Scholar] [CrossRef] [PubMed]
  83. Gordy, C.; Pua, H.; Sempowski, G.D.; He, Y.-W. Regulation of Steady-State Neutrophil Homeostasis by Macrophages. Blood 2011, 117, 618–629. [Google Scholar] [CrossRef] [PubMed]
  84. Tittel, A.P.; Heuser, C.; Ohliger, C.; Llanto, C.; Yona, S.; Hämmerling, G.J.; Engel, D.R.; Garbi, N.; Kurts, C. Functionally Relevant Neutrophilia in CD11c Diphtheria Toxin Receptor Transgenic Mice. Nat. Methods 2012, 9, 385–390. [Google Scholar] [CrossRef]
  85. Jiao, J.; Dragomir, A.-C.; Kocabayoglu, P.; Rahman, A.H.; Chow, A.; Hashimoto, D.; Leboeuf, M.; Kraus, T.; Moran, T.; Carrasco-Avino, G.; et al. Central Role of Conventional Dendritic Cells in Regulation of Bone Marrow Release and Survival of Neutrophils. J. Immunol. 2014, 192, 3374–3382. [Google Scholar] [CrossRef]
  86. Mei, J.; Liu, Y.; Dai, N.; Hoffmann, C.; Hudock, K.M.; Zhang, P.; Guttentag, S.H.; Kolls, J.K.; Oliver, P.M.; Bushman, F.D.; et al. Cxcr2 and Cxcl5 Regulate the IL-17/G-CSF Axis and Neutrophil Homeostasis in Mice. J. Clin. Invest. 2012, 122, 974–986. [Google Scholar] [CrossRef]
  87. Deshmukh, H.S.; Liu, Y.; Menkiti, O.R.; Mei, J.; Dai, N.; O’Leary, C.E.; Oliver, P.M.; Kolls, J.K.; Weiser, J.N.; Worthen, G.S. The Microbiota Regulates Neutrophil Homeostasis and Host Resistance to Escherichia Coli K1 Sepsis in Neonatal Mice. Nat. Med. 2014, 20, 524–530. [Google Scholar] [CrossRef]
  88. Ueda, Y.; Cain, D.W.; Kuraoka, M.; Kondo, M.; Kelsoe, G. IL-1R Type I-Dependent Hemopoietic Stem Cell Proliferation Is Necessary for Inflammatory Granulopoiesis and Reactive Neutrophilia. J. Immunol. 2009, 182, 6477–6484. [Google Scholar] [CrossRef]
  89. Mankan, A.K.; Canli, O.; Schwitalla, S.; Ziegler, P.; Tschopp, J.; Korn, T.; Greten, F.R. TNF-α–Dependent Loss of IKKβ-Deficient Myeloid Progenitors Triggers a Cytokine Loop Culminating in Granulocytosis. Proc. Natl. Acad. Sci. USA 2011, 108, 6567–6572. [Google Scholar] [CrossRef]
  90. Korniluk, A.; Koper, O.; Kemona, H.; Dymicka-Piekarska, V. From Inflammation to Cancer. Ir. J. Med. Sci. 2017, 186, 57–62. [Google Scholar] [CrossRef]
  91. Balkwill, F.; Mantovani, A. Inflammation and Cancer: Back to Virchow? Lancet 2001, 357, 539–545. [Google Scholar] [CrossRef] [PubMed]
  92. Mantovani, A. The Inflammation—Cancer Connection. FEBS J. 2018, 285, 638–640. [Google Scholar] [CrossRef] [PubMed]
  93. Lasry, A.; Zinger, A.; Ben-Neriah, Y. Inflammatory Networks Underlying Colorectal Cancer. Nat. Immunol. 2016, 17, 230–240. [Google Scholar] [CrossRef] [PubMed]
  94. Elinav, E.; Nowarski, R.; Thaiss, C.A.; Hu, B.; Jin, C.; Flavell, R.A. Inflammation-Induced Cancer: Crosstalk between Tumours, Immune Cells and Microorganisms. Nat. Rev. Cancer 2013, 13, 759–771. [Google Scholar] [CrossRef]
  95. Axelrad, J.E.; Lichtiger, S.; Yajnik, V. Inflammatory Bowel Disease and Cancer: The Role of Inflammation, Immunosuppression, and Cancer Treatment. WJG 2016, 22, 4794. [Google Scholar] [CrossRef]
  96. Tu, T.; Bühler, S.; Bartenschlager, R. Chronic Viral Hepatitis and Its Association with Liver Cancer. Biol. Chem. 2017, 398, 817–837. [Google Scholar] [CrossRef]
  97. Naumann, M.; Sokolova, O.; Tegtmeyer, N.; Backert, S. Helicobacter Pylori: A Paradigm Pathogen for Subverting Host Cell Signal Transmission. Trends Microbiol. 2017, 25, 316–328. [Google Scholar] [CrossRef]
  98. Pastille, E.; Frede, A.; McSorley, H.J.; Gräb, J.; Adamczyk, A.; Kollenda, S.; Hansen, W.; Epple, M.; Buer, J.; Maizels, R.M.; et al. Intestinal Helminth Infection Drives Carcinogenesis in Colitis-Associated Colon Cancer. PLoS Pathog. 2017, 13, e1006649. [Google Scholar] [CrossRef]
  99. Casper, C.; Fitzmaurice, C. Infection-Related Cancers: Prioritising an Important and Eliminable Contributor to the Global Cancer Burden. Lancet Glob. Health 2016, 4, e580–e581. [Google Scholar] [CrossRef]
  100. Butin-Israeli, V.; Bui, T.M.; Wiesolek, H.L.; Mascarenhas, L.; Lee, J.J.; Mehl, L.C.; Knutson, K.R.; Adam, S.A.; Goldman, R.D.; Beyder, A.; et al. Neutrophil-Induced Genomic Instability Impedes Resolution of Inflammation and Wound Healing. J. Clin. Investig. 2019, 129, 712–726. [Google Scholar] [CrossRef]
  101. Bui, T.M.; Butin-Israeli, V.; Wiesolek, H.L.; Zhou, M.; Rehring, J.F.; Wiesmüller, L.; Wu, J.D.; Yang, G.-Y.; Hanauer, S.B.; Sebag, J.A.; et al. Neutrophils Alter DNA Repair Landscape to Impact Survival and Shape Distinct Therapeutic Phenotypes of Colorectal Cancer. Gastroenterology 2021, 161, 225–238.e15. [Google Scholar] [CrossRef]
  102. Roessner, A.; Kuester, D.; Malfertheiner, P.; Schneider-Stock, R. Oxidative Stress in Ulcerative Colitis-Associated Carcinogenesis. Pathol.-Res. Pract. 2008, 204, 511–524. [Google Scholar] [CrossRef]
  103. Wu, S.; Chen, Y.; Chen, Z.; Wei, F.; Zhou, Q.; Li, P.; Gu, Q. Reactive Oxygen Species and Gastric Carcinogenesis: The Complex Interaction between Helicobacter Pylori and Host. Helicobacter 2023, 28, e13024. [Google Scholar] [CrossRef]
  104. Poto, R.; Cristinziano, L.; Modestino, L.; De Paulis, A.; Marone, G.; Loffredo, S.; Galdiero, M.R.; Varricchi, G. Neutrophil Extracellular Traps, Angiogenesis and Cancer. Biomedicines 2022, 10, 431. [Google Scholar] [CrossRef]
  105. Sionov, R.V.; Fridlender, Z.G.; Granot, Z. The Multifaceted Roles Neutrophils Play in the Tumor Microenvironment. Cancer Microenviron. 2015, 8, 125–158. [Google Scholar] [CrossRef]
  106. Raftopoulou, S.; Valadez-Cosmes, P.; Mihalic, Z.N.; Schicho, R.; Kargl, J. Tumor-Mediated Neutrophil Polarization and Therapeutic Implications. IJMS 2022, 23, 3218. [Google Scholar] [CrossRef]
  107. Gordon-Weeks, A.N.; Lim, S.Y.; Yuzhalin, A.E.; Jones, K.; Markelc, B.; Kim, K.J.; Buzzelli, J.N.; Fokas, E.; Cao, Y.; Smart, S.; et al. Neutrophils Promote Hepatic Metastasis Growth through Fibroblast Growth Factor 2–Dependent Angiogenesis in Mice. Hepatology 2017, 65, 1920–1935. [Google Scholar] [CrossRef]
  108. Deryugina, E.I.; Zajac, E.; Juncker-Jensen, A.; Kupriyanova, T.A.; Welter, L.; Quigley, J.P. Tissue-Infiltrating Neutrophils Constitute the Major In Vivo Source of Angiogenesis-Inducing MMP-9 in the Tumor Microenvironment. Neoplasia 2014, 16, 771–788. [Google Scholar] [CrossRef]
  109. Tecchio, C.; Cassatella, M.A. Neutrophil-Derived Cytokines Involved in Physiological and Pathological Angiogenesis. In Chemical Immunology and Allergy; Marone, G., Granata, F., Eds.; Karger Publishers: Basel, Switzerland, 2014; Volume 99, pp. 123–137. ISBN 978-3-318-02480-7. [Google Scholar]
  110. Loffredo, S.; Borriello, F.; Iannone, R.; Ferrara, A.L.; Galdiero, M.R.; Gigantino, V.; Esposito, P.; Varricchi, G.; Lambeau, G.; Cassatella, M.A.; et al. Group V Secreted Phospholipase A2 Induces the Release of Proangiogenic and Antiangiogenic Factors by Human Neutrophils. Front. Immunol. 2017, 8, 443. [Google Scholar] [CrossRef]
  111. Jablonska, J.; Leschner, S.; Westphal, K.; Lienenklaus, S.; Weiss, S. Neutrophils Responsive to Endogenous IFN-β Regulate Tumor Angiogenesis and Growth in a Mouse Tumor Model. J. Clin. Invest. 2010, 120, 1151–1164. [Google Scholar] [CrossRef]
  112. Webb, N.J.A.; Myers, C.R.; Watson, C.J.; Bottomley, M.J.; Brenchley, P.E.C. Activated human neutrophils express vascular endothelial growth factor (VEGF). Cytokine 1998, 10, 254–257. [Google Scholar] [CrossRef]
  113. Grenier, A.; Chollet-Martin, S.; Crestani, B.; Delarche, C.; El Benna, J.; Boutten, A.; Andrieu, V.; Durand, G.; Gougerot-Pocidalo, M.-A.; Aubier, M.; et al. Presence of a Mobilizable Intracellular Pool of Hepatocyte Growth Factor in Human Polymorphonuclear Neutrophils. Blood 2002, 99, 2997–3004. [Google Scholar] [CrossRef]
  114. Scapini, P.; Morini, M.; Tecchio, C.; Minghelli, S.; Di Carlo, E.; Tanghetti, E.; Albini, A.; Lowell, C.; Berton, G.; Noonan, D.M.; et al. CXCL1/Macrophage Inflammatory Protein-2-Induced Angiogenesis In Vivo Is Mediated by Neutrophil-Derived Vascular Endothelial Growth Factor-A. J. Immunol. 2004, 172, 5034–5040. [Google Scholar] [CrossRef]
  115. Gaudry, M.; Brégerie, O.; Andrieu, V.; El Benna, J.; Pocidalo, M.-A.; Hakim, J. Intracellular Pool of Vascular Endothelial Growth Factor in Human Neutrophils. Blood 1997, 90, 4153–4161. [Google Scholar] [CrossRef]
  116. Neagoe, P.-E.; Brkovic, A.; Hajjar, F.; Sirois, M.G. Expression and Release of Angiopoietin-1 from Human Neutrophils: Intracellular Mechanisms. Growth Factors 2009, 27, 335–344. [Google Scholar] [CrossRef]
  117. Ardi, V.C.; Kupriyanova, T.A.; Deryugina, E.I.; Quigley, J.P. Human Neutrophils Uniquely Release TIMP-Free MMP-9 to Provide a Potent Catalytic Stimulator of Angiogenesis. Proc. Natl. Acad. Sci. USA 2007, 104, 20262–20267. [Google Scholar] [CrossRef]
  118. Coussens, L.M.; Tinkle, C.L.; Hanahan, D.; Werb, Z. MMP-9 Supplied by Bone Marrow–Derived Cells Contributes to Skin Carcinogenesis. Cell 2000, 103, 481–490. [Google Scholar] [CrossRef]
  119. Tao, L.; Zhang, L.; Peng, Y.; Tao, M.; Li, L.; Xiu, D.; Yuan, C.; Ma, Z.; Jiang, B. Neutrophils Assist the Metastasis of Circulating Tumor Cells in Pancreatic Ductal Adenocarcinoma: A New Hypothesis and a New Predictor for Distant Metastasis. Medicine 2016, 95, e4932. [Google Scholar] [CrossRef]
  120. Rayes, R.F.; Mouhanna, J.G.; Nicolau, I.; Bourdeau, F.; Giannias, B.; Rousseau, S.; Quail, D.; Walsh, L.; Sangwan, V.; Bertos, N.; et al. Primary Tumors Induce Neutrophil Extracellular Traps with Targetable Metastasis-Promoting Effects. JCI Insight 2019, 4, e128008. [Google Scholar] [CrossRef]
  121. Munir, H.; Jones, J.O.; Janowitz, T.; Hoffmann, M.; Euler, M.; Martins, C.P.; Welsh, S.J.; Shields, J.D. Stromal-Driven and Amyloid β-Dependent Induction of Neutrophil Extracellular Traps Modulates Tumor Growth. Nat. Commun. 2021, 12, 683. [Google Scholar] [CrossRef]
  122. Yang, L.; Liu, Q.; Zhang, X.; Liu, X.; Zhou, B.; Chen, J.; Huang, D.; Li, J.; Li, H.; Chen, F.; et al. DNA of Neutrophil Extracellular Traps Promotes Cancer Metastasis via CCDC25. Nature 2020, 583, 133–138. [Google Scholar] [CrossRef] [PubMed]
  123. Park, J.; Wysocki, R.W.; Amoozgar, Z.; Maiorino, L.; Fein, M.R.; Jorns, J.; Schott, A.F.; Kinugasa-Katayama, Y.; Lee, Y.; Won, N.H.; et al. Cancer Cells Induce Metastasis-Supporting Neutrophil Extracellular DNA Traps. Sci. Transl. Med. 2016, 8, 361ra138. [Google Scholar] [CrossRef] [PubMed]
  124. Lee, W.; Ko, S.Y.; Mohamed, M.S.; Kenny, H.A.; Lengyel, E.; Naora, H. Neutrophils Facilitate Ovarian Cancer Premetastatic Niche Formation in the Omentum. J. Exp. Med. 2019, 216, 176–194. [Google Scholar] [CrossRef]
  125. El Rayes, T.; Catena, R.; Lee, S.; Stawowczyk, M.; Joshi, N.; Fischbach, C.; Powell, C.A.; Dannenberg, A.J.; Altorki, N.K.; Gao, D.; et al. Lung Inflammation Promotes Metastasis through Neutrophil Protease-Mediated Degradation of Tsp-1. Proc. Natl. Acad. Sci. USA 2015, 112, 16000–16005. [Google Scholar] [CrossRef] [PubMed]
  126. Albrengues, J.; Shields, M.A.; Ng, D.; Park, C.G.; Ambrico, A.; Poindexter, M.E.; Upadhyay, P.; Uyeminami, D.L.; Pommier, A.; Küttner, V.; et al. Neutrophil Extracellular Traps Produced during Inflammation Awaken Dormant Cancer Cells in Mice. Science 2018, 361, eaao4227. [Google Scholar] [CrossRef]
  127. Germann, M.; Zangger, N.; Sauvain, M.; Sempoux, C.; Bowler, A.D.; Wirapati, P.; Kandalaft, L.E.; Delorenzi, M.; Tejpar, S.; Coukos, G.; et al. Neutrophils Suppress Tumor-infiltrating T Cells in Colon Cancer via Matrix Metalloproteinase-mediated Activation of TGF β. EMBO Mol. Med. 2020, 12, e10681. [Google Scholar] [CrossRef]
  128. Yang, M.; Zhang, G.; Wang, Y.; He, M.; Xu, Q.; Lu, J.; Liu, H.; Xu, C. Tumour-Associated Neutrophils Orchestrate Intratumoural IL-8-Driven Immune Evasion through Jagged2 Activation in Ovarian Cancer. Br. J. Cancer 2020, 123, 1404–1416. [Google Scholar] [CrossRef]
  129. Domnich, M.; Pylaeva, E.; Siakaeva, E.; Kabankova, N.; Bedzinska, A.; Sojka, D.; Zebrowska, A.; Gawin, M.; Soldierer, M.; Rist, M.; et al. Serpins A1/A3 within Tumor-Derived Extracellular Vesicles Support pro-Tumoral Bias of Neutrophils in Cancer. bioRxiv 2025. [Google Scholar] [CrossRef]
  130. Coffelt, S.B.; Kersten, K.; Doornebal, C.W.; Weiden, J.; Vrijland, K.; Hau, C.-S.; Verstegen, N.J.M.; Ciampricotti, M.; Hawinkels, L.J.A.C.; Jonkers, J.; et al. IL-17-Producing Γδ T Cells and Neutrophils Conspire to Promote Breast Cancer Metastasis. Nature 2015, 522, 345–348. [Google Scholar] [CrossRef]
  131. Apte, R.N.; Segal, S.; Dinarello, C.A.; Song, X.; Krelin, Y.; Dvorkin, T. Bearing Tumors of IL-1β-Secreting Cells Mediate Suppression of T Cells in Mice CD11b+/Gr-1+ Immature Myeloid Cells. J. Immunol. 2005, 175, 8200–8208. [Google Scholar] [CrossRef]
  132. Bunt, S.K.; Sinha, P.; Clements, V.K.; Leips, J.; Ostrand-Rosenberg, S. Inflammation Induces Myeloid-Derived Suppressor Cells That Facilitate Tumor Progression. J. Immunol. 2006, 176, 284–290. [Google Scholar] [CrossRef] [PubMed]
  133. Elkabets, M.; Ribeiro, V.S.G.; Dinarello, C.A.; Ostrand-Rosenberg, S.; Di Santo, J.P.; Apte, R.N.; Vosshenrich, C.A.J. IL-1β Regulates a Novel Myeloid-derived Suppressor Cell Subset That Impairs NK Cell Development and Function. Eur. J. Immunol. 2010, 40, 3347–3357. [Google Scholar] [CrossRef] [PubMed]
  134. Tu, S.; Bhagat, G.; Cui, G.; Takaishi, S.; Kurt-Jones, E.A.; Rickman, B.; Betz, K.S.; Penz-Oesterreicher, M.; Bjorkdahl, O.; Fox, J.G.; et al. Overexpression of Interleukin-1β Induces Gastric Inflammation and Cancer and Mobilizes Myeloid-Derived Suppressor Cells in Mice. Cancer Cell 2008, 14, 408–419. [Google Scholar] [CrossRef]
  135. Gershkovitz, M.; Caspi, Y.; Fainsod-Levi, T.; Katz, B.; Michaeli, J.; Khawaled, S.; Lev, S.; Polyansky, L.; Shaul, M.E.; Sionov, R.V.; et al. TRPM2 Mediates Neutrophil Killing of Disseminated Tumor Cells. Cancer Res. 2018, 78, 2680–2690. [Google Scholar] [CrossRef]
  136. Buckley, C.D.; Ross, E.A.; McGettrick, H.M.; Osborne, C.E.; Haworth, O.; Schmutz, C.; Stone, P.C.W.; Salmon, M.; Matharu, N.M.; Vohra, R.K.; et al. Identification of a Phenotypically and Functionally Distinct Population of Long-Lived Neutrophils in a Model of Reverse Endothelial Migration. J. Leukoc. Biol. 2005, 79, 303–311. [Google Scholar] [CrossRef] [PubMed]
  137. Finisguerra, V.; Di Conza, G.; Di Matteo, M.; Serneels, J.; Costa, S.; Thompson, A.A.R.; Wauters, E.; Walmsley, S.; Prenen, H.; Granot, Z.; et al. MET Is Required for the Recruitment of Anti-Tumoural Neutrophils. Nature 2015, 522, 349–353. [Google Scholar] [CrossRef]
  138. Mensurado, S.; Rei, M.; Lança, T.; Ioannou, M.; Gonçalves-Sousa, N.; Kubo, H.; Malissen, M.; Papayannopoulos, V.; Serre, K.; Silva-Santos, B. Tumor-Associated Neutrophils Suppress pro-Tumoral IL-17+ Γδ T Cells through Induction of Oxidative Stress. PLoS Biol. 2018, 16, e2004990. [Google Scholar] [CrossRef]
  139. Cui, C.; Chakraborty, K.; Tang, X.A.; Zhou, G.; Schoenfelt, K.Q.; Becker, K.M.; Hoffman, A.; Chang, Y.-F.; Blank, A.; Reardon, C.A.; et al. Neutrophil Elastase Selectively Kills Cancer Cells and Attenuates Tumorigenesis. Cell 2021, 184, 3163–3177.e21. [Google Scholar] [CrossRef]
  140. Shrestha, S.; Noh, J.M.; Kim, S.-Y.; Ham, H.-Y.; Kim, Y.-J.; Yun, Y.-J.; Kim, M.-J.; Kwon, M.-S.; Song, D.-K.; Hong, C.-W. Angiotensin Converting Enzyme Inhibitors and Angiotensin II Receptor Antagonist Attenuate Tumor Growth via Polarization of Neutrophils toward an Antitumor Phenotype. OncoImmunology 2016, 5, e1067744. [Google Scholar] [CrossRef]
  141. Fridlender, Z.G.; Sun, J.; Kim, S.; Kapoor, V.; Cheng, G.; Ling, L.; Worthen, G.S.; Albelda, S.M. Polarization of Tumor-Associated Neutrophil Phenotype by TGF-β: “N1” versus “N2” TAN. Cancer Cell 2009, 16, 183–194. [Google Scholar] [CrossRef]
  142. Schernberg, A.; Blanchard, P.; Chargari, C.; Deutsch, E. Neutrophils, a Candidate Biomarker and Target for Radiation Therapy? Acta Oncol. 2017, 56, 1522–1530. [Google Scholar] [CrossRef] [PubMed]
  143. Bodogai, M.; Moritoh, K.; Lee-Chang, C.; Hollander, C.M.; Sherman-Baust, C.A.; Wersto, R.P.; Araki, Y.; Miyoshi, I.; Yang, L.; Trinchieri, G.; et al. Immunosuppressive and Prometastatic Functions of Myeloid-Derived Suppressive Cells Rely upon Education from Tumor-Associated B Cells. Cancer Res. 2015, 75, 3456–3465. [Google Scholar] [CrossRef] [PubMed]
  144. Pang, Y.; Gara, S.K.; Achyut, B.R.; Li, Z.; Yan, H.H.; Day, C.-P.; Weiss, J.M.; Trinchieri, G.; Morris, J.C.; Yang, L. TGF-β Signaling in Myeloid Cells Is Required for Tumor Metastasis. Cancer Discov. 2013, 3, 936–951. [Google Scholar] [CrossRef]
  145. Jackstadt, R.; Van Hooff, S.R.; Leach, J.D.; Cortes-Lavaud, X.; Lohuis, J.O.; Ridgway, R.A.; Wouters, V.M.; Roper, J.; Kendall, T.J.; Roxburgh, C.S.; et al. Epithelial NOTCH Signaling Rewires the Tumor Microenvironment of Colorectal Cancer to Drive Poor-Prognosis Subtypes and Metastasis. Cancer Cell 2019, 36, 319–336.e7. [Google Scholar] [CrossRef]
  146. Yoo, C.; Javle, M.M.; Verdaguer Mata, H.; De Braud, F.; Trojan, J.; Raoul, J.-L.; Kim, J.W.; Ueno, M.; Lee, C.; Hijioka, S.; et al. Phase 2 Trial of Bintrafusp Alfa as Second-Line Therapy for Patients with Locally Advanced/Metastatic Biliary Tract Cancers. Hepatology 2023, 78, 758–770. [Google Scholar] [CrossRef] [PubMed]
  147. Yamazaki, T.; Gunderson, A.J.; Gilchrist, M.; Whiteford, M.; Kiely, M.X.; Hayman, A.; O’Brien, D.; Ahmad, R.; Manchio, J.V.; Fox, N.; et al. Galunisertib plus Neoadjuvant Chemoradiotherapy in Patients with Locally Advanced Rectal Cancer: A Single-Arm, Phase 2 Trial. Lancet Oncol. 2022, 23, 1189–1200. [Google Scholar] [CrossRef]
  148. Takeshima, T.; Pop, L.M.; Laine, A.; Iyengar, P.; Vitetta, E.S.; Hannan, R. Key Role for Neutrophils in Radiation-Induced Antitumor Immune Responses: Potentiation with G-CSF. Proc. Natl. Acad. Sci. USA 2016, 113, 11300–11305. [Google Scholar] [CrossRef]
  149. 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. 2019, 38, 6. [Google Scholar] [CrossRef]
  150. Große-Steffen, T.; Giese, T.; Giese, N.; Longerich, T.; Schirmacher, P.; Hänsch, G.M.; Gaida, M.M. Epithelial-to-Mesenchymal Transition in Pancreatic Ductal Adenocarcinoma and Pancreatic Tumor Cell Lines: The Role of Neutrophils and Neutrophil-Derived Elastase. Clin. Dev. Immunol. 2012, 2012, 720768. [Google Scholar] [CrossRef]
  151. Mittal, V. Epithelial Mesenchymal Transition in Tumor Metastasis. Annu. Rev. Pathol. 2018, 13, 395–412. [Google Scholar] [CrossRef]
  152. Zhang, X.; Shi, H.; Yuan, X.; Jiang, P.; Qian, H.; Xu, W. Tumor-Derived Exosomes Induce N2 Polarization of Neutrophils to Promote Gastric Cancer Cell Migration. Mol. Cancer 2018, 17, 146. [Google Scholar] [CrossRef] [PubMed]
  153. Szczerba, B.M.; Castro-Giner, F.; Vetter, M.; Krol, I.; Gkountela, S.; Landin, J.; Scheidmann, M.C.; Donato, C.; Scherrer, R.; Singer, J.; et al. Neutrophils Escort Circulating Tumour Cells to Enable Cell Cycle Progression. Nature 2019, 566, 553–557. [Google Scholar] [CrossRef] [PubMed]
  154. Huh, S.J.; Liang, S.; Sharma, A.; Dong, C.; Robertson, G.P. Transiently Entrapped Circulating Tumor Cells Interact with Neutrophils to Facilitate Lung Metastasis Development. Cancer Res. 2010, 70, 6071–6082. [Google Scholar] [CrossRef]
  155. Spicer, J.D.; McDonald, B.; Cools-Lartigue, J.J.; Chow, S.C.; Giannias, B.; Kubes, P.; Ferri, L.E. Neutrophils Promote Liver Metastasis via Mac-1–Mediated Interactions with Circulating Tumor Cells. Cancer Res. 2012, 72, 3919–3927. [Google Scholar] [CrossRef]
  156. Li, P.; Lu, M.; Shi, J.; Gong, Z.; Hua, L.; Li, Q.; Lim, B.; Zhang, X.H.-F.; Chen, X.; Li, S.; et al. Lung Mesenchymal Cells Elicit Lipid Storage in Neutrophils That Fuel Breast Cancer Lung Metastasis. Nat. Immunol. 2020, 21, 1444–1455. [Google Scholar] [CrossRef]
  157. Langley, R.R.; Fidler, I.J. The Seed and Soil Hypothesis Revisited—The Role of Tumor-stroma Interactions in Metastasis to Different Organs. Int. J. Cancer 2011, 128, 2527–2535. [Google Scholar] [CrossRef]
  158. Kaplan, R.N.; Riba, R.D.; Zacharoulis, S.; Bramley, A.H.; Vincent, L.; Costa, C.; MacDonald, D.D.; Jin, D.K.; Shido, K.; Kerns, S.A.; et al. VEGFR1-Positive Haematopoietic Bone Marrow Progenitors Initiate the Pre-Metastatic Niche. Nature 2005, 438, 820–827. [Google Scholar] [CrossRef]
  159. Kaplan, R.N.; Psaila, B.; Lyden, D. Bone Marrow Cells in the ‘Pre-Metastatic Niche’: Within Bone and Beyond. Cancer Metastasis Rev. 2007, 25, 521–529. [Google Scholar] [CrossRef] [PubMed]
  160. Wculek, S.K.; Malanchi, I. Neutrophils Support Lung Colonization of Metastasis-Initiating Breast Cancer Cells. Nature 2015, 528, 413–417. [Google Scholar] [CrossRef]
  161. Tyagi, A.; Sharma, S.; Wu, K.; Wu, S.-Y.; Xing, F.; Liu, Y.; Zhao, D.; Deshpande, R.P.; D’Agostino, R.B.; Watabe, K. Nicotine Promotes Breast Cancer Metastasis by Stimulating N2 Neutrophils and Generating Pre-Metastatic Niche in Lung. Nat. Commun. 2021, 12, 474. [Google Scholar] [CrossRef]
  162. Peinado, H.; Alečković, M.; Lavotshkin, S.; Matei, I.; Costa-Silva, B.; Moreno-Bueno, G.; Hergueta-Redondo, M.; Williams, C.; García-Santos, G.; Ghajar, C.M.; et al. Melanoma Exosomes Educate Bone Marrow Progenitor Cells toward a Pro-Metastatic Phenotype through MET. Nat. Med. 2012, 18, 883–891. [Google Scholar] [CrossRef] [PubMed]
  163. Hoshino, A.; Costa-Silva, B.; Shen, T.-L.; Rodrigues, G.; Hashimoto, A.; Tesic Mark, M.; Molina, H.; Kohsaka, S.; Di Giannatale, A.; Ceder, S.; et al. Tumour Exosome Integrins Determine Organotropic Metastasis. Nature 2015, 527, 329–335. [Google Scholar] [CrossRef]
  164. Liu, Y.; Gu, Y.; Han, Y.; Zhang, Q.; Jiang, Z.; Zhang, X.; Huang, B.; Xu, X.; Zheng, J.; Cao, X. Tumor Exosomal RNAs Promote Lung Pre-Metastatic Niche Formation by Activating Alveolar Epithelial TLR3 to Recruit Neutrophils. Cancer Cell 2016, 30, 243–256. [Google Scholar] [CrossRef]
  165. Spiegel, A.; Brooks, M.W.; Houshyar, S.; Reinhardt, F.; Ardolino, M.; Fessler, E.; Chen, M.B.; Krall, J.A.; DeCock, J.; Zervantonakis, I.K.; et al. Neutrophils Suppress Intraluminal NK Cell–Mediated Tumor Cell Clearance and Enhance Extravasation of Disseminated Carcinoma Cells. Cancer Discov. 2016, 6, 630–649. [Google Scholar] [CrossRef] [PubMed]
  166. Yang, C.; Wang, Z.; Li, L.; Zhang, Z.; Jin, X.; Wu, P.; Sun, S.; Pan, J.; Su, K.; Jia, F.; et al. Aged Neutrophils Form Mitochondria-Dependent Vital NETs to Promote Breast Cancer Lung Metastasis. J. Immunother. Cancer 2021, 9, e002875. [Google Scholar] [CrossRef]
  167. Zheng, Z.; Li, Y.; Jia, S.; Zhu, M.; Cao, L.; Tao, M.; Jiang, J.; Zhan, S.; Chen, Y.; Gao, P.-J.; et al. Lung Mesenchymal Stromal Cells Influenced by Th2 Cytokines Mobilize Neutrophils and Facilitate Metastasis by Producing Complement C3. Nat. Commun. 2021, 12, 6202. [Google Scholar] [CrossRef] [PubMed]
  168. Cackowski, F.C.; Heath, E.I. Prostate Cancer Dormancy and Recurrence. Cancer Lett. 2022, 524, 103–108. [Google Scholar] [CrossRef]
  169. Park, S.-Y.; Nam, J.-S. The Force Awakens: Metastatic Dormant Cancer Cells. Exp. Mol. Med. 2020, 52, 569–581. [Google Scholar] [CrossRef]
  170. Francescangeli, F.; De Angelis, M.L.; Zeuner, A. COVID-19: A Potential Driver of Immune-Mediated Breast Cancer Recurrence? Breast Cancer Res. 2020, 22, 117. [Google Scholar] [CrossRef]
  171. Barnes, B.J.; Adrover, J.M.; Baxter-Stoltzfus, A.; Borczuk, A.; Cools-Lartigue, J.; Crawford, J.M.; Daßler-Plenker, J.; Guerci, P.; Huynh, C.; Knight, J.S.; et al. Targeting Potential Drivers of COVID-19: Neutrophil Extracellular Traps. J. Exp. Med. 2020, 217, e20200652. [Google Scholar] [CrossRef]
  172. Luo, J.; Feng, X.-X.; Luo, C.; Wang, Y.; Li, D.; Shu, Y.; Wang, S.-S.; Qin, J.; Li, Y.-C.; Zou, J.-M.; et al. 14,15-EET Induces the Infiltration and Tumor-Promoting Function of Neutrophils to Trigger the Growth of Minimal Dormant Metastases. Oncotarget 2016, 7, 43324–43336. [Google Scholar] [CrossRef] [PubMed]
  173. Perego, M.; Tyurin, V.A.; Tyurina, Y.Y.; Yellets, J.; Nacarelli, T.; Lin, C.; Nefedova, Y.; Kossenkov, A.; Liu, Q.; Sreedhar, S.; et al. Reactivation of Dormant Tumor Cells by Modified Lipids Derived from Stress-Activated Neutrophils. Sci. Transl. Med. 2020, 12, eabb5817. [Google Scholar] [CrossRef] [PubMed]
  174. Hirz, T.; Mei, S.; Sarkar, H.; Kfoury, Y.; Wu, S.; Verhoeven, B.M.; Subtelny, A.O.; Zlatev, D.V.; Wszolek, M.W.; Salari, K.; et al. Dissecting the Immune Suppressive Human Prostate Tumor Microenvironment via Integrated Single-Cell and Spatial Transcriptomic Analyses. Nat. Commun. 2023, 14, 663. [Google Scholar] [CrossRef] [PubMed]
  175. De Zuani, M.; Xue, H.; Park, J.S.; Dentro, S.C.; Seferbekova, Z.; Tessier, J.; Curras-Alonso, S.; Hadjipanayis, A.; Athanasiadis, E.I.; Gerstung, M.; et al. Single-Cell and Spatial Transcriptomics Analysis of Non-Small Cell Lung Cancer. Nat. Commun. 2024, 15, 4388. [Google Scholar] [CrossRef]
  176. 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 2023, 72, 958–971. [Google Scholar] [CrossRef]
  177. Hwang, W.L.; Jagadeesh, K.A.; Guo, J.A.; Hoffman, H.I.; Yadollahpour, P.; Reeves, J.W.; Mohan, R.; Drokhlyansky, E.; Van Wittenberghe, N.; Ashenberg, O.; et al. Single-Nucleus and Spatial Transcriptome Profiling of Pancreatic Cancer Identifies Multicellular Dynamics Associated with Neoadjuvant Treatment. Nat. Genet. 2022, 54, 1178–1191. [Google Scholar] [CrossRef]
  178. Surendran, V.; Rutledge, D.; Colmon, R.; Chandrasekaran, A. A Novel Tumor-Immune Microenvironment (TIME)-on-Chip Mimics Three Dimensional Neutrophil-Tumor Dynamics and Neutrophil Extracellular Traps (NETs)-Mediated Collective Tumor Invasion. Biofabrication 2021, 13, 035029. [Google Scholar] [CrossRef]
  179. Yeo, A.T.; Rawal, S.; Delcuze, B.; Christofides, A.; Atayde, A.; Strauss, L.; Balaj, L.; Rogers, V.A.; Uhlmann, E.J.; Varma, H.; et al. Single-Cell RNA Sequencing Reveals Evolution of Immune Landscape during Glioblastoma Progression. Nat. Immunol. 2022, 23, 971–984. [Google Scholar] [CrossRef]
  180. Zhou, C.; Guo, L.; Cai, Q.; Xi, W.; Yuan, F.; Zhang, H.; Yan, C.; Huang, L.; Zhu, Z.; Zhang, J. Circulating Neutrophils Activated by Cancer Cells and M2 Macrophages Promote Gastric Cancer Progression during PD-1 Antibody-Based Immunotherapy. Front. Mol. Biosci. 2023, 10, 1081762. [Google Scholar] [CrossRef]
  181. Wu, Y.; Ma, J.; Yang, X.; Nan, F.; Zhang, T.; Ji, S.; Rao, D.; Feng, H.; Gao, K.; Gu, X.; et al. Neutrophil Profiling Illuminates Anti-Tumor Antigen-Presenting Potency. Cell 2024, 187, 1422–1439.e24. [Google Scholar] [CrossRef]
  182. Nieto, P.; Elosua-Bayes, M.; Trincado, J.L.; Marchese, D.; Massoni-Badosa, R.; Salvany, M.; Henriques, A.; Nieto, J.; Aguilar-Fernández, S.; Mereu, E.; et al. A Single-Cell Tumor Immune Atlas for Precision Oncology. Genome Res. 2021, 31, 1913–1926. [Google Scholar] [CrossRef] [PubMed]
  183. Lv, Y.; Chen, C.; Han, M.; Tian, C.; Song, F.; Feng, S.; Xu, M.; Zhao, Z.; Zhou, H.; Su, W.; et al. CXCL2: A Key Player in the Tumor Microenvironment and Inflammatory Diseases. Cancer Cell Int. 2025, 25, 133. [Google Scholar] [CrossRef] [PubMed]
  184. Guo, C.; Sharp, A.; Gurel, B.; Crespo, M.; Figueiredo, I.; Jain, S.; Vogl, U.; Rekowski, J.; Rouhifard, M.; Gallagher, L.; et al. Targeting Myeloid Chemotaxis to Reverse Prostate Cancer Therapy Resistance. Nature 2023, 623, 1053–1061. [Google Scholar] [CrossRef]
  185. Yaeger, M.J.; Ngatikaura, T.; Zecchino, N.; Dunigan-Russell, K.; Lovins, H.B.; Schott, E.; Hutton, G.; Saunders, B.; Lin, Y.; Zhang, J. (Jim); et al. ALX/FPR2 Contributes to Serum Amyloid A-Induced Lung Neutrophil Recruitment Following Acute Ozone Exposure. FASEB J. 2025, 39, e70555. [Google Scholar] [CrossRef]
  186. Wang, J.; Man, K.; Ng, K.T.-P. Emerging Roles of C-C Motif Ligand 11 (CCL11) in Cancers and Liver Diseases: Mechanisms and Therapeutic Implications. IJMS 2025, 26, 4662. [Google Scholar] [CrossRef]
  187. Simoncello, F.; Piperno, G.M.; Caronni, N.; Amadio, R.; Cappelletto, A.; Canarutto, G.; Piazza, S.; Bicciato, S.; Benvenuti, F. CXCL5-Mediated Accumulation of Mature Neutrophils in Lung Cancer Tissues Impairs the Differentiation Program of Anticancer CD8 T Cells and Limits the Efficacy of Checkpoint Inhibitors. OncoImmunology 2022, 11, 2059876. [Google Scholar] [CrossRef]
  188. Chen, M.; Qi, Z.; Meng, X.; Wang, S.; Zheng, X.; Hu, M.; Liu, X.; Song, Y.; Deng, Y. Blockade of Neutrophil Recruitment to Tumor Sites Based on Sialic Acid-Modified Nanoplatforms Enhances the Efficacy of Checkpoint Blockade Immunotherapy. Asian J. Pharm. Sci. 2023, 18, 100784. [Google Scholar] [CrossRef]
  189. Kato, T.; Fukushima, H.; Furusawa, A.; Okada, R.; Wakiyama, H.; Furumoto, H.; Okuyama, S.; Takao, S.; Choyke, P.L.; Kobayashi, H. Selective Depletion of Polymorphonuclear Myeloid Derived Suppressor Cells in Tumor Beds with near Infrared Photoimmunotherapy Enhances Host Immune Response. OncoImmunology 2022, 11, 2152248. [Google Scholar] [CrossRef] [PubMed]
  190. Hu, T.; Zhai, J.; Yang, Z.; Peng, J.; Wang, C.; Liu, X.; Li, Y.; Yao, J.; Chen, F.; Li, H.; et al. Myeloid-Derived Suppressor Cells in Cancer: Mechanistic Insights and Targeted Therapeutic Innovations. MedComm 2025, 6, e70231. [Google Scholar] [CrossRef]
  191. Zhang, T.; Li, Y.; Zhai, E.; Zhao, R.; Qian, Y.; Huang, Z.; Liu, Y.; Zhao, Z.; Xu, X.; Liu, J.; et al. Intratumoral Fusobacterium Nucleatum Recruits Tumor-Associated Neutrophils to Promote Gastric Cancer Progression and Immune Evasion. Cancer Res. 2025, 85, 1819–1841. [Google Scholar] [CrossRef]
  192. Kasama, T.; Miwa, Y.; Isozaki, T.; Odai, T.; Adachi, M.; Kunkel, S. Neutrophil-Derived Cytokines: Potential Therapeutic Targets in Inflammation. CDTIA 2005, 4, 273–279. [Google Scholar] [CrossRef] [PubMed]
  193. Hein, L.E.; SenGupta, S.; Gunasekaran, G.; Johnson, C.N.; Parent, C.A. TGF-Β1 Activates Neutrophil Signaling and Gene Expression but Not Migration. PLoS ONE 2023, 18, e0290886. [Google Scholar] [CrossRef] [PubMed]
  194. Heemskerk, N.; Gruijs, M.; Temming, A.R.; Heineke, M.H.; Gout, D.Y.; Hellingman, T.; Tuk, C.W.; Winter, P.J.; Lissenberg-Thunnissen, S.; Bentlage, A.E.H.; et al. Augmented Antibody-Based Anticancer Therapeutics Boost Neutrophil Cytotoxicity. J. Clin. Investig. 2021, 131, e134680. [Google Scholar] [CrossRef] [PubMed]
  195. Yuan, S.; Hu, Q. Convergence of Nanomedicine and Neutrophils for Drug Delivery. Bioact. Mater. 2024, 35, 150–166. [Google Scholar] [CrossRef]
  196. Chu, D.; Dong, X.; Shi, X.; Zhang, C.; Wang, Z. Neutrophil-Based Drug Delivery Systems. Adv. Mater. 2018, 30, 1706245. [Google Scholar] [CrossRef]
  197. Xue, J.; Zhao, Z.; Zhang, L.; Xue, L.; Shen, S.; Wen, Y.; Wei, Z.; Wang, L.; Kong, L.; Sun, H.; et al. Neutrophil-Mediated Anticancer Drug Delivery for Suppression of Postoperative Malignant Glioma Recurrence. Nat. Nanotech 2017, 12, 692–700. [Google Scholar] [CrossRef]
  198. Gryka-Marton, M.; Grabowska, A.D.; Szukiewicz, D. Breaking the Barrier: The Role of Proinflammatory Cytokines in BBB Dysfunction. IJMS 2025, 26, 3532. [Google Scholar] [CrossRef]
  199. Shimizu, F.; Nakamori, M. Blood–Brain Barrier Disruption in Neuroimmunological Disease. IJMS 2024, 25, 10625. [Google Scholar] [CrossRef]
  200. Chu, Y.; Luo, Y.; Su, B.; Li, C.; Guo, Q.; Zhang, Y.; Liu, P.; Chen, H.; Zhao, Z.; Zhou, Z.; et al. A Neutrophil-Biomimic Platform for Eradicating Metastatic Breast Cancer Stem-like Cells by Redox Microenvironment Modulation and Hypoxia-Triggered Differentiation Therapy. Acta Pharm. Sin. B 2023, 13, 298–314. [Google Scholar] [CrossRef]
  201. Hersh, A.M.; Alomari, S.; Tyler, B.M. Crossing the Blood-Brain Barrier: Advances in Nanoparticle Technology for Drug Delivery in Neuro-Oncology. IJMS 2022, 23, 4153. [Google Scholar] [CrossRef]
  202. Wang, J.; Tang, W.; Yang, M.; Yin, Y.; Li, H.; Hu, F.; Tang, L.; Ma, X.; Zhang, Y.; Wang, Y. Inflammatory Tumor Microenvironment Responsive Neutrophil Exosomes-Based Drug Delivery System for Targeted Glioma Therapy. Biomaterials 2021, 273, 120784. [Google Scholar] [CrossRef] [PubMed]
  203. Guo, Y.; Li, Y.; Li, J.; Cai, H.; Liu, K.; Duan, D.; Zhang, W.; Han, G.; Zhao, Y. Controlled Inflammation Drives Neutrophil-Mediated Precision Drug Delivery in Heterogeneous Tumors. Adv. Sci. 2025, 12, 2411307. [Google Scholar] [CrossRef]
  204. Wang, P.-F.; Zhang, Y.-X.; Su, J.; Yao, K.; Li, S.-W.; Huang, G.-R.; Yan, C.-X. Neutrophil Depletion Enhances the Therapeutic Effect of PD-1 Antibody on Glioma. Aging 2020, 12, 15290–15301. [Google Scholar] [CrossRef]
  205. Ager, A. Cancer Immunotherapy: T Cells and Neutrophils Working Together to Attack Cancers. Cell 2023, 186, 1304–1306. [Google Scholar] [CrossRef] [PubMed]
  206. De Castro Silva, I.; Bianchi, A.; Deshpande, N.U.; Sharma, P.; Mehra, S.; Garrido, V.T.; Saigh, S.J.; England, J.; Hosein, P.J.; Kwon, D.; et al. Neutrophil-Mediated Fibroblast-Tumor Cell Il-6/Stat-3 Signaling Underlies the Association between Neutrophil-to-Lymphocyte Ratio Dynamics and Chemotherapy Response in Localized Pancreatic Cancer: A Hybrid Clinical-Preclinical Study. eLife 2022, 11, e78921. [Google Scholar] [CrossRef]
  207. De Los Reyes, A.A.; Kim, Y. Optimal Regulation of Tumour-Associated Neutrophils in Cancer Progression. R. Soc. Open Sci. 2022, 9, 210705. [Google Scholar] [CrossRef] [PubMed]
  208. Chan, Y.; Tan, H.; Lu, Y.; Zhang, C.; Cheng, C.; Wu, J.; Wang, N.; Feng, Y. Pancreatic Melatonin Enhances Anti-Tumor Immunity in Pancreatic Adenocarcinoma through Regulating Tumor-Associated Neutrophils Infiltration and NETosis. Acta Pharm. Sin. B 2023, 13, 1554–1567. [Google Scholar] [CrossRef]
  209. Mangrolia, U.; Osborne, J.W. Probiotics in Counteracting the Role of Neutrophils in Cancer Metastasis. Vaccines 2021, 9, 1306. [Google Scholar] [CrossRef]
  210. Garcia-Flores, L.A.; Dawid De Vera, M.T.; Pilo, J.; Rego, A.; Gomez-Casado, G.; Arranz-Salas, I.; Hierro Martín, I.; Alcaide, J.; Torres, E.; Ortega-Gomez, A.; et al. Increased Neutrophil Counts Are Associated with Poor Overall Survival in Patients with Colorectal Cancer: A Five-Year Retrospective Analysis. Front. Immunol. 2024, 15, 1415804. [Google Scholar] [CrossRef]
  211. Zhang, W.; Qin, T.; Yang, Z.; Yin, L.; Zhao, C.; Feng, L.; Lin, S.; Liu, B.; Cheng, S.; Zhang, K. Telomerase-Positive Circulating Tumor Cells Are Associated with Poor Prognosis via a Neutrophil-Mediated Inflammatory Immune Environment in Glioma. BMC Med. 2021, 19, 277. [Google Scholar] [CrossRef]
  212. Anderson, R.; Blidner, A.G.; Rapoport, B.L. Frontiers in Pharmacology: Review Manuscript Targeting of the Neutrophil as an Adjunctive Strategy in Non-Small Cell Lung Cancer. Front. Pharmacol. 2021, 12, 676399. [Google Scholar] [CrossRef]
  213. Kast, R.E. High Neutrophil-to-Lymphocyte Ratio Facilitates Cancer Growth—Currently Marketed Drugs Tadalafil, Isotretinoin, Colchicine, and Omega-3 to Reduce It: The TICO Regimen. Cancers 2022, 14, 4965. [Google Scholar] [CrossRef] [PubMed]
  214. Meng, Y.; Sun, J.; Zhang, G.; Yu, T.; Piao, H. Approaches for Neutrophil Imaging: An Important Step in Personalized Medicine. Bioengineered 2022, 13, 14844–14855. [Google Scholar] [CrossRef] [PubMed]
  215. Zhu, Q.; Tao, Y.; Han, Y.; He, Y.; Fu, Y.; Yang, H.; Chen, Y.; Shi, Y. Quercetin Alleviates Breast Cancer-Related Depression by Inhibiting Neutrophil Extracellular Traps via Inhibition of Sphingosine 1-Phosphate/Sphingosine 1-Phosphate Receptor Axis. Phytother. Res. 2025, ptr.8513. [Google Scholar] [CrossRef]
  216. Xiao, L.; Li, J.; Liao, J.; Wu, M.; Lu, X.; Li, J.; Zeng, Y. BCL2A1- and G0S2-driven Neutrophil Extracellular Traps: A Protective Mechanism Linking Preeclampsia to Reduced Breast Cancer Risk. Oncol. Rep. 2025, 53, 64. [Google Scholar] [CrossRef] [PubMed]
  217. Mi, J.; Guo, J.; Kang, K.; Wang, S.; Huang, M. Advances in Targeting Neutrophil Extracellular Traps as a PromisingApproach for Breast Cancer Treatment. CCHTS 2025, 28. [Google Scholar] [CrossRef]
  218. Yan, S.; Zhao, W.; Du, J.; Teng, L.; Yu, T.; Xu, P.; Liu, J.; Yang, R.; Dong, Y.; Wang, H.; et al. C-FOS Promotes the Formation of Neutrophil Extracellular Traps and the Recruitment of Neutrophils in Lung Metastasis of Triple-Negative Breast Cancer. J. Exp. Clin. Cancer Res. 2025, 44, 108. [Google Scholar] [CrossRef]
  219. Huang, Z.; Mo, C.; Li, L.; Hou, Q.; Pan, Y.; Zhu, G.; Qiu, F.; Zou, Q.; Yang, J. Identification of Novel Neutrophil-Extracellular-Traps-Related Genes as Biomarkers for Breast Cancer Prognosis and Immunotherapy. Transl. Cancer Res. 2025, 14, 1737–1752. [Google Scholar] [CrossRef]
  220. Aierken, Y.; Tan, K.; Liu, T.; Lv, Z. Prognosis and Immune Infiltration Prediction in Neuroblastoma Based on Neutrophil Extracellular Traps-Related Gene Signature. Sci. Rep. 2025, 15, 5343. [Google Scholar] [CrossRef]
  221. Shen, J.; Lin, H.; Mo, K.; Liang, Z.; Zhang, Y.; Quan, H.; Wang, X.; Zhang, C.; Chen, C. Bidirectional Roles of Neutrophil Extracellular Traps in Oral Microbiota Carcinogenesis: A Systematic Review. Transl. Oncol. 2025, 56, 102361. [Google Scholar] [CrossRef]
  222. Wang, Q.; Zhang, Y.; Ding, W.; Feng, C.; Wang, Y.; Wei, X.; Qu, Z.; Wang, H.; Liu, X.; Wang, H.; et al. Neutrophil Extracellular Traps Induced by Interleukin 8 via CXCR1/2 Promote the Progression of Gastric Carcinoma through Transcription Factor IIB-Related Factor 1 and Cyclin. Genes Dis. 2024, 11, 575–578. [Google Scholar] [CrossRef] [PubMed]
  223. Ma, Z.; Li, X.; Yang, S.; Yang, H.; Zhang, A.; Li, N.; Zou, X. Molecular Mechanisms of Neutrophil Extracellular Traps in Promoting Gastric Cancer Epithelial–Mesenchymal Transition Through SERPINE-1 Expression. J. Biochem. Amp; Mol. Tox 2025, 39, e70157. [Google Scholar] [CrossRef]
  224. Qian, Y.-Y.; Xu, M.; Huang, X.-K.; Zhu, B. Bioinformatic Analysis Indicated That LINC01150 Might Be a Novel Neutrophil Extracellular Traps-Related Biomarker of Gastric Cancer. Sci. Rep. 2025, 15, 7875. [Google Scholar] [CrossRef]
  225. Liu, Y.; Ma, J.; Ma, Y.; Wang, B.; Wang, Y.; Yuan, J.; Zhang, F.; Zhao, X.; Chen, K.; Zhang, X.; et al. Neutrophil Extracellular Traps Impede Cancer Metastatic Seeding via Protease-Activated Receptor 2-Mediated Downregulation of Phagocytic Checkpoint CD24. J. Immunother. Cancer 2025, 13, e010813. [Google Scholar] [CrossRef] [PubMed]
  226. Xia, Y.; Liu, J.; Meng, Q.; Zuo, X.; Sun, B.; Liu, Y.; Guo, Y.; Wang, M.; Yan, X.; Zhang, J.; et al. Neutrophil Extracellular Traps Promote a Liver Metastatic Phenotype in Human Colorectal Cancer Cells through Epithelial—Mesenchymal Transition. Preprint 2025. [Google Scholar] [CrossRef]
  227. Zhou, M.; Guan, B.; Liu, Y.; Gu, Q.; Chen, W.; Xie, B.; Zhou, M.; Xiang, J.; Zhao, S.; Zhao, Q.; et al. Fibrinogen-like 2 in Tumor-Associated Macrophage-Derived Extracellular Vesicles Shapes an Immunosuppressive Microenvironment in Colorectal Liver Metastases by Promoting Tumor Stemness and Neutrophil Extracellular Traps Formation. Cancer Lett. 2025, 618, 217642. [Google Scholar] [CrossRef]
  228. Wang, X.; He, S.; Gong, X.; Lei, S.; Zhang, Q.; Xiong, J.; Liu, Y. Neutrophils in Colorectal Cancer: Mechanisms, Prognostic Value, and Therapeutic Implications. Front. Immunol. 2025, 16, 1538635. [Google Scholar] [CrossRef]
  229. Kong, X.; Zhang, Y.; Xiang, L.; You, Y.; Duan, Y.; Zhao, Y.; Li, S.; Wu, R.; Zhang, J.; Zhou, L.; et al. Fusobacterium Nucleatum-Triggered Neutrophil Extracellular Traps Facilitate Colorectal Carcinoma Progression. J. Exp. Clin. Cancer Res. 2023, 42, 236. [Google Scholar] [CrossRef]
  230. Suzuki, T.; Tsujimoto, H.; Watanabe, T.; Ishibashi, Y.; Fujishima, S.; Itazaki, Y.; Kariya, R.; Uehata, N.; Shinada, H.; Mochizuki, S.; et al. Clinical Significance of Coiled-Coil Domain-Containing Protein 25 Expression in Esophageal Squamous Cell Carcinoma. Ann. Surg. Oncol. 2025, 32, 3839–3850. [Google Scholar] [CrossRef]
  231. Hong, Z.; Liu, Q.; Zhang, Y.; Shi, X.; Jing, D.; Cheng, T.; Liu, H.; Piao, H.; Gou, Y. The Tumor-Promoting Role of Neutrophil Extracellular Traps in Esophageal Squamous Cell Carcinoma and Their Interaction with the Gut Microbiota. Preprint 2025. [Google Scholar] [CrossRef]
  232. Baron, S.; Binenbaum, Y.; Maman, R.; Fidel, V.; Shusterman, A.; Vaisman, D.; Sher, O.; Manisterski, M.; Shukrun, R.; Rössig, C.; et al. Neutrophil Extracellular Traps Are Associated with Poor Response to Neoadjuvant Therapy and Poor Survival in Pediatric Osteosarcoma. Front. Oncol. 2025, 15, 1472716. [Google Scholar] [CrossRef] [PubMed]
  233. Chu, D.; Huang, R.; Shi, J.; Xu, R.; Wei, D. NETs-Related Genes Predict Prognosis and Are Correlated with the Immune Microenvironment in Osteosarcoma. Front. Oncol. 2025, 15, 1551074. [Google Scholar] [CrossRef] [PubMed]
  234. Lee, W.; Ko, S.Y.; Akasaka, H.; Weigert, M.; Lengyel, E.; Naora, H. Neutrophil Extracellular Traps Promote Pre-Metastatic Niche Formation in the Omentum by Expanding Innate-like B Cells That Express IL-10. Cancer Cell 2025, 43, 69–85.e11. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The function of neutrophils in tumors. Neutrophils have varied roles across tumor stages. During tumor initiation, they can foster tumorigenesis via chronic inflammation, pro-inflammatory cytokines, and non-coding RNAs. As the tumor progresses, they can either support or hinder tumor growth via mechanisms including N1/N2 polarization, NETs, angiogenesis modulation, and the release of cell-killing substances. In the metastatic phase, neutrophils promote the dissemination of tumor cells to distant sites by facilitating tumor migration, promotion of PMN formation, and activation dormant tumor cells.
Figure 1. The function of neutrophils in tumors. Neutrophils have varied roles across tumor stages. During tumor initiation, they can foster tumorigenesis via chronic inflammation, pro-inflammatory cytokines, and non-coding RNAs. As the tumor progresses, they can either support or hinder tumor growth via mechanisms including N1/N2 polarization, NETs, angiogenesis modulation, and the release of cell-killing substances. In the metastatic phase, neutrophils promote the dissemination of tumor cells to distant sites by facilitating tumor migration, promotion of PMN formation, and activation dormant tumor cells.
Biomedicines 13 01473 g001
Figure 2. Neutrophil granulopoiesis and granule changes. Neutrophil development begins with lymphoid-primed multipotent progenitors (LMPP). These cells, derived from multipotent progenitors (MPP), first differentiate into granulocyte-monocyte progenitors (GMP) during development. The GMP gives rise to the myeloblast, the initial recognizable myeloid form. Myeloblasts mature into promyelocytes, which acquire primary/azurophilic granules, then into myelocytes, which have secondary/specific granules and an eccentric nucleus. These develop into metamyelocytes and subsequently into band cells, characterized by a band-shaped, non-segmented nucleus. The final maturation step sees the band cell’s nucleus segmenting to form the phagocytic mature neutrophil, which acquires tertiary/gelatinase granules. Neutrophil granules include three main types: primary/azurophilic, containing MPO, NE, proteinase 3, defensins, and cathepsins; secondary/specific, which contain cathelicidin, lysozyme, and lactoferrin; and tertiary/gelatinase, containing lysozyme and cathelicidin.
Figure 2. Neutrophil granulopoiesis and granule changes. Neutrophil development begins with lymphoid-primed multipotent progenitors (LMPP). These cells, derived from multipotent progenitors (MPP), first differentiate into granulocyte-monocyte progenitors (GMP) during development. The GMP gives rise to the myeloblast, the initial recognizable myeloid form. Myeloblasts mature into promyelocytes, which acquire primary/azurophilic granules, then into myelocytes, which have secondary/specific granules and an eccentric nucleus. These develop into metamyelocytes and subsequently into band cells, characterized by a band-shaped, non-segmented nucleus. The final maturation step sees the band cell’s nucleus segmenting to form the phagocytic mature neutrophil, which acquires tertiary/gelatinase granules. Neutrophil granules include three main types: primary/azurophilic, containing MPO, NE, proteinase 3, defensins, and cathepsins; secondary/specific, which contain cathelicidin, lysozyme, and lactoferrin; and tertiary/gelatinase, containing lysozyme and cathelicidin.
Biomedicines 13 01473 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Li, P.; Fan, F.; Zhang, B.; Yuan, C.; Liang, H. Neutrophil Spatiotemporal Regulatory Networks: Dual Roles in Tumor Growth Regulation and Metastasis. Biomedicines 2025, 13, 1473. https://doi.org/10.3390/biomedicines13061473

AMA Style

Li P, Fan F, Zhang B, Yuan C, Liang H. Neutrophil Spatiotemporal Regulatory Networks: Dual Roles in Tumor Growth Regulation and Metastasis. Biomedicines. 2025; 13(6):1473. https://doi.org/10.3390/biomedicines13061473

Chicago/Turabian Style

Li, Pengcheng, Feimu Fan, Bixiang Zhang, Chaoyi Yuan, and Huifang Liang. 2025. "Neutrophil Spatiotemporal Regulatory Networks: Dual Roles in Tumor Growth Regulation and Metastasis" Biomedicines 13, no. 6: 1473. https://doi.org/10.3390/biomedicines13061473

APA Style

Li, P., Fan, F., Zhang, B., Yuan, C., & Liang, H. (2025). Neutrophil Spatiotemporal Regulatory Networks: Dual Roles in Tumor Growth Regulation and Metastasis. Biomedicines, 13(6), 1473. https://doi.org/10.3390/biomedicines13061473

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop