Next Article in Journal
Molecular Mechanisms of the Stripe Rust Interaction with Resistant and Susceptible Wheat Genotypes
Previous Article in Journal
Identification of Key Ubiquitination Sites Involved in the Proteasomal Degradation of AtACS7 in Arabidopsis
Previous Article in Special Issue
SIGLEC-5/14 Inhibits CD11b/CD18 Integrin Activation and Neutrophil-Mediated Tumor Cell Cytotoxicity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Neutrophils at the Crossroads: Unraveling the Multifaceted Role in the Tumor Microenvironment

by
Deepika Awasthi
1,* and
Aditya Sarode
2
1
Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY 10065, USA
2
Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(5), 2929; https://doi.org/10.3390/ijms25052929
Submission received: 31 January 2024 / Revised: 23 February 2024 / Accepted: 26 February 2024 / Published: 2 March 2024

Abstract

:
Over the past decade, research has prominently established neutrophils as key contributors to the intricate landscape of tumor immune biology. As polymorphonuclear granulocytes within the innate immune system, neutrophils play a pivotal and abundant role, constituting approximately ∼70% of all peripheral leukocytes in humans and ∼10–20% in mice. This substantial presence positions them as the frontline defense against potential threats. Equipped with a diverse array of mechanisms, including reactive oxygen species (ROS) generation, degranulation, phagocytosis, and the formation of neutrophil extracellular traps (NETs), neutrophils undeniably serve as indispensable components of the innate immune system. While these innate functions enable neutrophils to interact with adaptive immune cells such as T, B, and NK cells, influencing their functions, they also engage in dynamic interactions with rapidly dividing tumor cells. Consequently, neutrophils are emerging as crucial regulators in both pro- and anti-tumor immunity. This comprehensive review delves into recent research to illuminate the multifaceted roles of neutrophils. It explores their diverse functions within the tumor microenvironment, shedding light on their heterogeneity and their impact on tumor recruitment, progression, and modulation. Additionally, the review underscores their potential anti-tumoral capabilities. Finally, it provides valuable insights into clinical therapies targeting neutrophils, presenting a promising approach to leveraging innate immunity for enhanced cancer treatment.

1. Introduction

Neutrophils stand out as the predominant immune cells, serving as crucial effectors in the antimicrobial response within human blood. Often referred to as polymorphonuclear cells due to their distinctive lobulated nuclei, these cells have a brief lifespan of 12–24 h [1]. The bone marrow orchestrates their constant replenishment at an astonishing rate of 1011 neutrophils per day, rendering them the powerhouse of the innate immune system [2]. Beyond their baseline characteristics, the lifespan of neutrophils can be altered upon activation, and the degree of activation plays a pivotal role in septic or aseptic pathology outcomes. As granulocytes, neutrophils harbor a formidable arsenal of cytotoxic factors, including antimicrobial compounds, serine proteases, lysozyme, defensins, and metalloproteases. Additionally, they possess highly intense machinery of enzymes that orchestrate oxidative bursts, endowing them with proficiency in defense mechanisms like degranulation, phagocytosis, and the release of neutrophil extracellular traps (NETs).
Advancements in techniques such as in vivo imaging, high-dimensional transcriptomic and epigenomic analyses, and studies conducted at the single-cell resolution have prompted a reevaluation of neutrophil biology in the context of cancer [3]. Notably, cancers often coincide with the recruitment of neutrophils to tumors and an increase in their circulation, correlating with poor prognosis in many instances. Consequently, the neutrophil/lymphocyte ratio emerges as a clinically relevant prognostic indicator in certain cancers [1]. Recent research has underscored the multifaceted role of neutrophils in cancer, influencing tumor growth, metastasis, cancer stem cell maintenance, exit from dormancy, cell cycle progression, immunosurveillance impairment, therapeutic resistance, antigen presentation, T cell response co-regulation, and antibody-dependent tumor cell clearance [4,5,6,7,8,9]. Neutrophils exhibit a remarkable plasticity, assuming both pro- and anti-tumor functions within the tumor microenvironment.
Recent reviews have comprehensively analyzed neutrophil functions in the context of cancer and tumor progression and resistance, shedding light on their clinical relevance for cancer patients [3,9,10]. In this review, we aim to consolidate the current understanding of the intricate interplay between neutrophils and cancer, with a focus on recent research elucidating their diverse functions at different stages of cancer metastasis.

2. Neutrophil Heterogeneity in Tumors

Tumor-associated neutrophils (TANs) play a pivotal role in the complex landscape of the tumor microenvironment (TME), exerting both stimulatory and suppressive effects on tumor development. The dual nature of their impact is contingent upon their activation status and the signals present in the TME. Originally categorized into low-density neutrophils (LDNs) and high-density neutrophils (HDNs) based on cell density, recent advancements in single-cell RNA sequencing (scRNA-seq) and mass cytometry by time-of-flight (CyTOF) have unveiled a more intricate and heterogeneous neutrophil subpopulation.
In liver cancer, scRNA-seq revealed 11 distinct neutrophil subsets, each displaying unique gene signatures regulated by transcription factors in a spatiotemporal manner. Notably, three subsets (Neu_01_MMP8, Neu_07_APOA2, and Neu_08_CD74) were predominantly enriched in tumors, signifying TANs. Three TAN subsets (Neu_01_MMP8, Neu_07_APOA2, and Neu_08_CD74) were associated with a worse prognosis, underscoring their pro-tumor functions. Integrating machine learning in cohorts of hepatocellular carcinoma (HCC) patients′ bulk-sequencing and single-cell sequencing data resulted in the development of a risk model termed the “neutrophil-derived signature” (NDS), which demonstrated superior accuracy in predicting prognosis compared to clinical variables [11].
In melanoma patients, mass cytometry identified seven neutrophil clusters, revealing terminally differentiated subsets that decreased during tumor progression and others that increased. Similarly, in lung cancer, mass cytometry identified subpopulations within LDN/HDN neutrophil subsets, with LDN subsets being highly enriched in the low-density fraction of advanced lung cancer patients. A unique CD66b+/CD10low/CXCR4+/PDL1 intermediate subset was exclusively found in advanced lung cancer patients, and its presence correlated with poorer prognosis [12].
Moreover, in non-small cell lung carcinoma (NSCLC), the activation of Smad3 in N2 TANs was negatively correlated with the N1 population and patient survival, suggesting heterogeneity in neutrophil tumor subset infiltration may impact tumor progression/regression [13]. Collectively, these findings underscore the critical importance of understanding neutrophil plasticity in the context of tumor progression, offering insights that may inform prognostic models and therapeutic interventions.

3. Pro-Tumoral Roles of Neutrophils

3.1. Neutrophil Recruitment in Tumors

The recruitment of neutrophils orchestrates a complex interplay influenced by various factors, including local signals at the injury/circulation site. As neutrophils migrate to the tumor site, alterations in their transcriptional profiles occur, shaped by the organs where they are recruited. These changes give rise to distinct phenotypic functions primarily driven by the varied expression of adhesion molecules and signaling through CXCL12-CXCR4 [14]. Notably, these studies underscore that neutrophils exhibit specialized functions in a tissue-specific manner.
Neutrophil recruitment is further intricately regulated by signaling between CXCR1 and CXCL8 at the recruitment site [15]. Once recruited, neutrophils exert an influence on T cell tumoricidal activity by enhancing regulatory T cell recruitment and activity within the tumor microenvironment (TME) across various cancer types [16,17]. Additionally, neutrophils secrete various factors, including neutrophil elastase (NE), matrix metalloproteinases (MMPs), reactive oxygen species (ROS), vascular endothelial growth factor (VEGF), and others. These factors can modify the TME, fostering conditions conducive to cancer cell growth and spread. Another mechanism by which neutrophil recruitment into the tumors can happen is by CXCL1/CXCR2 interaction, where the monomeric and dimeric CXCL1 can mediate neutrophil recruitment by binding with the glycosaminoglycans (GAC) and forming a gradient, helping the recruitment of neutrophils [18,19]. The GAC is a major component in the ECM and affects many properties of the tumor; one of the mechanisms can be the recruitment of the neutrophils via the CXCL1/CXCR2 interaction [20,21]. This can be more clearly seen in the study by Sengupta et al., where the tumor-conditioned media from highly aggressive breast cancer was able to recruit more neutrophils compared to a less aggressive one [22]. The authors showed that neutrophil recruitment and trafficking in breast cancer was mediated by CXCL1/2/3 in tandem with TGF-β. Moreover, the monomeric and dimeric forms of CXCL1 and CXCL2 and their ability to interact with CXCR2/GAG/Ackr1 may determine tissue-specific neutrophil recruitment [19]. Recently it was also shown that the CXCL1-CXCL2 heterodimer is potent in recruiting neutrophils [23].
Neutrophils induce a pro-tumoral environment by generating neutrophil extracellular traps (NETs), composed of modified chromatin adorned with bactericidal proteins. This NETosis, influenced by changes in mitochondrial metabolism, inhibits cytotoxic T lymphocyte access to cancer cells, promoting cancer progression. NETs play a crucial role in metastasis, with the dysregulation of gut microbiota being essential for colorectal cancer metastasis to the liver [24,25]. The interaction between NETs and CCDC25 on tumor cells enhances cancer cell motility via the activation of the IL-beta parvin pathway, facilitating metastasis [26]. Additionally, NETs activate dormant cancer cells through inflammation and are mechanistically linked to the proteolytic remodeling of laminin activated by integrin alpha3beta1 signaling [27].
Beyond its role in metastasis and angiogenesis, NETs impact immunotherapy by excluding cytotoxic CD8+ T cells from tumors, a process that can be reversed by blocking IL17 [28]. Colorectal cancer metastasis to the liver is primarily mediated by bacteria dissemination from the primary site, recruiting neutrophils via cytokines such as IL1-β, CCL2, TNFα, and IL6, ultimately forming a pre-metastatic niche [29]. Furthermore, CXCL1 and CXCL3 production, facilitated by TGF-β and SMAD3 signaling, leads to increased infiltration of immunosuppressive neutrophils in the liver, contributing to metastasis [30].
The immune response and outcomes are contingent on the type of neutrophils, i.e., immature recruited neutrophils or neutrophils aged at the site, resulting in differential ROS production and NET formation. Circadian rhythms, governed in part by neutrophil release from the bone marrow and the expression of transcription factors C/EBP and GATA1, influence this process [31,32]. CXCL12 signaling to CXCR4 further regulates neutrophil release, and recent studies suggest that CXCR2, a circadian rhythm-controlling factor, affects neutrophil granule loss and NET formation [33]. External factors like the microbiota impact neutrophil aging via TLR and MyD88 pathways, influencing its function toward a proinflammatory phenotype at the recruitment site [34].
Neutrophils recruited to the TME can be functionally classified into two types: N1, with anti-tumor functions induced by IFN-β and N2, with pro-tumor functions induced by TGF-β [35,36]. The N1 and N2 subsets exhibit plasticity during tumor progression, with the protumor effects of N1 associated with TNFα, ICAM-1, ROS, Fas, and reduced arginase expression. Conversely, the effects of N2 stem from MMP9, VEGF, arginase, and chemokines that promote tumor metastasis [37]. NK cells can modify neutrophil function via the IL-17A-neutrophil axis, dependent on IFN-γ. Interestingly, when neutrophils are depleted using the antimetabolite drug, the anti-tumor effect is highly dependent on the absence of NK cells, highlighting the crucial role of NK cells in neutrophil function [38].

3.2. Role in DNA Damage

In the context of lung cancer, the well-established association between smoking and increased lung cancer risk is mechanistically linked to the recruitment of neutrophils in the lungs, leading to the generation of elevated levels of reactive oxygen species (ROS) due to the presence of urethane in cigarette smoke. This heightened ROS production results in DNA damage, a pivotal factor in the progression toward cancer [39]. Notably, studies have demonstrated that G-CSF knock-out mice do not exhibit urethane-induced lung carcinogenesis, underscoring the specific role of neutrophils in this process [40].
Recent insights highlight that excessive ROS production and widespread transcription by neutrophils are the primary sources of NETosis, a process initiating chromatin decondensation. During NADPH-dependent ROS production, the key DNA repair protein PCNA translocates from the cytoplasm to the nucleus. Inhibiting this DNA repair protein formation impedes NETosis [41]. DNA, in this context, serves as an immunogenic mediator of inflammation alongside its role in NETosis. Toll-like receptor-9 (TLR9), cyclic GMP-AMP synthase (cGAS), NOD-like receptor protein 3 (NLRP3), and absence in melanoma 2 (AIM2) have been implicated in the formation of NETs [42].
Beyond the established role of ROS in NETosis, recent evidence highlights an ROS-independent mechanism of DNA damage by neutrophils, involving the release of microRNAs as microparticles. MicroRNAs such as miR-23a and miR-155 play a role in colorectal cancer (CRC) progression by inducing double-stranded breaks, inhibiting homology-directed repair carried out by Rad51 [43]. Targeting these microRNAs presents a promising avenue for inhibiting neoplasia. Interestingly, the same miR-155 can have both pro- and anti-tumor effects in different stages of CRC. In the early stages, miR-155 targets the Rad51 axis, decreasing homology-directed repair and reducing cancer cell survival. However, in later stages, miR-155 targets the non-homologous end-joining (NHEJ) pathway, increasing cancer cell survival by enhancing Ku70 foci formation [44,45].
Spontaneous NETosis is regulated by DNA polymerase Beta and DNA polymerase delta, controlling the baseline of ROS-induced DNA damage. ROS-induced NETosis leads to DNA lesions, halting genome-wide transcription and replication. This effect can be reversed by scavenging ROS or using NOX inhibitors. Inhibitors targeting later steps of DNA repair, predominantly involving PCNA and polymerase β/δ, increase NETosis levels, while inhibitors of earlier steps inhibit NETosis, suggesting that DNA damage in neutrophils has a regulatory role [46]. APE1, PARP, and DNA ligase are key proteins involved in this regulation. Targeting these DNA damage response pathway proteins using nuclear-penetrating anti-DNA autoantibodies significantly inhibits NETosis in both NADPH oxidase-dependent and -independent mice models [47].
DNA damage caused by neutrophils also regulates telomere dysfunction and senescence. The recruitment of neutrophils via the senescence-associated secretory phenotype results in an aged liver. Co-culturing neutrophils with fibroblast cells increases DNA damage and foci formation involving DNA damage response proteins γH2A.X (or 53BP1) and telomeres in fibroblasts [48].

3.3. Role in Tumor Cell Proliferation

Tumor initiation is a multi-step process involving oncogenic mutations in tissue progenitor cells, followed by proliferation and the eventual triggering of a pro-inflammatory response. Given the established role of neutrophils as key contributors to inflammatory responses, they play a crucial role in shaping tumor onset and progression, particularly within the tumor microenvironment (TME).
Following their development in the bone marrow, neutrophils undergo mobilization into the bloodstream and are subsequently recruited to tumor sites, facilitated by factors such as CXCL1. The interaction between CXCL1 and the abundance of CXCR2 on neutrophils leads to the accumulation of tumor-associated neutrophils (TANs) at these sites. This accumulation inhibits CD4+ and CD8+ T cells, primarily through the expression of myeloperoxidase (MPO) and Fas/FasL, thereby promoting tumor growth [49,50]. Recent findings highlight the potential prognostic significance of local CXCL1/2 expression within the TME [51]. The expression of CXCL1 is intricately regulated by miR-146a, influencing neutrophil recruitment and subsequently promoting tumor growth. Conversely, upregulating miR142 reduces tumor burden by diminishing neutrophil accumulation and enhancing CD8+ T cell presence in the tumor [52]. Tumor cells actively produce agonists of CXCR1 and CXCR2 to induce a neutrophil trap, preventing immune cytotoxicity and protecting tumors from CTL and NK cell attacks [53]. Increased neutrophil recruitment is often associated with an elevated expression of granulocyte colony-stimulating factor (G-CSF) and granulocyte-macrophage colony-stimulating factor (GM-CSF), which drive hematopoiesis toward granulocyte production [54].
Cancer cells, particularly cancer stem cells, produce chitinase-3-like protein 1 (CHI3L1), fostering neutrophil recruitment and exerting a pro-tumor function by modulating immunosuppressive T cell functions [55,56]. The release of IL-8 from dendritic cells is involved in the retention of neutrophils, forming a potential positive feedback loop between neutrophils and dendritic cells via CHI3L1 [56,57].
While both human and mouse neutrophils share consensus functions, some factors exhibit distinct effects. Notably, neutrophil elastase (ELANE) expression selectively targets cancer cells without harming normal cells [58,59]. The lungs, characterized by increased expression of platelet endothelial cell adhesion molecule-1 (PECAM-1), facilitate enhanced recruitment of neutrophils, allowing their trans-endothelial migration into the alveolar space and contributing to pro-tumorigenic effects [60].

3.4. Role in Extracellular Matrix (ECM) Remodeling

The Extracellular Matrix (ECM) constitutes a meticulously organized network of macromolecules, including proteins and polysaccharides, synthesized, and secreted by cells into the extracellular space. Major ECM components encompass collagen, elastin, fibronectin, laminin, aminoglycans, and proteoglycans. Neutrophils play a pivotal role in ECM remodeling through the release of specific matrix-remodeling enzymes such as neutrophil elastase and metalloproteinase, the formation of neutrophil extracellular traps (NETs), and the discharge of exosomes via secondary and tertiary particles [61].
The interstitial matrix and basement membrane of the ECM regulate tissue homeostasis and immune responses, influencing cell survival, growth, differentiation, and migration of immune cells in response to external stimuli [62]. Neutrophils expressing the integrin α6β1 receptor facilitate their transmigration across the ECM, primarily composed of collagen, laminin, and proteoglycans [63]. Neutrophil-derived proteases, including neutrophil elastase, matrix metalloproteinases, cathepsins, neutrophil gelatinase-associated lipocalin, NETs, and neutrophil-derived exosomes, modulate the ECM. The expression levels of neutrophil elastase can serve as a recurrence marker for differentiated thyroid cancer patients [59,64].
The activation of extracellular signal-regulated kinase (ERK) by neutrophil elastase contributes to the migration of colorectal cancer cells [65]. ECM remodeling, mediated by neutrophils, influences tumor cell survival and migration through Src/PI3K-dependent activation of Akt signaling, crucial for tumor cell dissemination in vivo [66]. Secreted neutrophil elastase sequentially cleaves laminin, activating dormant cancer cells by triggering integrin α3β1 signaling [27]. Stromal cell remodeling in the lung by neutrophil elastase promotes lung cancer [67].
Neutrophils, through heat shock–integrin signaling, participate in ECM healing in wounds. Increased expression of collagen-binding integrins αMβ2, ITGAM, and ITGβ2, coupled with neutrophil homing dependent on CXCR2, NOS, and LTB4R, enhances the healing process [68]. NETs released by neutrophils cleave fibronectin in lung alveolar tissues with the assistance of MMP9 [69]. In the mucosa of ulcerative colitis patients, neutrophil-derived NET elastase exhibits high elastinolytic activity, cleaving the therapeutic monoclonal antibody TNFα. Restoring balance is crucial, and the use of elafin is instrumental in achieving this [70]. In bone matrix scenarios, neutrophil-derived NET elastase disrupts the cartilage matrix, leading to the release of membrane-bound peptidylarginine deiminase-2 by fibroblast-like synoviocytes. Subsequent citrullination of cartilage fragments is internalized by fibroblast-like synoviocytes and presented to antigen-specific CD4+ T cells [69]. Neutrophil-derived MMP9 activates TGF-β, suppressing T cells [71].
NETs deposited by neutrophils facilitate neovascularization improve ischemia and hypoxia, activate CCDC25, and enhance metastasis through epithelial–mesenchymal transition (EMT) in gastric cancer. Blocking or knocking down CCDC25 and inhibiting NETs with DNaseI counteract these effects [72]. In diffuse large B-cell lymphoma (DLBCL), NETs recruited via the IL8-CXCR2 axis activate TLR9, increasing NF-κB and p38 pathways and promoting tumor proliferation and growth [73,74,75]. Neutrophil-derived APRIL is another factor contributing to increased tumor aggressiveness in DLBCL [76].
The ANGPT2 and Tie2 axis plays a role in vascular remodeling and tumor progression, with NETs increasing ANGPT2 expression in gastric cancer models [77]. NETs significantly stimulate endothelial cells, decrease CD31, ZO-1, and VE-cadherin expression, contract the cytoskeleton, increase intercellular area, and support proliferation and migration [72]. Neutrophil-derived MMP9 emerges as a major source of angiogenesis for cancer cells, being highly potent and uncomplexed with other MMP inhibitors [78].

3.5. Role in Immunosuppression

Due to their immunosuppressive characteristics, immature neutrophils within the tumor microenvironment (TME) are commonly identified as “polymorphonucler myeloid-derived suppressor cells (PMN-MDSC)” [79]. However, reports also indicate that mature neutrophils also exhibit an immunosuppressive phenotype [80,81]. In mice, these cells are defined as CD11b+Ly6G+Ly6Clow, while in humans, they are identified as CD66b+CD14CD11b+CD15+ [82]. Human PMN-MDSCs additionally express lectin-type oxidized LDL receptor-1 (LOX-1) [83]. PMN-MDSCs predominantly employ reactive oxygen species (ROS), peroxynitrite, arginase-1 (Arg1), and prostaglandin E2 (PGE2) to suppress the functions of T cells, B cells, and natural killer (NK) cells [84,85,86]. These MDSCs elevate arginase-1 levels, which degrade arginine, which is crucial for T cell activity via CD3z expression, modulating the interaction between CCR5 and its ligands [87].
Bianchi et al. highlighted cell-autonomous CXCL1 as a key mediator of spatial T cell restriction through interactions with CXCR2+ neutrophilic MDSCs in human pancreatic ductal adenocarcinoma (PDAC). Neutrophil-derived TNF regulates immunologic rewiring, CXCL1 overproduction from tumor cells, and T cell dysfunction [88]. Gong et al. revealed the tissue-specific immunosuppressive capacity of neutrophils, demonstrating that lung-infiltrating neutrophils mediated immunosuppression through CD140a+ mesenchymal cells (MCs) and prostaglandin-endoperoxide synthase 2 (PTGS2), the rate-limiting enzyme for PGE2 biosynthesis [89]. Furthermore, PMN-MDSCs undergoing ferroptosis exhibit heightened immunosuppression, producing oxygenated lipids and abundant PGE2, limiting the activity of human and mouse T cells [90].
Another avenue through which neutrophils acquire an immunosuppressive phenotype is the expression of PD-L1, interacting with PD-1 on T cells, inhibiting their proliferation and activation. In pancreatic ductal adenocarcinoma (PDAC), neutrophils deficient in purinergic receptor P2RX1 are mobilized and recruited at metastatic sites, accompanied by PD-L1 expression, contributing to immunosuppression [91]. Additionally, Arg1+ and PD-L1+ immunosuppressive neutrophils infiltrate the brain, facilitating brain metastasis development. Elevated expression of enhancer of zest homolog 2 (EZH2), phosphorylated at tyrosine-696 (pY696)-EZH2 by nuclear-localized Src tyrosine kinase, alters its binding preference, increasing c-JUN expression and promoting the recruitment of immunosuppressive neutrophils into the brain [92]. In gastric cancer, cancer cell-derived extracellular vesicles (GC-EVs) transport high-mobility group box-1 (HMGB1), inducing PD-L1 expression on neutrophils and inhibiting T cell immunity [93]. In gastric cancer, activated neutrophils exhibit increased programmed death-ligand 1 (PD-L1) expression, induced by cancer cell-derived GM-CSF. This upregulation of PD-L1 leads to the suppression of normal T cell immunity [94]. The combination of interleukin-17 (IL-17) and G-CSF contributes to heightened hemostasis and neutrophil inflammation [95]. Subtypes of IL-17, such as IL-17C from epithelial cells, further promote neutrophil recruitment in lung tumor growth, influenced by the lung microbiota [96].

3.6. Role in Cytokines/Chemokines Production

Neutrophils constitute a diverse reservoir of cytokines, chemokines, and growth factors that exert significant influence on the tumor microenvironment, consequently shaping tumor outcomes.
In a study by Queen et al., a co-culture of neutrophils with human breast cancer cell lines led to the release of oncostatin M (OSM) by neutrophils, fostering angiogenesis through the induction of vascular endothelial growth factor (VEGF) [97]. In the context of intrahepatic cholangiocarcinoma (ICC), tumor-associated neutrophils (TANs) exhibited a pro-tumorigenic character by producing elevated levels of oncostatin M, activating STAT3 signaling in ICC cells [17]. TGF-β released by neutrophils in breast cancer has been implicated in promoting tumor cell resistance to gemcitabine by inducing epithelial-to-mesenchymal changes in tumor cells [98]. Furthermore, breast cancer cell–neutrophil interaction regulated the pro-tumorigenic phenotype of neutrophils, leading to cancer progression and therapy resistance. Neutrophils exposed to breast cancer cell-derived supernatant displayed heightened levels of interleukin-1β (IL-1β), CC-chemokine ligand-2-4 (CCL2, CCL3, and CCL4), inducible nitric oxide synthase (iNOS), and matrix metallopeptidase-9 (MMP9) [99].
In gastric cancer, TANs were widely distributed in gastric tissues and correlated with tumor stage and poor prognosis. Notably, these TANs produced IL-17A, enhancing the migration, invasion, and epithelial–mesenchymal transition (EMT) of gastric cancer cells through the activation of the Janus kinase 2/signal transducers and activators of transcription (JAK2/STAT3) pathway [94]. Within a lung cancer model, local microbiota activated neutrophil release of IL-1β and IL-23, inducing lung-resident γδ T cells that promoted inflammation and tumor growth [100]. Neutrophils played a role in facilitating the extravasation of tumor cells in lung parenchyma through the secretion of IL-1β and matrix metalloproteinases [101]. Moreover, TANs, as demonstrated by Zhou et al., produced bone morphogenetic protein-2 (BMP2) and transforming growth factor beta2 (TGF-β2), inducing stem cell characteristics in hepatocellular carcinoma (HCC) cells. These stem-like HCC cells, in turn, recruited more TANs via hyperactive NF-κB signaling, establishing a positive feedback loop that increased the proliferation rate of HCCs [102].

3.7. Role in Neutrophil Extracellular Trap Formation

The exploration of neutrophil extracellular traps (NETs) has significantly advanced since their initial observation in 1996 [103] and the subsequent discovery by Brickman et al. in 2004 [104]. These intricate web-like structures, composed of DNA strands and proteins, serve as novel extracellular killing mechanisms adapted by neutrophils to trap and eliminate microorganisms [105,106]. Over almost two decades, ongoing research has unveiled a deeper understanding of NETs’ potential mechanisms and implications in both physiological processes and various pathophysiologies. The involvement of NETs in cancer progression was initially demonstrated when induced by sepsis in the context of liver and lung metastasis [107]. A recent genome-wide association study (GWAS) and exome sequencing linking genetic variants of patients with chronic diseases to circulating NET levels found an association between genetic variants in 10 genes and NET levels, with cancer emerging as the top disease linked to NET-associated genes [108]. NETs’ association has been documented across various cancer types, including breast cancer [109], pancreatic cancer [110], lung cancer [111], ovarian cancer [112], bladder cancer [113], gastric cancer [73], skin cancer [114], and colorectal cancer [115].
Nevertheless, studies in both animal models and patient blood and tumors suggest that NETs may function as either pro- or anti-tumoral factors, contingent on the immune system status or tumor microenvironment [116,117]. Indications of NETs being protumorigenic point to their involvement in cancer progression, metastatic spread, and cancer-associated thrombosis [118,119]. Several mechanisms underline the pro-tumor effects of NETs. The main NET components, neutrophil elastase (NE) and cathepsin G (CG), activate dormant cancer cells by degrading the extracellular matrix (ECM) protein laminin [120]. This process generates an epitope binding to tumor integrins, promoting cancer cell extravasation, proliferation, and metastasis [27,121,122]. NETs enhance angiogenesis, thereby mediating cancer proliferation [123], entrapping cancer cells, acting as adhesion substrates to facilitate metastatic dissemination [107,124], promoting endothelial-to-mesenchymal transition [125], and shielding tumor cells from CD8+ T cells and natural killer (NK) cell-mediated cytotoxicity [53].
Contrastingly, studies have shown that NETs can exert anti-tumor effects. NET components such as myeloperoxidases (MPO), proteinases, and histones demonstrate the ability to eliminate tumors, inhibit tumor growth, and hinder metastasis [126,127]. Intriguingly, there exists a positive feedback loop between tumor-induced NETosis and NET-driven tumor growth, with various components in the tumor microenvironment acting as NET inducers. Cytokines like G-CSF [128], IL-1β [129], Interleukin-8 [73], and CXCR1 and CXCR2 agonists [53] have been demonstrated to stimulate cancer-associated NETosis. In orthotopic tumors, the induction of NET formation via IL17, through the induction of CXCR1/2 agonist chemokines, was observed. This attracted neutrophils, leading to NETosis inhibition, increased infiltration of activated CD8+ T cells, and heightened sensitivity to PD-1 blocking mAbs [28]. Another study by Wang et al. revealed that the pseudo serine protease PRSS35 induces CXCL2 degradation, attenuating neutrophil recruitment to tumors and NET formation, ultimately suppressing hepatocellular carcinoma (HCC) progression [130]. Tumor-induced HMGB1 has also been identified as a stimulator of cancer-associated NETosis [131]. The HMGB1/RAGE/IL-8 axis plays a crucial role in neutrophil recruitment into the tumor microenvironment (TME) and NETosis as HMGB1 was found to bind the RAGE receptor on glioma cells, activating NF-κB and upregulating IL-8 expression—an influential neutrophil chemoattractant [132]. Additionally, tumor cells have been shown to upregulate extracellular vesicle (EV) production, communicating with the stroma to create a tumor-permissive environment. Specifically, human melanoma cell-derived EVs promote neutrophil recruitment and NET production by signaling through the CXCR2/PI3K-AKT axis [114]. Recent findings by Li et al. demonstrated that fibroblast growth factor 19 (FGF19)-induced inflammatory cancer-associated fibroblasts (iCAFs) promote neutrophil infiltration and mediate NET formation in liver metastatic niches via the production of complement C5a and IL-1β, accelerating the liver colonization of colorectal cancer (CRC) cells [133]. Furthermore, Yao et al. reported that NETs interact with TLR9 to decrease merlin phosphorylation, contributing to triple-negative breast cancer (TNBC) cell ferroptosis resistance and promoting TNBC progression [134].

3.8. Role in ROS Generation

Neutrophil-derived reactive oxygen species (ROS) play a pivotal role in fostering mutational burden, thereby propelling cancer initiation and advancement. This occurs through the promotion of oxidative DNA damage in critical tissues such as the lungs and intestines [135]. In a lung tumor model, researchers identified the impact of neutrophil NOX2-derived ROS in facilitating tumor colonization through an IL-1β-dependent pathway [136]. Another study demonstrated that neutrophil-produced ROS induced DNA damage in the lungs, thereby fostering tumor formation, especially under co-treatment with carcinogens [40].
Neutrophils can inhibit the activity of T cells within the TME. The reduced levels of glucose within the TME force the neutrophils to use mitochondrial-derived NADHP for ROS production, which then inhibits the CD4+ T cells and has been shown using a 4T1 tumor-bearing mice model. This mainly occurs by decreasing the viability of these T cells and decreasing the T cells’ ability to produce IFN-γ [137]. Moreover, the ROS derived from the neutrophils inhibit IL17-producing gamma delta T cells, which can act as a major pro-tumoral agent. This mainly occurs as a virtue of reduced production of antioxidant glutathione in these gamma delta T cells, rendering them susceptible to this neutrophil-derived ROS [138]. In the case of circulating neutrophils, they can still inhibit the activity of the T cells by suppressing their proliferation. This inhibition in the immunological synapse requires the expression of integrin Mac-1 on the neutrophil’s surface [139]. However, the immature neutrophils have less T cell suppression activity compared to the mature subset in the case of cancer and the increase in circulating immature neutrophils in the bloodstream of cancer patients represents a mere correlation with the suppressive activity [140]. This has also been shown by Minns et al., where, if the T cell interacts with the neutrophils during the early stage of activation, they are suppressive, but if the T cell has previously been activated, they are not susceptible to the neutrophil-mediated suppression [141].
In the context of breast cancer, tumor-associated neutrophils strategically leverage ROS production to subdue T cells by engaging in oxidative mitochondrial metabolism. This priming of neutrophils, induced by the tumor, allows these cells to sustain an immunosuppressive phenotype, particularly within a glucose-deprived tumor microenvironment [137]. Neutrophil-derived ROS can also exert immunosuppressive effects on T cells by diminishing CD3ζ chain and cytokine expression [142]. Furthermore, neutrophil-derived hydrogen peroxide (H2O2) exhibits the capability to inhibit natural killer (NK) cell function, diminishing tumor clearance and promoting lung colonization in a mouse model of breast cancer metastasis [143]. In mice with a myeloid cell-specific deletion of glutathione peroxidase 4 (Gpx4), a crucial ROS scavenger, more invasive colon tumors developed after repeated injections of the carcinogenic agent azoxymethane. This outcome resulted from excessive ROS production by Gpx4-deficient myeloid cells, amplifying the mutational load of colonic epithelial cells and consequently rendering arising tumors more aggressive [135].
In a subcutaneous cancer mouse model, the expression of inducible nitric oxide synthase (iNOS) by infiltrating neutrophils closely correlates to the number of hypoxanthine phosphoribosyl transferase (Hprt) mutations. These mutations significantly contribute to the genetic abnormalities associated with tumor progression [144]. In the context of inflammatory bowel disease, activated neutrophils release microparticles containing pro-inflammatory microRNAs, including miR-23a and miR-155, resulting in DNA double-strand breaks and subsequent genomic instability. Remarkably, this process occurs in an ROS-independent manner. Evidence of such metabolic shifts is increasingly apparent across various cancer contexts and is intricately linked to immunosuppressive capabilities [43].

4. Anti-Tumoral Roles of Neutrophils

4.1. Role in Tumor Cell Killing

Neutrophils, like other immune cells, exhibit diverse and sometimes opposing functions throughout various stages of cancer. They employ surface receptors, such as Fc receptors, to recognize antibody-opsonized tumor cells. Notably, cancer patients often undergo treatment with tumor-targeting monoclonal antibodies (mAbs). The interaction of Fc receptors promotes neutrophil activation and effector functions. Therefore, opsonizing tumor cells with tumor-associated antigens (TAA)-targeting antibodies enables neutrophils to identify and eradicate tumor cells. Neutrophils play a crucial role in cancer cell elimination through antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP) [145]. Additionally, IgA antibodies, in particular, demonstrate superior cancer cell-killing capabilities compared to IgG. This heightened efficacy is linked to the checkpoint inhibition of the CD47–SIRPα interaction [146,147,148]. SIRPα acts as a signal for inhibiting ADCC by cancer cells, presenting it as a potential target for enhancing neutrophil-mediated ADCC.
Another mechanism facilitating the interaction between neutrophils and various types of tumor cells is Mac-1-dependent cellular contacts, crucial for neutrophil-mediated tumor cell killing [147,149]. In vitro studies revealed close interactions between neutrophils and trastuzumab (IgG)-opsonized SK-BR-3 cells, leading to tumor cell killing [147]. Another study highlighting neutrophil-killing potential demonstrated that combination therapy involving tumor necrosis factor, CD40 agonist, and a tumor-binding antibody induced rapid mobilization and tumor infiltration of neutrophils [150]. The activation of complement component C5a further stimulated neutrophils to produce leukotriene B4, promoting reactive oxygen species production via xanthine oxidase. This cascade resulted in oxidative damage and T cell-independent clearance of multiple tumor types [151].
Neutrophils employ various mechanisms to induce the death of tumor cells, including apoptosis, necrosis, and autophagy. One such pathway through which neutrophils may trigger tumor cell death is the Fas–Fas ligand (FasL) pathway. The Fas receptor, a death receptor (DR) expressed on various cells, undergoes trimerization of the death domain upon cross-linking with its ligand FasL. This activation leads to the subsequent initiation of the caspase pathway and induction of apoptosis [152]. Neutrophils express FasL, a member of the TNF superfamily, and a study suggests that they may release soluble FasL, thereby inducing tumor cell death via the Fas/FasL pathway [153].
TNF-related apoptosis-inducing ligand (TRAIL), another member of the TNF superfamily, interacts with various TRAIL receptors, including death receptors DR4 (TRAIL-R1) and DR5 (TRAIL-R2), as well as decoy receptors DcR1 (TRAIL-R3) and DcR2 (TRAIL-R4) [145]. IFN-stimulated neutrophils are reported to express TRAIL, leading to the induction of tumor cell apoptosis [154]. In another study, evidence of neutrophil-mediated tumor cytotoxicity was observed in IgG-opsonized tumor cells, resulting in necrosis-like cell death and the release of cellular contents. The incubation of human mammary carcinoma cells with human neutrophils and anti-FcαRIxHer2/neu BsAbs revealed autophagic structures and LC3B+ autophagosomes in different human epithelial carcinoma cells, ultimately resulting in neutrophil-mediated autophagic tumor cell death [155].

4.2. Role in Trogocytosis and Trogoptosis of Tumor Cells

Trogocytosis, an intricate mechanism utilized by immune cells, involves the acquisition of membrane proteins from other cells through an endocytic process. During trogocytosis, the immunological synapse facilitates the transfer of plasma membrane fragments from one cell to another [156]. Neutrophils, engaging in close contact with rituximab-opsonized CLL B cells, absorb tumor cell membrane fragments, resulting in a notable decrease in CD20 expression in the tumor cells [157]. Additionally, when neutrophils are incubated with epratuzumab (anti-CD22)-opsonized Daudi B lymphoblast cells, there is a transfer of CD22 from tumor cells to neutrophils [158]. Other tumor-derived membrane proteins, including CD19 and CD79b, have been identified on neutrophils following co-incubation with tumor cells. Likewise, after incubation with rituximab-opsonized B cells, neutrophils express both CD19 and CD20 [159]. Given the expression of various Fc receptors (FcRs) on neutrophils, they are capable of trogocytosis of opsonized tumor cells, forming close cell–cell interactions and a Mac-1-dependent cytotoxic synapse. Subsequently, neutrophils engage in trogocytosis, nibbling small fragments of the cell membrane and cytosol from the tumor cell. Horner et al. proposed that neutrophil trogocytosis may lead to opsonized Raji tumor cell destruction [160]. Matlung et al. demonstrated that neutrophils employ antibody-mediated trogocytosis as a mode of cancer cell destruction, both in vitro and in vivo. This process involves CD11b/CD18-dependent conjugate formation, resulting in trogoptotic (lytic) death of antibody-opsonized solid cancer cells. Interestingly, inhibiting neutrophil trogocytosis prevents trogoptosis of target cells, while enhancing trogocytosis through interference with CD47–SIRPα interactions enhances killing. This highlights a potential avenue for optimizing the clinical efficacy of antibody therapy in cancer [147].

4.3. Role in Tumor Cell Cytotoxicity via Neutrophil Degranulation

Neutrophils, often referred to as granulocytes due to their densely packed cytoplasm filled with numerous cytotoxic granules, house an array of substances, including myeloperoxidase (MPO), neutrophil elastase (NE), lactoferrins, gelatinase, and defensins. Neutrophil degranulation is a process by which neutrophils release their granules, including cytotoxic antimicrobial proteases as a way of non-oxidative killing mechanism. Degranulation can occur at the plasma membrane for extracellular release (killing extracellular microorganisms) or in the phagosome for intracellular delivery (killing intracellular microorganisms) [161]. Neutrophil granules can be broadly classified into four main types: secretory, tertiary, secondary, and primary. Secretory granules include plasma proteins and Fc and complement receptors. Tertiary granules include matrix metalloproteases, such as matrix metallopeptidase 9 (MMP9). Secondary granules involve lysozyme, pre-cathelicidin, and lactoferrin. Primary granules include myeloperoxidase, defensins, elastase, and azurocidin [161]. Although these granules are mostly known for their pro-inflammatory and antimicrobial functions, some studies have suggested that these granules also possess the capability to induce tumor cell cytotoxicity [162,163]. A recent study demonstrated that human neutrophils release catalytically active neutrophil elastase (ELANE), which proteolytically cleaves the CD95 death domain, binding it to histone H1 in cancer cells [27]. The released ELANE from neutrophils exhibits an abscopal effect, mediated by CD8+ T cells [58].

4.4. Role in Crosstalk with Adaptive Immune System

During the initial stages of lung cancer, neutrophils assume a stimulatory role, bolstering T cell function, leading to heightened T cell proliferation and an increased release of IFN-γ. This sequential cascade results in elevated proinflammatory factors and upregulation of costimulatory molecules on T cells [164]. Studies by Fridlender et al. showcased that blocking TGF-β facilitates neutrophil recruitment, thereby exhibiting anti-tumor activity by supporting cytotoxic T cell-mediated responses [36,165]. Similar antitumor effects of neutrophils, achieved through TGF-β inhibition, were observed in colorectal cancer (CRC) [166].
The anti-tumor activity of neutrophils can be further enhanced by obstructing the PD-L1/PD-1 axis on cancer cells [167]. Neutrophil recruitment, crucial for nitric oxide (NO) production, contributes to the effective killing of cancer cells, particularly evident in lung cancer. This process is mediated by the MET and HGF receptors, where blocking MET enhances neutrophil recruitment while concurrently suppressing T cell functions and expansion [168,169,170].
Neutrophils also exert their anti-tumor effects by modulating tumor-associated microbiota through the regulation of IL17, thereby promoting intratumor B cells [171]. However, conflicting results in studies using the same transplanted cell lines indicate context-dependent functions of neutrophils. Discrepancies in experimental methods, sampling time, antibodies used for neutrophil depletion, and cancer progression stages may contribute to these variations, underscoring the necessity for further investigation. In a recent study by Yam et al., tumor neutrophils were found to undergo a transformation from a wound-healing to an acutely activated cytotoxic phenotype. Microbial-activated neutrophils upregulated chemokines, facilitating CD8+ T cell recruitment. They transitioned from sessile VEGF producers to highly motile neutrophils, forming neutrophil-rich domains in tumors, thereby repressing tumor growth [172].

4.5. Role as of Antigen-Presenting Cells

Tumor-infiltrating neutrophils observed in different cancers help in transporting the tumor antigens from the tumor site to the draining lymph nodes, where they are presented to the T cells to initiate the immune response. Along with the expression of CD40 and CD80, this phenomenon of neutrophils is attributed to its potent phagocytosis characteristic [173,174,175]. They are the earliest immune cells to be presenting the tumor antigen. Neutrophil antigen presentation characteristic is conferred by GM-CSF, IFN-γ, and IL-3 [176,177,178]. However, higher expression of the MHC molecule for the antigen presentation on the neutrophils requires cues from the activated T cells, and innate signals like TLR or DAMPs are not sufficient for this [74,179,180]. The neutrophil’s increased ability to process antigen and present it is important for priming Th1 and Th17 cells in an antigen-specific manner [181]. The neutrophil’s APC role primarily occurs as a virtue of decreased DC and T cell migration into the tumor site due to the increased neutrophil-secreted thromboxane A2 [182,183]. Although nAPC presents the antigens, it lacks the conventional ability of robust signaling to the T cells as seen for professional DC. However, this ability to strongly activate T cells at par with the DC can be increased by endocytosis of the antibody-antigen complex via FcγR in vivo and has been shown to elicit CD8+ T cell-dependent anti-tumor immunity [184]. The role of transcription factor PU.1 is critical for this enhanced ability of neutrophils to work as an APC. This approach is gathering much-needed attention as an immunotherapeutic strategy to overcome the drawbacks associated with the use of conventional DC use. A novel approach that enhances immune-mediated tumor regression by activating the noncanonical antigen presentation by the neutrophils makes use of radiotherapy–radiodynamic therapy (RT-RDT) with nanoscale metal–organic frameworks (nMOFs). This has also been found to increase the surface expression of CD80 and CD86 as well as MHCII molecules on the neutrophils for an effective cross-presentation of antigens [185].

5. Clinical Relevance and Future Directions

Neutrophils, with their dynamic responsiveness to infection, inflammation, and shifts in homeostasis, represent complex and intriguing cells. Recent research has unveiled their dual impact on the tumor microenvironment (Figure 1), yielding both favorable and adverse outcomes contingent on tumor types and disease stages. However, the challenges stem from their transient lifespan, low RNA content, and adaptable nature, presenting a lack of distinct markers for discerning between different neutrophil subpopulations [186]. Despite substantial strides in comprehending neutrophil biology, functions, and heterogeneity, numerous questions persist regarding their roles in cancer.
Of note, currently, there are several undergoing preclinical and clinical studies to exploit neutrophil survival and recruitment and their potential anti-tumor functions as a therapeutic strategy in various cancers.
In a phase I study of patients with stage III head and neck squamous cell carcinoma, DS-8273a (an agonistic death receptor 5 (TRAIL-R2) antibody) was used to selectively target MDSCs. Treatment with DS8273a reduced the number of circulating PMN-MDSCs (CD11b+/CD14+/CD33+/CD15+; low-density fraction cells) in patients [187]. Moreover, in a phase II clinical trial, gemcitabine increased the efficacy of nivolumab by killing MDSCs to reduce immunosuppression in stage IIIB NSCLC (NCT03302247). In a phase II clinical trial of patients with stage IV CRC, 5-FU plus bevacizumab and anakinra resulted in an improvement of median progression-free survival [188]. However, these drugs also depleted other subsets of neutrophils as side effects causing neutropenia.
Moreover, current research indicates their involvement in cancer therapy as drugs targeting oncogenic signaling in tumor cells through c-MET, a receptor tyrosine kinase, not only impedes tumor progression but also blocks neutrophil recruitment into tumors and the draining lymph nodes. This absence of neutrophils facilitates T cell expansion, fostering tumor elimination [189]. Similarly, therapies targeting the CXCR2 signaling pathway, which plays a critical role in the trafficking of neutrophils from the bone marrow into the bloodstream and subsequently into the peripheral tissues, are being actively investigated in various cancers. AZD5069, a CXCR2 inhibitor that is used to reduce absolute neutrophil recruitment in bronchiectasis patients [174], is also under a phase I/II study in combination with durvalumab in patients with advanced hepatocellular carcinoma (HCC) [190]. Similarly, clinical trials targeting SX-682, a small-molecule inhibitor of CXCR1 and CXCR2 in combination with pembrolizumab (anti-PD-1 immunotherapy), are undergoing for metastatic or recurrent stage IIIC or IV non-small cell lung cancer [NCT05570825], metastatic melanoma [NCT03161431], and metastatic colorectal cancer (mCRC) [NCT06149481]. Another mechanism that exploits the tumor-killing ability of neutrophils is using TGF-β-blocking antibodies either as monotherapy or as combination therapy with [191] fresolimumab, which has been shown to be used in patients with melanoma [192], renal cell carcinoma [193], and metastatic breast cancer [194]. Moreover, fresolimumab and stereotactic ablative radiotherapy is currently being used in early-stage non-small cell lung cancer in phase I/II trials [NCT02581787]. Similarly, galunisertib is a small-molecule selective inhibitor of TGFβ receptor I that has been used in HCC [195]. In antibody cancer therapy, neutrophils engage with cancer cells, initiating antibody-dependent cellular cytotoxicity (ADCC) and eliminating cancer cells through trogocytosis or potentially degranulation [147].
While existing research predominantly emphasizes targeting the immunosuppressive or other pro-tumoral functions of neutrophils, there is a burgeoning yet promising avenue for exploring their cancer-killing capabilities. A nuanced approach is vital, considering the integral role neutrophils play in innate immunity. Utter suppression of this cell population could elevate infection risks. Hence, identifying markers to selectively target protumor neutrophil subtypes or intervening in signaling pathways that drive their protumorigenic nature emerges as a more prudent and promising therapeutic strategy against cancer. Future studies aimed at unraveling these intricate mechanisms will undoubtedly redefine neutrophils’ roles in cancer growth and metastasis, paving the way for innovative treatments that modulate their behavior in a context-dependent manner.

Author Contributions

Conceptualization by D.A. Manuscript written by D.A. and A.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Burn, G.L.; Foti, A.; Marsman, G.; Patel, D.F.; Zychlinsky, A. The Neutrophil. Immunity 2021, 54, 1377–1391. [Google Scholar] [CrossRef] [PubMed]
  2. Lawrence, S.M.; Corriden, R.; Nizet, V. The Ontogeny of a Neutrophil: Mechanisms of Granulopoiesis and Homeostasis. Microbiol. Mol. Biol. Rev. 2018, 82, e00057-17. [Google Scholar] [CrossRef] [PubMed]
  3. Hedrick, C.C.; Malanchi, I. Neutrophils in cancer: Heterogeneous and multifaceted. Nat. Rev. Immunol. 2022, 22, 173–187. [Google Scholar] [CrossRef]
  4. 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]
  5. Engblom, C.; Pfirschke, C.; Zilionis, R.; Da Silva Martins, J.; Bos, S.; Courties, G.; Rickelt, S.; Severe, N.; Baryawno, N.; Faget, J.; et al. Osteoblasts remotely supply lung tumors with cancer-promoting SiglecFhigh neutrophils. Science 2017, 358, eaal5081. [Google Scholar] [CrossRef] [PubMed]
  6. Wculek, S.K.; Malanchi, I. Neutrophils support lung colonization of metastasis-initiating breast cancer cells. Nature 2015, 528, 413–417. [Google Scholar] [CrossRef] [PubMed]
  7. Shaul, M.E.; Zlotnik, A.; Tidhar, E.; Schwartz, A.; Arpinati, L.; Kaisar-Iluz, N.; Mahroum, S.; Mishalian, I.; Fridlender, Z.G. Tumor-Associated Neutrophils Drive B-cell Recruitment and Their Differentiation to Plasma Cells. Cancer Immunol. Res. 2021, 9, 811–824. [Google Scholar] [CrossRef]
  8. Ancey, P.-B.; Contat, C.; Boivin, G.; Sabatino, S.; Pascual, J.; Zangger, N.; Perentes, J.Y.; Peters, S.; Abel, E.D.; Kirsch, D.G.; et al. GLUT1 Expression in Tumor-Associated Neutrophils Promotes Lung Cancer Growth and Resistance to Radiotherapy. Cancer Res. 2021, 81, 2345–2357. [Google Scholar] [CrossRef]
  9. Quail, D.F.; Amulic, B.; Aziz, M.; Barnes, B.J.; Eruslanov, E.; Fridlender, Z.G.; Goodridge, H.S.; Granot, Z.; Hidalgo, A.; Huttenlocher, A.; et al. Neutrophil phenotypes and functions in cancer: A consensus statement. J. Exp. Med. 2022, 219, e20220011. [Google Scholar] [CrossRef]
  10. McFarlane, A.J.; Fercoq, F.; Coffelt, S.B.; Carlin, L.M. Neutrophil dynamics in the tumor microenvironment. J. Clin. Invest. 2021, 131, e143759. [Google Scholar] [CrossRef]
  11. Gong, Q.; Chen, X.; Liu, F.; Cao, Y. Machine learning-based integration develops a neutrophil-derived signature for improving outcomes in hepatocellular carcinoma. Front. Immunol. 2023, 14, 1216585. [Google Scholar] [CrossRef] [PubMed]
  12. Shaul, M.E.; Eyal, O.; Guglietta, S.; Aloni, P.; Zlotnik, A.; Forkosh, E.; Levy, L.; Weber, L.M.; Levin, Y.; Pomerantz, A.; et al. Circulating neutrophil subsets in advanced lung cancer patients exhibit unique immune signature and relate to prognosis. FASEB J. 2020, 34, 4204–4218. [Google Scholar] [CrossRef] [PubMed]
  13. Yamamoto, M.; Toya, Y.; Jensen, R.A.; Ishikawa, Y. Caveolin is an inhibitor of platelet-derived growth factor receptor signaling. Exp. Cell Res. 1999, 247, 380–388. [Google Scholar] [CrossRef] [PubMed]
  14. Margraf, A.; Ley, K.; Zarbock, A. Neutrophil Recruitment: From Model Systems to Tissue-Specific Patterns. Trends Immunol. 2019, 40, 613–634. [Google Scholar] [CrossRef] [PubMed]
  15. Powell, D.; Lou, M.; Barros Becker, F.; Huttenlocher, A. Cxcr1 mediates recruitment of neutrophils and supports proliferation of tumor-initiating astrocytes in vivo. Sci. Rep. 2018, 8, 13285. [Google Scholar] [CrossRef] [PubMed]
  16. Mishalian, I.; Bayuh, R.; Eruslanov, E.; Michaeli, J.; Levy, L.; Zolotarov, L.; Singhal, S.; Albelda, S.; Granot, Z.; Fridlender, Z. Neutrophils recruit regulatory T-cells into tumors via secretion of CCL17—A new mechanism of impaired antitumor immunity. Int. J. Cancer 2014, 135, 1178–1186. [Google Scholar] [CrossRef] [PubMed]
  17. Zhou, S.-L.; Zhou, Z.-J.; Hu, Z.-Q.; Huang, X.-W.; Wang, Z.; Chen, E.-B.; Fan, J.; Cao, Y.; Dai, Z.; Zhou, J. Tumor-Associated Neutrophils Recruit Macrophages and T-Regulatory Cells to Promote Progression of Hepatocellular Carcinoma and Resistance to Sorafenib. Gastroenterology 2016, 150, 1646–1658.e17. [Google Scholar] [CrossRef]
  18. Sawant, K.V.; Poluri, K.M.; Dutta, A.K.; Sepuru, K.M.; Troshkina, A.; Garofalo, R.P.; Rajarathnam, K. Chemokine CXCL1 mediated neutrophil recruitment: Role of glycosaminoglycan interactions. Sci. Rep. 2016, 6, 33123. [Google Scholar] [CrossRef]
  19. Sawant, K.V.; Sepuru, K.M.; Lowry, E.; Penaranda, B.; Frevert, C.W.; Garofalo, R.P.; Rajarathnam, K. Neutrophil recruitment by chemokines Cxcl1/KC and Cxcl2/MIP2: Role of Cxcr2 activation and glycosaminoglycan interactions. J. Leukoc. Biol. 2021, 109, 777–791. [Google Scholar] [CrossRef]
  20. Wei, J.; Hu, M.; Huang, K.; Lin, S.; Du, H. Roles of Proteoglycans and Glycosaminoglycans in Cancer Development and Progression. Int. J. Mol. Sci. 2020, 21, 5983. [Google Scholar] [CrossRef]
  21. Wieboldt, R.; Läubli, H. Glycosaminoglycans in cancer therapy. Am. J. Physiol. Cell Physiol. 2022, 322, C1187–C1200. [Google Scholar] [CrossRef] [PubMed]
  22. SenGupta, S.; Hein, L.E.; Xu, Y.; Zhang, J.; Konwerski, J.R.; Li, Y.; Johnson, C.; Cai, D.; Smith, J.L.; Parent, C.A. Triple-Negative Breast Cancer Cells Recruit Neutrophils by Secreting TGF-β and CXCR2 Ligands. Front. Immunol. 2021, 12, 659996. [Google Scholar] [CrossRef] [PubMed]
  23. Sawant, K.V.; Sepuru, K.M.; Penaranda, B.; Lowry, E.; Garofalo, R.P.; Rajarathnam, K. Chemokine Cxcl1-Cxcl2 heterodimer is a potent neutrophil chemoattractant. J. Leukoc. Biol. 2023, 114, 666–671. [Google Scholar] [CrossRef] [PubMed]
  24. Wu, J.; Dong, W.; Pan, Y.; Wang, J.; Wu, M.; Yu, Y. Crosstalk between gut microbiota and metastasis in colorectal cancer: Implication of neutrophil extracellular traps. Front. Immunol. 2023, 14, 1296783. [Google Scholar] [CrossRef]
  25. 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] [PubMed]
  26. 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]
  27. 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] [PubMed]
  28. Zhang, Y.; Chandra, V.; Sanchez, E.R.; Dutta, P.; Quesada, P.R.; Rakoski, A.; Zoltan, M.; Arora, N.; Baydogan, S.; Horne, W.; et al. Interleukin-17–induced neutrophil extracellular traps mediate resistance to checkpoint blockade in pancreatic cancer. J. Exp. Med. 2020, 217, e20190354. [Google Scholar] [CrossRef]
  29. Cheng, P.; Wu, J.; Zong, G.; Wang, F.; Deng, R.; Tao, R.; Qian, C.; Shan, Y.; Wang, A.; Zhao, Y.; et al. Capsaicin shapes gut microbiota and pre-metastatic niche to facilitate cancer metastasis to liver. Pharmacol. Res. 2023, 188, 106643. [Google Scholar] [CrossRef]
  30. Wang, H.; Zhang, B.; Li, R.; Chen, J.; Xu, G.; Zhu, Y.; Li, J.; Liang, Q.; Hua, Q.; Wang, L.; et al. KIAA1199 drives immune suppression to promote colorectal cancer liver metastasis by modulating neutrophil infiltration. Hepatology 2022, 76, 967–981. [Google Scholar] [CrossRef]
  31. Aroca-Crevillén, A.; Adrover, J.M.; Hidalgo, A. Circadian Features of Neutrophil Biology. Front. Immunol. 2020, 11, 576. [Google Scholar] [CrossRef] [PubMed]
  32. Adrover, J.M.; Del Fresno, C.; Crainiciuc, G.; Cuartero, M.I.; Casanova-Acebes, M.; Weiss, L.A.; Huerga-Encabo, H.; Silvestre-Roig, C.; Rossaint, J.; Cossio, I.; et al. A Neutrophil Timer Coordinates Immune Defense and Vascular Protection. Immunity 2019, 50, 390–402.e10. [Google Scholar] [CrossRef] [PubMed]
  33. Adrover, J.M.; Aroca-Crevillén, A.; Crainiciuc, G.; Ostos, F.; Rojas-Vega, Y.; Rubio-Ponce, A.; Cilloniz, C.; Bonzón-Kulichenko, E.; Calvo, E.; Rico, D.; et al. Programmed ‘disarming’ of the neutrophil proteome reduces the magnitude of inflammation. Nat. Immunol. 2020, 21, 135–144. [Google Scholar] [CrossRef] [PubMed]
  34. Zhang, D.; Chen, G.; Manwani, D.; Mortha, A.; Xu, C.; Faith, J.J.; Burk, R.D.; Kunisaki, Y.; Jang, J.-E.; Scheiermann, C.; et al. Neutrophil ageing is regulated by the microbiome. Nature 2015, 525, 528–532. [Google Scholar] [CrossRef] [PubMed]
  35. Andzinski, L.; Kasnitz, N.; Stahnke, S.; Wu, C.F.; Gereke, M.; von Köckritz-Blickwede, M.; Schilling, B.; Brandau, S.; Weiss, S.; Jablonska, J. Type I IFN s induce anti-tumor polarization of tumor associated neutrophils in mice and human. Int. J. Cancer 2016, 138, 1982–1993. [Google Scholar] [CrossRef] [PubMed]
  36. 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] [PubMed]
  37. 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]
  38. Ogura, K.; Sato-Matsushita, M.; Yamamoto, S.; Hori, T.; Sasahara, M.; Iwakura, Y.; Saiki, I.; Tahara, H.; Hayakawa, Y. NK Cells Control Tumor-Promoting Function of Neutrophils in Mice. Cancer Immunol. Res. 2018, 6, 348–357. [Google Scholar] [CrossRef]
  39. Jetten, A.M.; Shirley, J.E.; Stoner, G. Regulation of proliferation and differentiation of respiratory tract epithelial cells by TGFβ. Exp. Cell Res. 1986, 167, 539–549. [Google Scholar] [CrossRef]
  40. Wculek, S.K.; Bridgeman, V.L.; Peakman, F.; Malanchi, I. Early Neutrophil Responses to Chemical Carcinogenesis Shape Long-Term Lung Cancer Susceptibility. iScience 2020, 23, 101277. [Google Scholar] [CrossRef]
  41. Azzouz, D.; Khan, M.A.; Palaniyar, N. ROS induces NETosis by oxidizing DNA and initiating DNA repair. Cell Death Discov. 2021, 7, 113. [Google Scholar] [CrossRef] [PubMed]
  42. Aubé, F.-A.; Bidias, A.; Pépin, G. Who and how, DNA sensors in NETs-driven inflammation. Front. Immunol. 2023, 14, 1190177. [Google Scholar] [CrossRef] [PubMed]
  43. Sumagin, R.; Bui, T.M.; Wiesolek, H.L.; Butin-Israeli, V. Neutrophil-induced genomic instability impedes resolution of inflammation and wound healing. FASEB J. 2019, 33, 34.10. [Google Scholar] [CrossRef]
  44. 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] [PubMed]
  45. Bui, T.M. Neutrophils Deregulate Tumoral DNA Damage Response and Contribute to Non-Homologous End-Joining (NHEJ)-dependent Survival of Colorectal Cancer. FASEB J. 2020, 34, 1. [Google Scholar] [CrossRef]
  46. Azzouz, D.; Palaniyar, N. ROS and DNA repair in spontaneous versus agonist-induced NETosis: Context matters. Front. Immunol. 2022, 13, 1033815. [Google Scholar] [CrossRef] [PubMed]
  47. Chen, X.; Cuffari, B.J.; Dubljevic, V.; Shirali, A.; Zhou, J.; Campbell, J.A.; Suits, S.C.; O’sullivan, K.M.; Hansen, J.E. Inhibition of NETosis by a Nuclear-Penetrating Anti-DNA Autoantibody. ImmunoHorizons 2022, 6, 356–365. [Google Scholar] [CrossRef]
  48. Lagnado, A.; Leslie, J.; Ruchaud-Sparagano, M.; Victorelli, S.; Hirsova, P.; Ogrodnik, M.; Collins, A.L.; Vizioli, M.G.; Habiballa, L.; Saretzki, G.; et al. Neutrophils induce paracrine telomere dysfunction and senescence in ROS-dependent manner. EMBO J. 2021, 40, e106048. [Google Scholar] [CrossRef]
  49. 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. Investig. 2010, 120, 2423–2431. [Google Scholar] [CrossRef]
  50. Yuan, M.; Zhu, H.; Xu, J.; Zheng, Y.; Cao, X.; Liu, Q. Corrigendum to “Tumor-Derived CXCL1 Promotes Lung Cancer Growth via Recruitment of Tumor-Associated Neutrophils”. J. Immunol. Res. 2020, 2020, 5106904. [Google Scholar]
  51. Cao, Z.; Fu, B.; Deng, B.; Zeng, Y.; Wan, X.; Qu, L. Overexpression of Chemokine (C-X-C) ligand 1 (CXCL1) associated with tumor progression and poor prognosis in hepatocellular carcinoma. Cancer Cell Int. 2014, 14, 86. [Google Scholar] [CrossRef] [PubMed]
  52. Chen, R.; Coleborn, E.; Bhavsar, C.; Wang, Y.; Alim, L.; Wilkinson, A.N.; Tran, M.A.; Irgam, G.; Atluri, S.; Wong, K.; et al. miR-146a inhibits ovarian tumor growth in vivo via targeting immunosuppressive neutrophils and enhancing CD8+ T cell infiltration. Mol. Ther. Oncolytics 2023, 31, 100725. [Google Scholar] [CrossRef] [PubMed]
  53. Teijeira, Á.; Garasa, S.; Gato, M.; Alfaro, C.; Migueliz, I.; Cirella, A.; de Andrea, C.; Ochoa, M.C.; Otano, I.; Etxeberria, I.; et al. CXCR1 and CXCR2 Chemokine Receptor Agonists Produced by Tumors Induce Neutrophil Extracellular Traps that Interfere with Immune Cytotoxicity. Immunity 2020, 52, 856–871.e8. [Google Scholar] [CrossRef] [PubMed]
  54. Bayne, L.J.; Beatty, G.L.; Jhala, N.; Clark, C.E.; Rhim, A.D.; Stanger, B.Z.; Vonderheide, R.H. Tumor-Derived Granulocyte-Macrophage Colony-Stimulating Factor Regulates Myeloid Inflammation and T Cell Immunity in Pancreatic Cancer. Cancer Cell 2012, 21, 822–835. [Google Scholar] [CrossRef] [PubMed]
  55. Xiao, L.-Y.; Su, Y.-L.; Huang, S.-Y.; Chen, Y.-H.; Hsueh, P.-R. Chitinase 3-like-1 Expression in the Microenvironment Is Associated with Neutrophil Infiltration in Bladder Cancer. Int. J. Mol. Sci. 2023, 24, 15990. [Google Scholar] [CrossRef] [PubMed]
  56. Van Gisbergen, K.P.J.M.; Geijtenbeek, T.B.H.; Van Kooyk, Y. Close encounters of neutrophils and DCs. Trends Immunol. 2005, 26, 626–631. [Google Scholar] [CrossRef] [PubMed]
  57. Alfaro, C.; Suárez, N.; Martínez-Forero, I.; Palazón, A.; Rouzaut, A.; Solano, S.; Feijoo, E.; Gúrpide, A.; Bolaños, E.; Erro, L.; et al. Carcinoma-Derived Interleukin-8 Disorients Dendritic Cell Migration Without Impairing T-Cell Stimulation. PLoS ONE 2011, 6, e17922. [Google Scholar] [CrossRef]
  58. 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]
  59. Jia, W.; Luo, Q.; Wu, J.; Shi, Y.; Guan, Q. Neutrophil elastase as a potential biomarker related to the prognosis of gastric cancer and immune cell infiltration in the tumor immune microenvironment. Sci. Rep. 2023, 13, 13447. [Google Scholar] [CrossRef]
  60. Chen, X.; Chen, J.; Liu, S.; Li, X. PECAM-1 mediates temsirolimus-induced increase in neutrophil transendothelial migration that leads to lung injury. Biochem. Biophys. Res. Commun. 2023, 682, 180–186. [Google Scholar] [CrossRef]
  61. Siddhartha, R.; Garg, M. Interplay Between Extracellular Matrix Remodeling and Angiogenesis in Tumor Ecosystem. Mol. Cancer Ther. 2023, 22, 291–305. [Google Scholar] [CrossRef] [PubMed]
  62. Bonnans, C.; Chou, J.; Werb, Z. Remodelling the extracellular matrix in development and disease. Nat. Rev. Mol. Cell Biol. 2014, 15, 786–801. [Google Scholar] [CrossRef] [PubMed]
  63. Dangerfield, J.; Larbi, K.Y.; Huang, M.-T.; Dewar, A.; Nourshargh, S. PECAM-1 (CD31) Homophilic Interaction Up-Regulates α6β1 on Transmigrated Neutrophils In Vivo and Plays a Functional Role in the Ability of α6 Integrins to Mediate Leukocyte Migration through the Perivascular Basement Membrane. J. Exp. Med. 2002, 196, 1201–1212. [Google Scholar] [CrossRef] [PubMed]
  64. Lee, H.; Kim, T.-S.; Gu, J.-Y.; Yu, M.R.; Lee, S.-E.; Kim, E.S.; Kim, H.K. Value of circulating neutrophil elastase for detecting recurrence of differentiated thyroid cancer. Endocr. Connect. 2023, 12, e230400. [Google Scholar] [CrossRef] [PubMed]
  65. Okamoto, M.; Mizuno, R.; Kawada, K.; Itatani, Y.; Kiyasu, Y.; Hanada, K.; Hirata, W.; Nishikawa, Y.; Masui, H.; Sugimoto, N.; et al. Neutrophil Extracellular Traps Promote Metastases of Colorectal Cancers through Activation of ERK Signaling by Releasing Neutrophil Elastase. Int. J. Mol. Sci. 2023, 24, 1118. [Google Scholar] [CrossRef] [PubMed]
  66. Deryugina, E.; Carré, A.; Ardi, V.; Muramatsu, T.; Schmidt, J.; Pham, C.; Quigley, J.P. Neutrophil Elastase Facilitates Tumor Cell Intravasation and Early Metastatic Events. iScience 2020, 23, 101799. [Google Scholar] [CrossRef] [PubMed]
  67. Topic, A.; Ljujic, M.; Nikolic, A.; Petrovic-Stanojevic, N.; Dopudja-Pantic, V.; Mitic-Milikic, M.; Radojkovic, D. Alpha-1-antitrypsin Phenotypes and Neutrophil Elastase Gene Promoter Polymorphisms in Lung Cancer. Pathol. Oncol. Res. 2011, 17, 75–80. [Google Scholar] [CrossRef]
  68. Fischer, A.; Wannemacher, J.; Christ, S.; Koopmans, T.; Kadri, S.; Zhao, J.; Gouda, M.; Ye, H.; Mück-Häusl, M.; Krenn, P.W.; et al. Neutrophils direct preexisting matrix to initiate repair in damaged tissues. Nat. Immunol. 2022, 23, 518–531. [Google Scholar] [CrossRef]
  69. Carmona-Rivera, C.; Carlucci, P.M.; Goel, R.R.; James, E.; Brooks, S.R.; Rims, C.; Hoffmann, V.; Fox, D.A.; Buckner, J.H.; Kaplan, M.J. Neutrophil extracellular traps mediate articular cartilage damage and enhance cartilage component immunogenicity in rheumatoid arthritis. JCI Insight 2020, 5, e139388. [Google Scholar] [CrossRef]
  70. Curciarello, R.; Sobande, T.; Jones, S.; Giuffrida, P.; Di Sabatino, A.; Docena, G.H.; MacDonald, T.T.; Kok, K. Human Neutrophil Elastase Proteolytic Activity in Ulcerative Colitis Favors the Loss of Function of Therapeutic Monoclonal Antibodies. J. Inflam. Res. 2020, 13, 233–243. [Google Scholar] [CrossRef]
  71. Germann, M.; Zangger, N.; Sauvain, M.O.; 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] [PubMed]
  72. Yang, S.; Sun, B.; Li, J.; Li, N.; Zhang, A.; Zhang, X.; Yang, H.; Zou, X. Neutrophil extracellular traps promote angiogenesis in gastric cancer. Cell Commun. Signal 2023, 21, 176. [Google Scholar] [CrossRef] [PubMed]
  73. Nie, M.; Yang, L.; Bi, X.; Wang, Y.; Sun, P.; Yang, H.; Liu, P.; Li, Z.; Xia, Y.; Jiang, W. Neutrophil Extracellular Traps Induced by IL8 Promote Diffuse Large B-cell Lymphoma Progression via the TLR9 Signaling. Clin. Cancer Res. 2019, 25, 1867–1879. [Google Scholar] [CrossRef] [PubMed]
  74. Patidar, A.; Selvaraj, S.; Sarode, A.; Chauhan, P.; Chattopadhyay, D.; Saha, B. DAMP-TLR-cytokine axis dictates the fate of tumor. Cytokine 2018, 104, 114–123. [Google Scholar] [CrossRef]
  75. Leeman-Neill, R.J.; Song, D.; Bizarro, J.; Wacheul, L.; Rothschild, G.; Singh, S.; Yang, Y.; Sarode, A.Y.; Gollapalli, K.; Wu, L.; et al. Noncoding mutations cause super-enhancer retargeting resulting in protein synthesis dysregulation during B cell lymphoma progression. Nat. Genet. 2023, 55, 2160–2174. [Google Scholar] [CrossRef] [PubMed]
  76. Schwaller, J.; Schneider, P.; Mhawech-Fauceglia, P.; McKee, T.; Myit, S.; Matthes, T.; Tschopp, J.; Donze, O.; Le Gal, F.-A.; Huard, B. Neutrophil-derived APRIL concentrated in tumor lesions by proteoglycans correlates with human B-cell lymphoma aggressiveness. Blood 2007, 109, 331–338. [Google Scholar] [CrossRef] [PubMed]
  77. Yang, S.; Zou, X.; Li, J.; Yang, H.; Zhang, A.; Zhu, Y.; Zhu, L.; Zhang, L. Immunoregulation and clinical significance of neutrophils/NETs-ANGPT2 in tumor microenvironment of gastric cancer. Front. Immunol. 2022, 13, 1010434. [Google Scholar] [CrossRef] [PubMed]
  78. 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]
  79. Talmadge, J.E.; Gabrilovich, D.I. History of myeloid-derived suppressor cells. Nat. Rev. Cancer 2013, 13, 739–752. [Google Scholar] [CrossRef]
  80. Vanhaver, C.; Nana, F.A.; Delhez, N.; Luyckx, M.; Hirsch, T.; Bayard, A.; Houbion, C.; Dauguet, N.; Brochier, A.; van der Bruggen, P.; et al. Immunosuppressive low-density neutrophils in the blood of cancer patients display a mature phenotype. Life Sci. Alliance 2024, 7, e202302332. [Google Scholar] [CrossRef]
  81. Furumaya, C.; Martinez-Sanz, P.; Bouti, P.; Kuijpers, T.W.; Matlung, H.L. Plasticity in Pro- and Anti-tumor Activity of Neutrophils: Shifting the Balance. Front. Immunol. 2020, 11, 2100. [Google Scholar] [CrossRef] [PubMed]
  82. Bronte, V.; Brandau, S.; Chen, S.-H.; Colombo, M.P.; Frey, A.B.; Greten, T.F.; Manduzzato, S.; Murray, P.J.; Ochoa, A.; Ostrand-Rosenberg, S.; et al. Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards. Nat. Commun. 2016, 7, 12150. [Google Scholar] [CrossRef] [PubMed]
  83. Condamine, T.; Dominguez, G.A.; Youn, J.-I.; Kossenkov, A.V.; Mony, S.; Alicea-Torres, K.; Tcyganov, E.; Hashimoto, A.; Nefedova, Y.; Lin, C.; et al. Lectin-type oxidized LDL receptor-1 distinguishes population of human polymorphonuclear myeloid-derived suppressor cells in cancer patients. Sci. Immunol. 2016, 1, aaf8943. [Google Scholar] [CrossRef] [PubMed]
  84. Giese, M.A.; Hind, L.E.; Huttenlocher, A. Neutrophil plasticity in the tumor microenvironment. Blood 2019, 133, 2159–2167. [Google Scholar] [CrossRef] [PubMed]
  85. Gabrilovich, D.I.; Ostrand-Rosenberg, S.; Bronte, V. Coordinated regulation of myeloid cells by tumours. Nat. Rev. Immunol. 2012, 12, 253–268. [Google Scholar] [CrossRef] [PubMed]
  86. Veglia, F.; Sanseviero, E.; Gabrilovich, D.I. Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity. Nat. Rev. Immunol. 2021, 21, 485–498. [Google Scholar] [CrossRef] [PubMed]
  87. Hawila, E.; Razon, H.; Wildbaum, G.; Blattner, C.; Sapir, Y.; Shaked, Y.; Umansky, V.; Karin, N. CCR5 Directs the Mobilization of CD11b+Gr1+Ly6Clow Polymorphonuclear Myeloid Cells from the Bone Marrow to the Blood to Support Tumor Development. Cell Rep. 2017, 21, 2212–2222. [Google Scholar] [CrossRef] [PubMed]
  88. Bianchi, A.; Silva, I.D.C.; Deshpande, N.U.; Singh, S.; Mehra, S.; Garrido, V.T.; Guo, X.; Nivelo, L.A.; Kolonias, D.S.; Saigh, S.J.; et al. Cell-Autonomous Cxcl1 Sustains Tolerogenic Circuitries and Stromal Inflammation via Neutrophil-Derived TNF in Pancreatic Cancer. Cancer Discov. 2023, 13, 1428–1453. [Google Scholar] [CrossRef]
  89. Gong, Z.; Li, Q.; Shi, J.; Li, P.; Hua, L.; Shultz, L.D.; Ren, G. Immunosuppressive reprogramming of neutrophils by lung mesenchymal cells promotes breast cancer metastasis. Sci. Immunol. 2023, 8, eadd5204. [Google Scholar] [CrossRef]
  90. Kim, R.; Hashimoto, A.; Markosyan, N.; Tyurin, V.A.; Tyurina, Y.Y.; Kar, G.; Fu, S.; Sehgal, M.; Garcia-Gerique, L.; Kossenkov, A.; et al. Ferroptosis of tumour neutrophils causes immune suppression in cancer. Nature 2022, 612, 338–346. [Google Scholar] [CrossRef]
  91. Wang, X.; Hu, L.P.; Qin, W.T.; Yang, Q.; Chen, D.Y.; Li, Q.; Zhou, K.X.; Huang, P.Q.; Xu, C.J.; Li, J.; et al. Identification of a subset of immunosuppressive P2RX1-negative neutrophils in pancreatic cancer liver metastasis. Nat. Commun. 2021, 12, 174. [Google Scholar] [CrossRef] [PubMed]
  92. Zhang, L.; Yao, J.; Wei, Y.; Zhou, Z.; Li, P.; Qu, J.; Badu-Nkansah, A.; Yuan, X.; Huang, Y.-W.; Fukumura, K.; et al. Blocking immunosuppressive neutrophils deters pY696-EZH2-driven brain metastases. Sci. Transl. Med. 2020, 12, eaaz5387. [Google Scholar] [CrossRef] [PubMed]
  93. Shi, Y.; Zhang, J.; Mao, Z.; Jiang, H.; Liu, W.; Shi, H.; Ji, R.; Xu, W.; Qian, H. Extracellular Vesicles From Gastric Cancer Cells Induce PD-L1 Expression on Neutrophils to Suppress T-Cell Immunity. Front. Oncol. 2020, 10, 629. [Google Scholar] [CrossRef] [PubMed]
  94. Wang, T.-T.; Zhao, Y.-L.; Peng, L.-S.; Chen, N.; Chen, W.; Lv, Y.-P.; Mao, F.-Y.; Zhang, J.-Y.; Cheng, P.; Teng, Y.-S.; et al. Tumour-activated neutrophils in gastric cancer foster immune suppression and disease progression through GM-CSF-PD-L1 pathway. Gut 2017, 66, 1900–1911. [Google Scholar] [CrossRef] [PubMed]
  95. Akbay, E.A.; Koyama, S.; Liu, Y.; Dries, R.; Bufe, L.E.; Silkes, M.; Alam, M.; Magee, D.M.; Jones, R.; Jinushi, M.; et al. Interleukin-17A Promotes Lung Tumor Progression through Neutrophil Attraction to Tumor Sites and Mediating Resistance to PD-1 Blockade. J. Thorac. Oncol. 2017, 12, 1268–1279. [Google Scholar] [CrossRef]
  96. Jungnickel, C.; Schmidt, L.H.; Bittigkoffer, L.; Wolf, L.; Wolf, A.; Ritzmann, F.; Kamyschnikow, A.; Herr, C.; Menger, M.D.; Spieker, T.; et al. IL-17C mediates the recruitment of tumor-associated neutrophils and lung tumor growth. Oncogene 2017, 36, 4182–4190. [Google Scholar] [CrossRef]
  97. Queen, M.M.; Ryan, R.E.; Holzer, R.G.; Keller-Peck, C.R.; Jorcyk, C.L. Breast cancer cells stimulate neutrophils to produce oncostatin M: Potential implications for tumor progression. Cancer Res. 2005, 65, 8896–8904. [Google Scholar] [CrossRef]
  98. 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]
  99. Wu, L.; Saxena, S.; Goel, P.; Prajapati, D.R.; Wang, C.; Singh, R.K. Breast Cancer Cell-Neutrophil Interactions Enhance Neutrophil Survival and Pro-Tumorigenic Activities. Cancers 2020, 12, 2884. [Google Scholar] [CrossRef]
  100. Jin, C.; Lagoudas, G.K.; Zhao, C.; Bullman, S.; Bhutkar, A.; Hu, B.; Ameh, S.; Sandel, D.; Liang, X.S.; Mazzilli, S.; et al. Commensal Microbiota Promote Lung Cancer Development via γδ T Cells. Cell 2019, 176, 998–1013.e16. [Google Scholar] [CrossRef]
  101. 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]
  102. Zhou, S.; Yin, D.; Hu, Z.; Luo, C.; Zhou, Z.; Xin, H.; Yang, X.; Shi, Y.; Wang, Z.; Huang, X.; et al. A Positive Feedback Loop Between Cancer Stem-Like Cells and Tumor-Associated Neutrophils Controls Hepatocellular Carcinoma Progression. Hepatology 2019, 70, 1214–1230. [Google Scholar] [CrossRef] [PubMed]
  103. Takei, H.; Araki, A.; Watanabe, H.; Ichinose, A.; Sendo, F. Rapid killing of human neutrophils by the potent activator phorbol 12-myristate 13-acetate (PMA) accompanied by changes different from typical apoptosis or necrosis. J. Leukoc. Biol. 1996, 59, 229–240. [Google Scholar] [CrossRef] [PubMed]
  104. Brinkmann, V.; Reichard, U.; Goosmann, C.; Fauler, B.; Uhlemann, Y.; Weiss, D.S.; Weinrauch, Y.; Zychlinsky, A. Neutrophil extracellular traps kill bacteria. Science 2004, 303, 1532–1535. [Google Scholar] [CrossRef] [PubMed]
  105. Fuchs, T.A.; Abed, U.; Goosmann, C.; Hurwitz, R.; Schulze, I.; Wahn, V.; Weinrauch, Y.; Brinkmann, V.; Zychlinsky, A. Novel cell death program leads to neutrophil extracellular traps. J. Cell Biol. 2007, 176, 231–241. [Google Scholar] [CrossRef] [PubMed]
  106. Papayannopoulos, V.; Zychlinsky, A. NETs: A new strategy for using old weapons. Trends Immunol. 2009, 30, 513–521. [Google Scholar] [CrossRef] [PubMed]
  107. Cools-Lartigue, J.; Spicer, J.; McDonald, B.; Gowing, S.; Chow, S.; Giannias, B.; Bourdeau, F.; Kubes, P.; Ferri, L. Neutrophil extracellular traps sequester circulating tumor cells and promote metastasis. J. Clin. Invest. 2013, 123, 3446–3458. [Google Scholar] [CrossRef]
  108. Donkel, S.J.; Fernández, E.P.; Ahmad, S.; Rivadeneira, F.; van Rooij, F.J.A.; Ikram, M.A.; Leebeek, F.W.G.; de Maat, M.P.M.; Ghanbari, M. Common and Rare Variants Genetic Association Analysis of Circulating Neutrophil Extracellular Traps. Front. Immunol. 2021, 12, 615527. [Google Scholar] [CrossRef]
  109. Xiao, Y.; Cong, M.; Li, J.; He, D.; Wu, Q.; Tian, P.; Wang, Y.; Yang, S.; Liang, C.; Liang, Y.; et al. Cathepsin C promotes breast cancer lung metastasis by modulating neutrophil infiltration and neutrophil extracellular trap formation. Cancer Cell 2021, 39, 423–437.e7. [Google Scholar] [CrossRef]
  110. Hisada, Y.; Grover, S.P.; Maqsood, A.; Houston, R.; Ay, C.; Noubouossie, D.F.; Cooley, B.C.; Wallén, H.; Key, N.S.; Thålin, C.; et al. Neutrophils and neutrophil extracellular traps enhance venous thrombosis in mice bearing human pancreatic tumors. Haematologica 2020, 105, 218–225. [Google Scholar] [CrossRef]
  111. Arpinati, L.; Shaul, M.E.; Kaisar-Iluz, N.; Mali, S.; Mahroum, S.; Fridlender, Z.G. NETosis in cancer: A critical analysis of the impact of cancer on neutrophil extracellular trap (NET) release in lung cancer patients vs. mice. Cancer Immunol. Immunother. 2020, 69, 199–213. [Google Scholar] [CrossRef] [PubMed]
  112. 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] [PubMed]
  113. Shinde-Jadhav, S.; Mansure, J.J.; Rayes, R.F.; Marcq, G.; Ayoub, M.; Skowronski, R.; Kool, R.; Bourdeau, F.; Brimo, F.; Spicer, J.; et al. Role of neutrophil extracellular traps in radiation resistance of invasive bladder cancer. Nat. Commun. 2021, 12, 2776. [Google Scholar] [CrossRef]
  114. Guimarães-Bastos, D.; Frony, A.C.; Barja-Fidalgo, C.; Moraes, J.A. Melanoma-derived extracellular vesicles skew neutrophils into a pro-tumor phenotype. J. Leukoc. Biol. 2022, 111, 585–596. [Google Scholar] [CrossRef] [PubMed]
  115. Zhang, Y.; Wang, C.; Yu, M.; Zhao, X.; Du, J.; Li, Y.; Jing, H.; Dong, Z.; Kou, J.; Bi, Y.; et al. Neutrophil extracellular traps induced by activated platelets contribute to procoagulant activity in patients with colorectal cancer. Thromb. Res. 2019, 180, 87–97. [Google Scholar] [CrossRef] [PubMed]
  116. Cools-Lartigue, J.; Spicer, J.; Najmeh, S.; Ferri, L. Neutrophil extracellular traps in cancer progression. Cell Mol. Life Sci. 2014, 71, 4179–4194. [Google Scholar] [CrossRef] [PubMed]
  117. Muqaku, B.; Pils, D.; Mader, J.C.; Aust, S.; Mangold, A.; Muqaku, L.; Slany, A.; Del Favero, G.; Gerner, C. Neutrophil Extracellular Trap Formation Correlates with Favorable Overall Survival in High Grade Ovarian Cancer. Cancers 2020, 12, 505. [Google Scholar] [CrossRef]
  118. Yazdani, H.O.; Roy, E.; Comerci, A.J.; van der Windt, D.J.; Zhang, H.; Huang, H.; Loughran, P.; Shiva, S.; Geller, D.A.; Bartlett, D.L.; et al. Neutrophil Extracellular Traps Drive Mitochondrial Homeostasis in Tumors to Augment Growth. Cancer Res. 2019, 79, 5626–5639. [Google Scholar] [CrossRef]
  119. Wolach, O.; Sellar, R.S.; Martinod, K.; Cherpokova, D.; McConkey, M.; Chappell, R.J.; Silver, A.J.; Adams, D.; Castellano, C.A.; Schneider, R.K.; et al. Increased neutrophil extracellular trap formation promotes thrombosis in myeloproliferative neoplasms. Sci. Transl. Med. 2018, 10, eaan8292. [Google Scholar] [CrossRef]
  120. 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]
  121. Cedervall, J.; Hamidi, A.; Olsson, A.-K. Platelets, NETs and cancer. Thromb. Res. 2018, 164 (Suppl. S1), S148–S152. [Google Scholar] [CrossRef] [PubMed]
  122. Rayes, R.F.; Vourtzoumis, P.; Rjeily, M.B.; Seth, R.; Bourdeau, F.; Giannias, B.; Berube, J.; Huang, Y.-H.; Rousseau, S.; Camilleri-Broet, S.; et al. Neutrophil Extracellular Trap-Associated CEACAM1 as a Putative Therapeutic Target to Prevent Metastatic Progression of Colon Carcinoma. J. Immunol. 2020, 204, 2285–2294. [Google Scholar] [CrossRef] [PubMed]
  123. Aldabbous, L.; Abdul-Salam, V.; McKinnon, T.; Duluc, L.; Pepke-Zaba, J.; Southwood, M.; Ainscough, A.J.; Hadinnapola, C.; Wilkins, M.R.; Toshner, M.; et al. Neutrophil Extracellular Traps Promote Angiogenesis: Evidence from Vascular Pathology in Pulmonary Hypertension. Arter. Thromb. Vasc. Biol. 2016, 36, 2078–2087. [Google Scholar] [CrossRef] [PubMed]
  124. Monti, M.; De Rosa, V.; Iommelli, F.; Carriero, M.V.; Terlizzi, C.; Camerlingo, R.; Belli, S.; Fonti, R.; Di Minno, G.; Del Vecchio, S. Neutrophil Extracellular Traps as an Adhesion Substrate for Different Tumor Cells Expressing RGD-Binding Integrins. Int. J. Mol. Sci. 2018, 19, 2350. [Google Scholar] [CrossRef] [PubMed]
  125. Pieterse, E.; Rother, N.; Garsen, M.; Hofstra, J.M.; Satchell, S.C.; Hoffmann, M.; Loeven, M.A.; Knaapen, H.K.; van der Heijden, O.W.; Berden, J.H.; et al. Neutrophil Extracellular Traps Drive Endothelial-to-Mesenchymal Transition. Arter. Thromb. Vasc. Biol. 2017, 37, 1371–1379. [Google Scholar] [CrossRef] [PubMed]
  126. Saffarzadeh, M.; Juenemann, C.; Queisser, M.A.; Lochnit, G.; Barreto, G.; Galuska, S.P.; Lohmeyer, J.; Preissner, K.T. Neutrophil extracellular traps directly induce epithelial and endothelial cell death: A predominant role of histones. PLoS ONE 2012, 7, e32366. [Google Scholar] [CrossRef] [PubMed]
  127. Schedel, F.; Mayer-Hain, S.; Pappelbaum, K.I.; Metze, D.; Stock, M.; Goerge, T.; Loser, K.; Sunderkötter, C.; Luger, T.A.; Weishaupt, C. Evidence and impact of neutrophil extracellular traps in malignant melanoma. Pigment. Cell Melanoma Res. 2020, 33, 63–73. [Google Scholar] [CrossRef]
  128. Mouchemore, K.A.; Anderson, R.L.; Hamilton, J.A. Neutrophils, G-CSF and their contribution to breast cancer metastasis. FEBS J. 2018, 285, 665–679. [Google Scholar] [CrossRef]
  129. Li, Y.; Yang, Y.; Gan, T.; Zhou, J.; Hu, F.; Hao, N.; Yuan, B.; Chen, Y.; Zhang, M. Extracellular RNAs from lung cancer cells activate epithelial cells and induce neutrophil extracellular traps. Int. J. Oncol. 2019, 55, 69–80. [Google Scholar] [CrossRef]
  130. Wang, T.; Zhou, Y.; Zhou, Z.; Zhang, P.; Yan, R.; Sun, L.; Ma, W.; Zhang, T.; Shen, S.; Liu, H.; et al. Secreted protease PRSS35 suppresses hepatocellular carcinoma by disabling CXCL2-mediated neutrophil extracellular traps. Nat. Commun. 2023, 14, 1513. [Google Scholar] [CrossRef]
  131. Tohme, S.; Yazdani, H.O.; Al-Khafaji, A.B.; Chidi, A.P.; Loughran, P.; Mowen, K.; Wang, Y.; Simmons, R.L.; Huang, H.; Tsung, A. Neutrophil Extracellular Traps Promote the Development and Progression of Liver Metastases after Surgical Stress. Cancer Res. 2016, 76, 1367–1380. [Google Scholar] [CrossRef] [PubMed]
  132. Zha, C.; Meng, X.; Li, L.; Mi, S.; Qian, D.; Li, Z.; Wu, P.; Hu, S.; Zhao, S.; Cai, J.; et al. Neutrophil extracellular traps mediate the crosstalk between glioma progression and the tumor microenvironment via the HMGB1/RAGE/IL-8 axis. Cancer Biol. Med. 2020, 17, 154–168. [Google Scholar] [CrossRef]
  133. Li, C.; Chen, T.; Liu, J.; Wang, Y.; Zhang, C.; Guo, L.; Shi, D.; Zhang, T.; Wang, X.; Li, J. FGF19-Induced Inflammatory CAF Promoted Neutrophil Extracellular Trap Formation in the Liver Metastasis of Colorectal Cancer. Adv. Sci. 2023, 10, e2302613. [Google Scholar] [CrossRef] [PubMed]
  134. Yao, L.; Sheng, X.; Dong, X.; Zhou, W.; Li, Y.; Ma, X.; Song, Y.; Dai, H.; Du, Y. Neutrophil extracellular traps mediate TLR9/Merlin axis to resist ferroptosis and promote triple negative breast cancer progression. Apoptosis 2023, 28, 1484–1495. [Google Scholar] [CrossRef] [PubMed]
  135. Canli, Ö.; Nicolas, A.M.; Gupta, J.; Finkelmeier, F.; Goncharova, O.; Pesic, M.; Neumann, T.; Horst, D.; Löwer, M.; Sahin, U.; et al. Myeloid Cell-Derived Reactive Oxygen Species Induce Epithelial Mutagenesis. Cancer Cell 2017, 32, 869–883.e5. [Google Scholar] [CrossRef] [PubMed]
  136. Zhong, J.; Li, Q.; Luo, H.; Holmdahl, R. Neutrophil-derived reactive oxygen species promote tumor colonization. Commun. Biol. 2021, 4, 865. [Google Scholar] [CrossRef] [PubMed]
  137. Rice, C.M.; Davies, L.C.; Subleski, J.J.; Maio, N.; Gonzalez-Cotto, M.; Andrews, C.; Patel, N.L.; Palmieri, E.M.; Weiss, J.M.; Lee, J.-M.; et al. Tumour-elicited neutrophils engage mitochondrial metabolism to circumvent nutrient limitations and maintain immune suppression. Nat. Commun. 2018, 9, 5099. [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. Pillay, J.; Kamp, V.M.; van Hoffen, E.; Visser, T.; Tak, T.; Lammers, J.; Ulfman, L.H.; Leenen, L.P.; Pickkers, P.; Koenderman, L. A subset of neutrophils in human systemic inflammation inhibits T cell responses through Mac-1. J. Clin. Invest. 2012, 122, 327–336. [Google Scholar] [CrossRef]
  140. Aarts, C.E.; Hiemstra, I.H.; Tool, A.T.; Van Den Berg, T.K.; Mul, E.; Van Bruggen, R.; Kuijpers, T.W. Neutrophils as Suppressors of T Cell Proliferation: Does Age Matter? Front. Immunol. 2019, 10, 2144. [Google Scholar] [CrossRef]
  141. Minns, D.; Smith, K.J.; Hardisty, G.; Rossi, A.G.; Gwyer Findlay, E. The Outcome of Neutrophil-T Cell Contact Differs Depending on Activation Status of Both Cell Types. Front. Immunol. 2021, 12, 633486. [Google Scholar] [CrossRef] [PubMed]
  142. Schmielau, J.; Finn, O.J. Activated granulocytes and granulocyte-derived hydrogen peroxide are the underlying mechanism of suppression of t-cell function in advanced cancer patients. Cancer Res. 2001, 61, 4756–4760. [Google Scholar] [PubMed]
  143. Li, P.; Lu, M.; Shi, J.; Hua, L.; Gong, Z.; Li, Q.; Shultz, L.D.; Ren, G. Dual roles of neutrophils in metastatic colonization are governed by the host NK cell status. Nat. Commun. 2020, 11, 4387. [Google Scholar] [CrossRef]
  144. Sandhu, J.K.; Privora, H.F.; Wenckebach, G.; Birnboim, H.C. Neutrophils, nitric oxide synthase, and mutations in the mutatect murine tumor model. Am. J. Pathol. 2000, 156, 509–518. [Google Scholar] [CrossRef] [PubMed]
  145. Behrens, L.M.; van Egmond, M.; van den Berg, T.K. Neutrophils as immune effector cells in antibody therapy in cancer. Immunol. Rev. 2023, 314, 280–301. [Google Scholar] [CrossRef] [PubMed]
  146. Treffers, L.W.; Ten Broeke, T.; Rösner, T.; Jansen, J.M.; van Houdt, M.; Kahle, S.; Schornagel, K.; Verkuijlen, P.J.; Prins, J.M.; Franke, K.; et al. IgA-Mediated Killing of Tumor Cells by Neutrophils Is Enhanced by CD47–SIRPα Checkpoint Inhibition. Cancer Immunol. Res. 2020, 8, 120–130. [Google Scholar] [CrossRef] [PubMed]
  147. Matlung, H.L.; Babes, L.; Zhao, X.W.; van Houdt, M.; Treffers, L.W.; van Rees, D.J.; Franke, K.; Schornagel, K.; Verkuijlen, P.; Janssen, H.; et al. Neutrophils Kill Antibody-Opsonized Cancer Cells by Trogoptosis. Cell Rep. 2018, 23, 3946–3959.e6. [Google Scholar] [CrossRef]
  148. Brandsma, A.M.; Bondza, S.; Evers, M.; Koutstaal, R.; Nederend, M.; Jansen, J.H.M.; Rösner, T.; Valerius, T.; Leusen, J.H.W.; Broeke, T.T. Potent Fc Receptor Signaling by IgA Leads to Superior Killing of Cancer Cells by Neutrophils Compared to IgG. Front. Immunol. 2019, 10, 704. [Google Scholar] [CrossRef]
  149. Metelitsa, L.S.; Gillies, S.D.; Super, M.; Shimada, H.; Reynolds, C.P.; Seeger, R.C. Antidisialoganglioside/granulocyte macrophage-colony-stimulating factor fusion protein facilitates neutrophil antibody-dependent cellular cytotoxicity and depends on FcgammaRII (CD32) and Mac-1 (CD11b/CD18) for enhanced effector cell adhesion and azurophil granule exocytosis. Blood 2002, 99, 4166–4173. [Google Scholar]
  150. Sarode, A.Y.; Jha, M.K.; Zutshi, S.; Ghosh, S.K.; Mahor, H.; Sarma, U.; Saha, B. Residue-Specific Message Encoding in CD40-Ligand. iScience 2020, 23, 101441. [Google Scholar] [CrossRef]
  151. Linde, I.L.; Prestwood, T.R.; Qiu, J.; Pilarowski, G.; Linde, M.H.; Zhang, X.; Shen, L.; Reticker-Flynn, N.E.; Chiu, D.K.-C.; Sheu, L.Y.; et al. Neutrophil-activating therapy for the treatment of cancer. Cancer Cell 2023, 41, 356–372.e10. [Google Scholar] [CrossRef] [PubMed]
  152. Nagata, S. Apoptosis by death factor. Cell 1997, 88, 355–365. [Google Scholar] [CrossRef] [PubMed]
  153. Serrao, K.L.; Fortenberry, J.D.; Owens, M.L.; Harris, F.L.; Brown, L.A. Neutrophils induce apoptosis of lung epithelial cells via release of soluble Fas ligand. Am. J. Physiol. Lung Cell Mol. Physiol. 2001, 280, L298–L305. [Google Scholar] [CrossRef] [PubMed]
  154. Koga, Y.; Matsuzaki, A.; Suminoe, A.; Hattori, H.; Hara, T. Neutrophil-derived TNF-related apoptosis-inducing ligand (TRAIL): A novel mechanism of antitumor effect by neutrophils. Cancer Res. 2004, 64, 1037–1043. [Google Scholar] [CrossRef] [PubMed]
  155. Bakema, J.E.; Ganzevles, S.H.; Fluitsma, D.M.; Schilham, M.W.; Beelen, R.H.J.; Valerius, T.; Lohse, S.; Glennie, M.J.; Medema, J.P.; van Egmond, M. Targeting FcαRI on polymorphonuclear cells induces tumor cell killing through autophagy. J. Immunol. 2011, 187, 726–732. [Google Scholar] [CrossRef] [PubMed]
  156. Joly, E.; Hudrisier, D. What is trogocytosis and what is its purpose? Nat. Immunol. 2003, 4, 815. [Google Scholar] [CrossRef] [PubMed]
  157. Valgardsdottir, R.; Cattaneo, I.; Klein, C.; Introna, M.; Figliuzzi, M.; Golay, J. Human neutrophils mediate trogocytosis rather than phagocytosis of CLL B cells opsonized with anti-CD20 antibodies. Blood 2017, 129, 2636–2644. [Google Scholar] [CrossRef]
  158. Rossi, E.A.; Goldenberg, D.M.; Michel, R.; Rossi, D.L.; Wallace, D.J.; Chang, C.-H. Trogocytosis of multiple B-cell surface markers by CD22 targeting with epratuzumab. Blood 2013, 122, 3020–3029. [Google Scholar] [CrossRef]
  159. Jones, J.D.; Hamilton, B.J.; Rigby, W.F.C. Rituximab mediates loss of CD19 on B cells in the absence of cell death. Arthritis Rheum. 2012, 64, 3111–3118. [Google Scholar] [CrossRef]
  160. Horner, H.; Frank, C.; Dechant, C.; Repp, R.; Glennie, M.; Herrmann, M.; Stockmeyer, B. Intimate cell conjugate formation and exchange of membrane lipids precede apoptosis induction in target cells during antibody-dependent, granulocyte-mediated cytotoxicity. J. Immunol. 2007, 179, 337–345. [Google Scholar] [CrossRef]
  161. Eichelberger, K.R.; Goldman, W.E. Manipulating neutrophil degranulation as a bacterial virulence strategy. PLoS Pathog. 2020, 16, e1009054. [Google Scholar] [CrossRef] [PubMed]
  162. Clark, R.A.; Olsson, I.; Klebanoff, S.J. Cytotoxicity for tumor cells of cationic proteins from human neutrophil granules. J. Cell Biol. 1976, 70, 719–723. [Google Scholar] [CrossRef] [PubMed]
  163. Lichtenstein, A. Mechanism of mammalian cell lysis mediated by peptide defensins. Evidence for an initial alteration of the plasma membrane. J. Clin. Invest. 1991, 88, 93–100. [Google Scholar] [CrossRef] [PubMed]
  164. Eruslanov, E.B.; Bhojnagarwala, P.S.; Quatromoni, J.G.; Stephen, T.L.; Ranganathan, A.; Deshpande, C.; Akimova, T.; Vachani, A.; Litzky, L.; Hancock, W.W.; et al. Tumor-associated neutrophils stimulate T cell responses in early-stage human lung cancer. J. Clin. Investig. 2014, 124, 5466–5480. [Google Scholar] [CrossRef] [PubMed]
  165. Lang, M.; Borgmann, M.; Oberhuber, G.; Evstatiev, R.; Jimenez, K.; Dammann, K.W.; Jambrich, M.; Khare, V.; Campregher, C.; Ristl, R.; et al. Thymoquinone attenuates tumor growth in ApcMin mice by interference with Wnt-signaling. Mol. Cancer 2013, 12, 41. [Google Scholar] [CrossRef] [PubMed]
  166. Qin, F.; Liu, X.; Chen, J.; Huang, S.; Wei, W.; Zou, Y.; Liu, X.; Deng, K.; Mo, S.; Chen, J.; et al. Anti-TGF-β attenuates tumor growth via polarization of tumor associated neutrophils towards an anti-tumor phenotype in colorectal cancer. J. Cancer 2020, 11, 2580–2592. [Google Scholar] [CrossRef] [PubMed]
  167. Yajuk, O.; Baron, M.; Toker, S.; Zelter, T.; Fainsod-Levi, T.; Granot, Z. The PD-L1/PD-1 Axis Blocks Neutrophil Cytotoxicity in Cancer. Cells 2021, 10, 1510. [Google Scholar] [CrossRef]
  168. 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]
  169. Glodde, N.; Bald, T.; van den Boorn-Konijnenberg, D.; Nakamura, K.; O’Donnell, J.S.; Szczepanski, S.; Brandes, M.; Eickhoff, S.; Das, I.; Shridhar, N.; et al. Reactive Neutrophil Responses Dependent on the Receptor Tyrosine Kinase c-MET Limit Cancer Immunotherapy. Immunity 2017, 47, 789–802.e9. [Google Scholar] [CrossRef]
  170. MET Promotes Antitumor Neutrophil Recruitment and Cytotoxicity. Cancer Discov. 2015, 5, 689. [CrossRef]
  171. Triner, D.; Devenport, S.N.; Ramakrishnan, S.K.; Ma, X.; Frieler, R.A.; Greenson, J.K.; Inohara, N.; Nunez, G.; Colacino, J.A.; Mortensen, R.M. Neutrophils Restrict Tumor-Associated Microbiota to Reduce Growth and Invasion of Colon Tumors in Mice. Gastroenterology 2019, 156, 1467–1482. [Google Scholar] [CrossRef] [PubMed]
  172. Yam, A.O.; Bailey, J.; Lin, F.; Jakovija, A.; Youlten, S.E.; Counoupas, C.; Gunzer, M.; Bald, T.; Woodruff, T.M.; Triccas, J.A.; et al. Neutrophil Conversion to a Tumor-Killing Phenotype Underpins Effective Microbial Therapy. Cancer Res. 2023, 83, 1315–1328. [Google Scholar] [CrossRef] [PubMed]
  173. Terasawa, M.; Nagata, K.; Kobayashi, Y. Neutrophils and monocytes transport tumor cell antigens from the peritoneal cavity to secondary lymphoid tissues. Biochem. Biophys. Res. Commun. 2008, 377, 589–594. [Google Scholar] [CrossRef] [PubMed]
  174. Lee, W.L.; Harrison, R.E.; Grinstein, S. Phagocytosis by neutrophils. Microbes Infect. 2003, 5, 1299–1306. [Google Scholar] [CrossRef] [PubMed]
  175. Meinderts, S.M.; Baker, G.; van Wijk, S.; Beuger, B.M.; Geissler, J.; Jansen, M.H.; Saris, A.; Brinke, A.T.; Kuijpers, T.W.; van den Berg, T.K.; et al. Neutrophils acquire antigen-presenting cell features after phagocytosis of IgG-opsonized erythrocytes. Blood Adv. 2019, 3, 1761–1773. [Google Scholar] [CrossRef] [PubMed]
  176. Reinisch, W.; Lichtenberger, C.; Steger, G.; Tillinger, W.; Scheiner, O.; Gangl, A.; Maurer, D.; Willheim, M. Donor dependent, interferon-γ induced HLA-DR expression on human neutrophils in vivo. Clin. Exp. Immunol. 2003, 133, 476–484. [Google Scholar] [CrossRef] [PubMed]
  177. Gosselin, E.J.; Wardwell, K.; Rigby, W.F.; Guyre, P.M. Induction of MHC class II on human polymorphonuclear neutrophils by granulocyte/macrophage colony-stimulating factor, IFN-gamma, and IL-3. J. Immunol. 1993, 151, 1482–1490. [Google Scholar] [CrossRef]
  178. Smith, W.; Guida, L.; Sun, Q.; Korpelainen, E.I.; van den Heuvel, C.; Gillis, D.; Hawrylowicz, C.M.; Vadas, M.A.; Lopez, A.F. Neutrophils activated by granulocyte-macrophage colony-stimulating factor express receptors for interleukin-3 which mediate class II expression. Blood 1995, 86, 3938–3944. [Google Scholar] [CrossRef]
  179. Vono, M.; Lin, A.; Norrby-Teglund, A.; Koup, R.A.; Liang, F.; Loré, K. Neutrophils acquire the capacity for antigen presentation to memory CD4+ T cells in vitro and ex vivo. Blood 2017, 129, 1991–2001. [Google Scholar] [CrossRef]
  180. Singel, K.L.; Segal, B.H. Neutrophils in the tumor microenvironment: Trying to heal the wound that cannot heal. Immunol. Rev. 2016, 273, 329–343. [Google Scholar] [CrossRef]
  181. Abi Abdallah, D.S.; Egan, C.E.; Butcher, B.A.; Denkers, E.Y. Mouse neutrophils are professional antigen-presenting cells programmed to instruct Th1 and Th17 T-cell differentiation. Int. Immunol. 2011, 23, 317–326. [Google Scholar] [CrossRef] [PubMed]
  182. Yang, C.-W.; Unanue, E.R. Neutrophils control the magnitude and spread of the immune response in a thromboxane A2-mediated process. J. Exp. Med. 2013, 210, 375–387. [Google Scholar] [CrossRef]
  183. Yang, C.-W.; Strong, B.S.I.; Miller, M.J.; Unanue, E.R. Neutrophils Influence the Level of Antigen Presentation during the Immune Response to Protein Antigens in Adjuvants. J. Immunol. 2010, 185, 2927–2934. [Google Scholar] [CrossRef] [PubMed]
  184. Mysore, V.; Cullere, X.; Mears, J.; Rosetti, F.; Okubo, K.; Liew, P.X.; Zhang, F.; Madera-Salcedo, I.; Rosenbauer, F.; Stone, R.M.; et al. FcγR engagement reprograms neutrophils into antigen cross-presenting cells that elicit acquired anti-tumor immunity. Nat. Commun. 2021, 12, 4791. [Google Scholar] [CrossRef] [PubMed]
  185. Guo, N.; Ni, K.; Luo, T.; Lan, G.; Arina, A.; Xu, Z.; Mao, J.; Weichselbaum, R.R.; Spiotto, M.; Lin, W. Reprogramming of Neutrophils as Non-canonical Antigen Presenting Cells by Radiotherapy–Radiodynamic Therapy to Facilitate Immune-Mediated Tumor Regression. ACS Nano 2021, 15, 17515–17527. [Google Scholar] [CrossRef] [PubMed]
  186. Lahoz-Beneytez, J.; Elemans, M.; Zhang, Y.; Ahmed, R.; Salam, A.; Block, M.; Niederalt, C.; Asquith, B.; Macallan, D. Human neutrophil kinetics: Modeling of stable isotope labeling data supports short blood neutrophil half-lives. Blood 2016, 127, 3431–3438. [Google Scholar] [CrossRef] [PubMed]
  187. Dominguez, G.A.; Condamine, T.; Mony, S.; Hashimoto, A.; Wang, F.; Liu, Q.; Forero, A.; Bendell, J.; Witt, R.; Hockstein, N.; et al. Selective Targeting of Myeloid-Derived Suppressor Cells in Cancer Patients Using DS-8273a, an Agonistic TRAIL-R2 Antibody. Clin. Cancer Res. 2017, 23, 2942–2950. [Google Scholar] [CrossRef]
  188. Isambert, N.; Hervieu, A.; Rébé, C.; Hennequin, A.; Borg, C.; Zanetta, S.; Chevriaux, A.; Richard, C.; Derangère, V.; Limagne, E.; et al. Fluorouracil and bevacizumab plus anakinra for patients with metastatic colorectal cancer refractory to standard therapies (IRAFU): A single-arm phase 2 study. Oncoimmunology 2018, 7, e1474319. [Google Scholar] [CrossRef]
  189. Bronte, V. From Oncogene Interference to Neutrophil Immune Modulation. Immunity 2017, 47, 613–615. [Google Scholar] [CrossRef]
  190. Ludford, K.; Ho, W.J.; Thomas, J.V.; Raghav, K.P.; Murphy, M.B.; Fleming, N.D.; Lee, M.S.; Smaglo, B.G.; You, Y.N.; Tillman, M.M.; et al. Neoadjuvant Pembrolizumab in Localized Microsatellite Instability High/Deficient Mismatch Repair Solid Tumors. J. Clin. Oncol. 2023, 41, 2181–2190. [Google Scholar] [CrossRef]
  191. Yan, M.; Zheng, M.; Niu, R.; Yang, X.; Tian, S.; Fan, L.; Li, Y.; Zhang, S. Roles of tumor-associated neutrophils in tumor metastasis and its clinical applications. Front. Cell Dev. Biol. 2022, 10, 938289. [Google Scholar] [CrossRef] [PubMed]
  192. Morris, J.C.; Tan, A.R.; Olencki, T.E.; Shapiro, G.I.; Dezube, B.J.; Reiss, M.; Hsu, F.J.; Berzofsky, J.A.; Lawrence, D.P. Phase I study of GC1008 (fresolimumab): A human anti-transforming growth factor-beta (TGFβ) monoclonal antibody in patients with advanced malignant melanoma or renal cell carcinoma. PLoS ONE 2014, 9, e90353. [Google Scholar] [CrossRef] [PubMed]
  193. Lacouture, M.E.; Morris, J.C.; Lawrence, D.P.; Tan, A.R.; Olencki, T.E.; Shapiro, G.I.; Dezube, B.J.; Berzofsky, J.A.; Hsu, F.J.; Guitart, J. Cutaneous keratoacanthomas/squamous cell carcinomas associated with neutralization of transforming growth factor β by the monoclonal antibody fresolimumab (GC1008). Cancer Immunol. Immunother. 2015, 64, 437–446. [Google Scholar] [CrossRef] [PubMed]
  194. Formenti, S.C.; Lee, P.; Adams, S.; Goldberg, J.D.; Li, X.; Xie, M.W.; Ratikan, J.A.; Felix, C.; Hwang, L.; Faull, K.F.; et al. Focal Irradiation and Systemic TGFβ Blockade in Metastatic Breast Cancer. Clin. Cancer Res. 2018, 24, 2493–2504. [Google Scholar] [CrossRef] [PubMed]
  195. Kelley, R.K.; Gane, E.; Assenat, E.; Siebler, J.; Galle, P.R.; Merle, P.; Hourmand, I.O.; Cleverly, A.; Zhao, Y.; Gueorguieva, I.; et al. A Phase 2 Study of Galunisertib (TGF-β1 Receptor Type I Inhibitor) and Sorafenib in Patients with Advanced Hepatocellular Carcinoma. Clin. Transl. Gastroenterol. 2019, 10, e00056. [Google Scholar] [CrossRef]
Figure 1. The multifaceted role of neutrophils in the tumor microenvironment: Neutrophils can act as both pro-tumorigenic and anti-tumorigenic, employing various functions depending on the disease stage and their location (as described in detail in the text). ROS-Reactive Oxygen Species; DNA-Deoxyribonucleic acid; IL1Ra-Interleukine-1 Receptor Alpha; NK cells-Natural Killer Cells; SIRPa- Signal Regulatory Protein alpha; MHCII- Major Histocompatibility complex II; PGE1-Prostaglandin E1; TRAIL/FasL-Tumor Necrosis Factor-Related Apoptosis Inducing Ligand/Fas Ligand; NO-Nitric Oxide; Arg- Arginase; PDL1-Programmed death-ligand 1; MMP- Matrix metalloproteinases; MPO-Myeloperoxidase; FcRs- Fc receptors. Created with BioRender.com (accessed on 25 January 2024).
Figure 1. The multifaceted role of neutrophils in the tumor microenvironment: Neutrophils can act as both pro-tumorigenic and anti-tumorigenic, employing various functions depending on the disease stage and their location (as described in detail in the text). ROS-Reactive Oxygen Species; DNA-Deoxyribonucleic acid; IL1Ra-Interleukine-1 Receptor Alpha; NK cells-Natural Killer Cells; SIRPa- Signal Regulatory Protein alpha; MHCII- Major Histocompatibility complex II; PGE1-Prostaglandin E1; TRAIL/FasL-Tumor Necrosis Factor-Related Apoptosis Inducing Ligand/Fas Ligand; NO-Nitric Oxide; Arg- Arginase; PDL1-Programmed death-ligand 1; MMP- Matrix metalloproteinases; MPO-Myeloperoxidase; FcRs- Fc receptors. Created with BioRender.com (accessed on 25 January 2024).
Ijms 25 02929 g001
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

Awasthi, D.; Sarode, A. Neutrophils at the Crossroads: Unraveling the Multifaceted Role in the Tumor Microenvironment. Int. J. Mol. Sci. 2024, 25, 2929. https://doi.org/10.3390/ijms25052929

AMA Style

Awasthi D, Sarode A. Neutrophils at the Crossroads: Unraveling the Multifaceted Role in the Tumor Microenvironment. International Journal of Molecular Sciences. 2024; 25(5):2929. https://doi.org/10.3390/ijms25052929

Chicago/Turabian Style

Awasthi, Deepika, and Aditya Sarode. 2024. "Neutrophils at the Crossroads: Unraveling the Multifaceted Role in the Tumor Microenvironment" International Journal of Molecular Sciences 25, no. 5: 2929. https://doi.org/10.3390/ijms25052929

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