Neutrophils in cancer development and progression: Roles, mechanisms, and implications (Review)

  • Authors:
    • Xu Zhang
    • Wen Zhang
    • Xiao Yuan
    • Min Fu
    • Hui Qian
    • Wenrong Xu
  • View Affiliations

  • Published online on: July 7, 2016     https://doi.org/10.3892/ijo.2016.3616
  • Pages: 857-867
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Neutrophils are predominant immune cells that protect the host from microbial infection. The roles of neutrophils in tumor have long been ignored due to their short life span and terminal differentiation phenotype. In recent years, emerging evidence indicates that neutrophils have phenotypic and functional plasticity. Neutrophils eliminate malignant cells by releasing the antimicrobial and cytotoxic contents in their granules or secreting immune mediators to recruit and activate other antitumor effector cells. On the contrary, tumor derived factors can convert neutrophils into a pro-tumor phenotype. Neutrophils have been shown to facilitate tumorigenesis, promote tumor growth and metastasis, stimulate tumor angiogenesis, and mediate immunosuppression. The number of neutrophils in blood and tumor tissues of cancer patients is associated with disease progression and patient outcome. In this review, we summarize the recent progress of neutrophils in the pathogenesis of cancer with an emphasis on neutrophil polarization. Better understanding of the mechanisms that regulate the dichotomy of neutrophils will not only shed light on their roles in cancer but also provide new approaches for cancer diagnosis and treatment.

1. Introduction

Neutrophils are innate immune cells that protect the host from infection by eliminating the invading pathogens. In recent years, these cells have been shown to play important roles in other pathological conditions including cancer. Neutrophils make up a significant portion of the inflammatory cell infiltrate in cancer, whereby they show high functional plasticity and display both antitumor and pro-tumor activities (1). The antitumor effects of neutrophils are related to their cytotoxicity and the regulation of antitumor immune responses, which has been denominated as N1 neutrophils. In addition, tumor derived signals can induce a pro-tumor phenotype in neutrophils, which supports tumor growth and metastasis (N2 neutrophils). N2 polarized neutrophils promote the proliferation, migration, and invasion of tumor cells, stimulate angiogenesis, as well as mediate immunosuppression (2,3). Moreover, increased number of neutrophils in blood and tumors has been linked to poor clinical outcome. Strategies designed to inhibit the pro-tumor activities of neutrophils have shown promising anticancer effects. In this review, we summarize the recent findings on the functional roles of neutrophils in cancer. We mainly focus on the molecular mechanisms that modulate the phenotypic and functional plasticity of neutrophils. The diagnostic value and therapeutic potential of neutrophils in cancer is also discussed.

2. The antitumor roles of neutrophils

The antitumor activities of neutrophils are supported by several lines of evidence. Neutrophils limit tumor growth and metastasis through distinct mechanisms including direct and antibody-dependent cytotoxic activity as well as the activation of other innate and adaptive immune cells such as T cells, B cells, natural killer (NK) cells, and dendritic cells (DCs).

3. The direct cytotoxicity of neutrophils

Neutrophils produce a number of antimicrobial mediators that have potential tumoricidal activity, including reactive oxygen species (ROS), myeloperoxidase (MPO), hydrogen peroxide (H2O2), and proteases. Neutrophils from healthy donors have potent cytotoxicity against tumor cells (4). The administration of neutrophils from healthy donors reduces experimental tumor growth and extends the survival of tumor-bearing animals (5). After stimulation with cytokines, neutrophils release ROS to trigger oxidative damage and consequent apoptotic cell death in melanoma cells (6). In addition, neutrophils could inhibit the metastatic potential of tumor cells. Granot and colleagues demonstrated that neutrophils generate H2O2 to suppress metastatic seeding of breast cancer cells in the lungs of mice (7), suggesting that neutrophils could prevent tumor metastasis via the generation of cytotoxic substances (8).

4. The antibody-dependent cell cytotoxicity of neutrophils

Neutrophils are critical effector cells that mediate the antitumor effects of mAb-mediated immunotherapy. Antibody-targeting cells could be destroyed by immune cells that express Fc receptors (FcR). Neutrophils express the family members of FcγR. The interactions between neutrophils and mAb through FcR induce the release of tumoricidal mediators (9,10). In several tumor models mAb-induced tumor reduction is abolished in mice with depleted neutrophils. In FcR-deficient mice, the transfer of normal neutrophils or transgenic expression of FcR restore the antitumor effects of mAb, suggesting that neutrophils are required for effective, mAb-induced cancer immunotherapy.

5. The recruitment and activation of innate and adaptive immune cells by neutrophils

In addition to direct and antibody-dependent cytotoxic effects on tumor cells, neutrophils could also recruit and activate immune cells to elicit antitumor immune responses (1115). Neutrophils release a wide array of factors including cytokines, chemokines, and proteases that have promoting roles in the proliferation and cytokine production of T cells. Neutrophils isolated from the surgically resected human lung cancer tissues could stimulate T cell proliferation and IFN-γ release (16). Neutrophils could efficiently process and present antigens to directly stimulate immune response. Moreover, TLR-stimulated neutrophils induce enhanced cytotoxicity and cytokine production in NK cells and trigger the maturation of dendritic cells, promoting T cell proliferation and IFN-γ production (17).

6. The pro-tumor roles of neutrophils

There is mounting evidence showing that neutrophils are critically involved in the development and progression of cancer (18). Neutrophils play important roles in neoplastic transformation, tumor growth and metastasis, angiogenesis, and the modulation of immunosuppression (Fig. 1).

7. Neutrophils and tumorigenesis

The accumulation of genetic instability is associated with increased cancer risk. Neutrophils release genotoxic substances to inflict DNA damage on epithelial cells and initiate carcinogenic response (1922). Exposure to activated neutrophils increases the number of replication errors in colon epithelial cells (23). In colitis-associated colon cancer (CAC) mouse model, depletion of neutrophils markedly reduces the number and size of tumors, indicating a crucial role for neutrophils in the initiation and progression of CAC (2426). Lakritz et al demonstrate that neutrophils are critical for mammary tumor development because systemic depletion of neutrophils entirely inhibits tumorigenesis (27). Wilson and colleagues demonstrated that neutrophils stimulate the production of ROS and telomere DNA damage in hepatocytes and promote diethylnitrosamine (DEN)-induced hepatocellular carcinoma (HCC) (28). Yan et al further demonstrated the promoting role of neutrophils in hepatocarcinogenesis by using a zebrafish model (29).

8. Neutrophils and tumor growth

Neutrophils generate and release a wide spectrum of factors to support tumor cell growth in vitro and in vivo (30). Neutrophil elastase (NE) was able to enter into tumor cells to degrade insulin receptor substrate-1 (IRS-1), resulting in increased interaction between PI3K and PDGFR and accelerated tumor cell proliferation (31,32). Neutrophils could promote tumor cell proliferation through COX-2-mediated prostaglandin E2 (PGE2) synthesis (33,34). Antonio et al suggested that acute wound (such as tumor biopsy) induces rapid recruitment of neutrophils to interact with nearby neoplastic cells, leading to increased proliferation of the neoplastic cells through PGE2 (35). Neutrophils enhanced the proliferation of glioblastoma-initiating cells through the upregulation of S100A4 expression (36). Neutrophils could promote the proliferation of renal cell carcinoma (RCC) cells via modulating androgen receptor (AR)/c-Myc signals (37). Moreover, neutrophils from B-cell lymphoma patients induce stromal cell activation to promote the growth of germinal center B-cell lymphoma cells (38). Neutrophils were able to promote multiple myeloma (MM) survival from doxorubicin and melphalan by secretion of soluble factors (39).

9. Neutrophils and tumor metastasis

Neutrophils play a key role in cancer metastasis (40). Neutrophils could promote tumor metastasis by increasing the migratory and invasive potential of tumor cells, degrading extracellular matrix, and promoting the colonization of tumor cells (4143).

Neutrophils recruited by LPS-induced inflammation could release proteinases such as catheptin G and elastase to degrade thrombospondin-1 (Tsp-1) and facilitate lung metastasis (44). When stimulated with GM-CSF from breast cancer cells, neutrophils release a high level of oncostatin M (OSM), which in turn promotes the detachment of breast cancer cells (45). Wu et al demonstrate that hyaluronan (HA) from tumor cells activates neutrophils, which in turn effectively enhances the motility of tumor cells via a cell contact-dependent mechanism (46). Macrophage migration inhibitory factor (MIF) from human head and neck squamous cell carcinoma (HNSCC) cells could activate neutrophils, which in turn enhances the migratory properties of HNSCC cells (47). Moreover, G-CSF from breast cancer cells expand and mobilize neutrophils to release of Bv8, resulting in the promotion of metastasis (48).

Neutrophils could induce epithelial-to-mesenchymal transition (EMT) in tumor cells, which significantly increases the migratory and invasive capacity of tumor cells (4952). Neutrophils increase bladder cancer cell invasion through the modulation of androgen receptor (AR)/MMP13 signals (53). In addition, neutrophils could promote renal cell carcinoma cell migration and invasion via the activation of VEGFa/HIF2α and estrogen receptor β signals (54). Moreover, neutrophils could also diminish immune protection to promote metastasis. Coffelt et al demonstrate that gamma delta (γδ) T cell-derived IL-17 induce G-CSF-dependent expansion and activation of neutrophils, which inhibits cytotoxic CD8+ T lymphocytes and helps establish metastases (55).

Neutrophils may serve as a carrier to assist tumor cell extravasation. Tumor-elicited neutrophils bind to tumor cells and facilitate tumor cell migration, which is dependent on the expression of intercellular adhesion molecule-1 (ICAM-1) on tumor cells and CD11b on neutrophils (56,57). In vivo, neutrophils regulate lung metastasis through physical interaction and anchoring of circulating tumor cells to endothelium (58). Neutrophils promote cancer cell adhesion within liver sinusoids, however, neutrophil depletion impairs the formation of liver metastasis (59,60).

10. Neutrophils and tumor angiogenesis

Neutrophils synthesize and release a number of molecules to activate endothelial cells and induce angiogenesis (61,62). Shojaei et al suggest that tumor derived G-CSF upregulates Bv8 expression, which mobilizes neutrophils to promote angiogenesis (63). MMP-9 is implicated in VEGF activation to induce and maintain angiogenesis. Neutrophils are found to be the major sources of MMP-9 (6466). Human neutrophils uniquely release TIMP-free MMP-9 to provide a potent stimulator of angiogenesis (67). Tumor infiltrating neutrophil-derived MMP-9 coordinately regulate tumor angiogenesis and tumor cell intravasation. Specific inhibition of neutrophil accumulation results in the coordinated inhibition of tumor angiogenesis and intravasation (68).

11. Neutrophils and tumor immunosuppression

Tumor-elicited neutrophils could inhibit the proliferation of T cells through the release of arginase 1 (ARG1) and the modulation of PD-L1/PD-1 signaling (6972). Neutrophils isolated from the circulation of tumor-bearing mice contribute to the survival of tumor cells by suppressing peripheral leukocyte activation (73). A subset of neutrophils with low density is enriched in the peripheral blood of cancer patients and display immature phenotype similar to that of MDSCs (74). Fridlender and colleagues have compared tumor-associated neutrophils (TANs) with granulocytic myeloid-derived suppressor cells (G-MDSCs) by using transcriptomic analysis and found that the two cell populations are significantly different in their mRNA profiles, pointing out the differences between TANs and MDSCs (75). Hypoxia within the primary tumor sites induce increased infiltration of immunosuppressive neutrophils into the lung, where these cells compromise NK cell cytotoxicity, resulting in a reduced antitumor response that allows metastasis formation (76). Neutrophils inhibit NK cell function to increase the intraluminal survival of tumor cells, facilitating tumor cell extravasation and metastatic dissemination (77). In addition, neutrophils isolated from murine tumor tissues secrete significant amounts of CCL17 to progressively attract Tregs during tumor development (78). CCL17 secretion is relevant to the number of tumor infiltrating neutrophils in human lung cancer patients, suggesting that neutrophils may suppress antitumor immunity and promote tumor growth by regulating Tregs. Zhou et al demonstrated that TANs recruit macrophages and Treg cells to promote HCC growth, progression, and resistance to sorafenib (79). The expansion of myeloid cells contributes to tumor progression. Using a multistage mouse model of breast cancer, Casbon et al demonstrate that the invasive breast cancer reprograms early differentiation of myeloid cells in the bone marrow to generate immunosuppressive neutrophils (80).

12. Neutrophil extracellular traps in cancer

Neutrophil extracellular traps (NETs) is a web-like structure to trap and kill invading microorganisms (81). The contribution of NETs to tumor has recently been demonstrated (8284). Cancer-associated thrombosis is linked to a poor prognosis and represents the second-leading cause of death in cancer patients. Using a murine model of chronic myelogenous leukemia, Demers et al demonstrated that cancers predispose the release of NETs to cause cancer-associated thrombosis (85,86). Cools-Lartigue et al have shown that circulating tumor cells are trapped within NETs in vitro under both static and dynamic conditions. In a murine model of lung cancer, deposition of NETs and consequent trapping of circulating lung carcinoma cells are associated with increased hepatic metastasis (87). Neutrophils isolated from mouse models of pancreatic ductal adenocarcinoma (PDA) have shown an increased ability to form NETs (88). Guglietta et al showed that increased circulating lipopolysaccharide induces upregulation of complement C3a receptor on neutrophils and activation of the complement cascade, which leads to NETosis and N2 polarization of neutrophils, inducing coagulation and promoting spontaneous intestinal tumorigenesis (89). Moreover, neutrophil extracellular traps have been shown to promote the development and progression of liver metastases after surgical stress (90).

13. The recruitment, expansion, and polarization of neutrophils in cancer

The origin of the infiltrating neutrophils in tumor has not been well characterized. Cortez-Retamozo et al demonstrated that the spleen is an important origin of tumor associated neutrophils. The precursors of neutrophils relocate from the spleen to the tumor stroma during tumor progression. Removal of the spleen reduces the number of the infiltrating neutrophils and delays tumor growth (91). A large number of molecules from tumor cells have been shown to recruit neutrophils. IL-8 is one of the potent neutrophil chemoattractants. Tumor cells with IL-8 overexpression recruit more neutrophils and display increased metastatic potential (92). IL-17 recruits blood neutrophils into the peritumoral stroma of hepatocellular carcinoma by inducing expression of chemokines in epithelial cells (66). Wu et al also suggest that tumor-infiltrating DCs induce the activation of IL-17 producing γδT cells to promote the accumulation and expansion of immunosuppressive neutrophils in colon cancer (93).

Tumor-derived oxysterols could recruit neutrophils to favor tumor growth by promoting angiogenesis and immunosuppression (94,95). CXCL5 has a direct chemoattractant effect on neutrophils. CXCL5 overexpression is positively correlated with neutrophil infiltration in hepatocellular carcinoma and intrahepatic cholangiocarcinoma patients (96,97). UV irradiation-damaged epidermal keratinocytes release high mobility group box 1 (HMGB1) to recruit and activate neutrophils by interacting with toll-like receptor 4 (TLR4), which stimulates angiogenesis and promotes the ability of melanoma cells to metastasize (98). Leukotriene B4 (LTB4), an inflammation mediator, induces the recruitment of neutrophils via interaction with BLT1 on neutrophils (30). Neutrophils from HNSCC patients display a significantly reduced apoptosis compared to those from healthy donors, which may be associated with the secretion of MIF by HNSCC cells (99). Hypoxia induces an HIF-1α-dependent activation of NF-κB to inhibit neutrophil apoptosis (100). Li et al demonstrated that the prolonged survival of neutrophils in tumor is associated with increased autophagy. Neutrophils in HCC intratumoral regions undergo increased autophagy and display long-lived phenotypes and sustained production of pro-metastatic factors (101). IFN-β negatively regulates the survival and recruitment of neutrophils. In the absence of endogenous IFN-β the life span of neutrophils from blood and tumors of IFN-β deficient mice is remarkably prolonged (102,103). On the contrary, MET is required for the recruitment of antitumor neutrophils (104). Met deletion in mouse neutrophils enhances tumor growth and metastasis.

Tumor-derived factors could modify the phenotype and function of myeloid cells (105,106). Neutrophils are polarized to N1 and N2 phenotypes in cancer. In general, the N1 and N2 polarized neutrophils could be distinguished based on their phenotype and function. The N1 polarized neutrophils are short-lived cells with mature phenotype and display high cytotoxicity and immunostimulating activity. The N2 polarized neutrophils are long-lived cells with immature phenotype and show low cytotoxicity but high pro-angiogenic, pro-metastatic, and immunosuppressive activities.

The potent drivers of neutrophil polarization have recently been demonstrated. Inhibition of TGF-β increases the expression of chemokines that recruit neutrophils, resulting in an influx of neutrophils that has strong cytotoxic activity to tumor cells. Following TGF-β blockade, depletion of these neutrophils significantly attenuates antitumor effects of treatment and reduces CD8+ T cell activation. In contrast, in control tumors, neutrophil depletion decreases tumor growth and results in more activated CD8+ T cells within tumor, suggesting that tumor associated neutrophils are driven by TGF-β to acquire N2 protumoral phenotype. In contrast, TGF-β inhibition induces an antitumor N1 phenotype (107). The anti- and pro-tumor functions of neutrophils imply its diversity and plasticity. Sagiv et al have identified a heterogeneous subset of low density neutrophils (LDNs) that progressively accumulate in tumors. LDNs consist of both immature MDSCs and mature cells that are derived from HDNs in a TGF-β-dependent mechanism (108). The plasticity of neutrophils has been determined in mouse tumor models at different time points during tumor progression. Neutrophils are mainly located at the peritumoral tissues at early stage of tumor development while these cells are found scattered in tumor cells at later stage. Neutrophils isolated from tumors at early stage are more cytotoxic toward tumor cells and produce higher levels of NO and H2O2. In established tumors, these functions are decreased and these cells acquire a more protumorigenic phenotype, suggesting the critical role of tumor niche in modulating neutrophil phenotype and function. In line with this phenotype, only depletion of neutrophils at later stage of tumor development inhibits tumor growth, indicating the functional changes in neutrophils with tumor progression (109).

In the absence of endogenous IFN-β, mice develop a fast-growing tumor accompanied with increased infiltration of neutrophils which produce a large amount of VEGF and MMP-9 to promote tumor angiogenesis and metastasis. In vitro treatment with recombinant IFN-β inhibit the activation of STAT3 pathway and the upregulation of VEGF and MMP-9 genes in tumor infiltrating neutrophils. In addition, the transplantation of neutrophils from control mice into IFN-β-deficient mice retards tumor growth, suggesting that IFN-β may be a factor that maintain the N1 polarization of neutrophils. The conversion of neutrophil phenotype and function may occur in the bone marrow of tumor-bearing mice (110).

In addition to tumor cells, the microenvironmental cells also participate in the regulation of neutrophil biology in cancer. Tumor-resident mesenchymal stem cell (MSC)-derived IL-6 induced N2 polarized activation of neutrophils (111). Intriguingly, Hu et al demonstrated that TNFα-primed mouse MSCs could program neutrophils into an immunosuppressive and tumor-promoting phenotype (112). Moreover, in response to tumor derived IL-1β signal, tumor infiltrating γδ T cells release IL-17 to recruit, expand, and activate neutrophils to promote cancer metastasis. Taken together, these findings indicate that neutrophils are polarized during tumor progression by the signals from tumor milieu (Table I).

Table I

Factors that mediate neutrophil recruitment, expansion, polarization, and pro-tumoral function.

Table I

Factors that mediate neutrophil recruitment, expansion, polarization, and pro-tumoral function.

FactorsCancer typeFunctionSpeciesRefs.
CXCL5HCCRecruit neutrophils to promote cancer growth and metastasisHuman, mouse(96,136)
IL-17Breast cancer, HCCRecruit neutrophils to promote tumor growth and metastasisMouse(55,66,137)
HMGB1MelanomaRecruit neutrophils to promote tumor angiogenesis and metastasisMouse(98)
OxysterolLung cancerRecruit neutrophils to favor tumor growthMouse(94)
GM-CSFBreast cancerRecruit neutrophils to promote tumor angiogenesis and metastasisHuman(45)
HGFLung cancerRecruit neutrophils to promote tumor metastasisHuman, mouse(138)
IL-8Lung cancer, skin cancerRecruit neutrophils to promote tumor initiation and progressionHuman, zebrafish(51,92
G-CSFBreast cancerRecruit and expand neutrophils to promote tumor growth and metastasisMouse(48,80)
HAHCCRecruit neutrophils to promote tumor angiogenesis and metastasisHuman(46)
IL-6HCC, gastric cancerRecruit neutrophils to promote tumor growth and metastasisHuman, mouse(110,111)
CXCL2Colon cancerRecruit neutrophils to promote tumor growth and metastasisMouse(24)
TGF-βLung cancerPolarize neutrophils to an N2 phenotype to promote tumor growthMouse(107)
MIFHead and neck cancerRecruit neutrophils to promote tumor cell migrationHuman(47)
LTB4Lung cancerRecruit neutrophils to promote tumor growthMouse(30)
a2NTDBreast cancerRecruit neutrophils to promote tumor cell invasionHuman(139)
PGE2Lung cancer, skin cancerPromote tumor cell proliferationHuman, zebrafish(33,35)
NELung cancer, pancreatic cancerPromote tumor cell proliferation and tumor cell dyshesionMouse(31,49)
OSMBreast cancerPromote tumor angiogenesis and metastasisHuman(45)
Arg-1Lung cancerPromote tumor immunosuppressionHuman(70)
CCL17Lung cancer, HCCPromote tumor immunosuppressionMouse(78,79)
PD-L1HCCPromote tumor immunosuppressionHuman(71)
NETCML, lung cancerPromote tumor-associated thrombosis and tumor metastasisHuman(85,87)
MMP-9HCC, lung cancer, pancreatic cancerPromotes tumor angiogenesisHuman, mouse(64,65)
IL-1βColon cancerPromote tumor initiation and progressionMouse(25)

14. Targeting neutrophils for cancer diagnosis and therapy

Cancer-related inflammation plays a key role in tumor progression. The increased neutrophil infiltration in tumor is associated with poor outcome in renal cell carcinoma (113), head and neck squamous cell carcinoma (114), melanoma (115), lung carcinoma (116,117), colorectal carcinoma (118), gastric carcinoma (119), cholangiocarcinoma (97), hepatocellular carcinoma (120), tongue squamous cell carcinoma (121), and esophageal squamous cell carcinoma (122,123). High intratumoral neutrophil is positively correlated with lymph node metastasis, tumor grade, and tumor stage. High densities of neutrophils in tumor are identified as an independent risk factor for poor prognosis (124). In addition, a high neutrophil-to-lymphocyte ratio (NLR) has also been suggested as a poor prognostic indicator in cancer (125). Moreover, the high numbers of neutrophils and NLR in cancer patients are associated with poor response to chemotherapy and immunotherapy (126).

The idea of targeting neutrophils represents a new approach for cancer therapy (127). Several strategies have been proposed to inhibit their recruitment, interfere with their survival, or reprogram them into N1 antitumor phenotype (Fig. 2). ‘Reeducation’ to activate the antitumor potential of cells or elimination of tumor promoting cells is a new strategy undergoing preclinical and clinical evaluation. Since tumor derived factors contribute to phenotypic and functional plasticity of neutrophils in cancer, modulation of tumor milieu can lead to reeducation of neutrophils. The conversion of pro-tumor activity of neutrophils into antitumor potential with appropriate stimulation and modulation provides new opportunities for cancer therapy. Sun et al have recently shown that priming with TNF-α and IFN-γ could convert the potential of neutrophils from tumor-promoting to tumor-suppressing through the activation of NK cells (128). TGF-β signaling regulates neutrophil N2 polarization. Depletion of the receptor for TGF-β decreases the production of arginase 1 and iNOS in neutrophils, which in turn increases IFN-γ expression in CD8+ T cells and inhibits tumor metastasis (129). IFN-β regulates the N1 polarization of neutrophils. In mice, the treatment with low dose of IFN-β induces antitumor activation of neutrophils (130). The chemokine receptor CXCR2 is a key mediator of neutrophil recruitment. CXCR2 inhibitor attenuates neutrophil recruitment and profoundly suppresses tumor growth (131,132). Moreover, pharmacological inhibition of ALOX5, a leukotriene-generating enzyme, inhibits the recruitment of pro-metastatic neutrophils and reduces lung metastasis (133). A recent study from Shrestha et al indicates that the inhibitors for angiotensin converting enzyme (ACEis) and the antagonists for angiotensin II type 1 receptor (AGTR1) could attenuate tumor growth via the polarization of neutrophils toward an antitumoral phenotype (134).

15. Concluding remarks

The roles of neutrophils in the pathogenesis of cancer have recently become an intense research area. Neutrophils have both pro-tumor and antitumor activities. Neutrophils are frequently recruited to local tumor sites, whereby neutrophils can be polarized towards distinct phenotypes by tumor derived signals. In turn, neutrophils suppress or promote tumor development and progression by cell contact-dependent mechanism or secretion of soluble factors. Herein, we summarize the roles of neutrophils in cancer and their potential as cancer diagnosis biomarker and therapy target. Although early studies indicate that neutrophils have direct cytotoxicity against tumor cells and regulate the functions of innate and adaptive immune cells, more recent reports have shown that neutrophils promote tumor development and progression by enhancing tumor cell growth and metastasis, stimulating tumor angiogenesis, and mediating immunosuppression. Previous studies have mainly focused on experimental animal models of cancer (135), however, more studies are needed to elucidate the cellular and molecular mechanisms that modulate the phenotype and function of neutrophils in human tumor, such as recruitment to the tumor site, prolonged survival and enhanced release of tumor-promoting factors. In addition, further studies are needed to elucidate the relationship between heterogeneous neutrophil subsets. Moreover, novel strategies to reeducate the tumor-promoting neutrophils to activate the host’s innate and adaptive immune responses will provide new approaches for tumor therapy.

Acknowledgements

This study was supported by the National Natural Science Foundation of China (81201660), the Natural Science Foundation of the Jiangsu Province (BK20141303), the Key Research and Development Project of Zhenjiang (SH2015034), the Jiangsu Key Laboratory of Medical Science and Laboratory Medicine Project (JSKLM-2014-006), Jiangsu Province’s Qing Lan project, Foundation for Young Academic Leader of Jiangsu University, Starting Foundation for Senior Talents of Jiangsu University (13JDG086).

Abbreviations:

ACEis

angiotensin converting enzyme inhibitors

AGTR1

angiotensin II type 1 receptor antagonist

ALOX5

arachidonic acid 5-lipoxygenase

ARG1

arginase 1

AR

androgen receptor

BLT1

leukotriene B4 receptor-1

CAC

colitis-associated colon cancer

CCL

chemokine (C-C motif) ligand

CS

crystalline silica

CXCR

chemokine (C-X-C motif) receptor

DC

dendritic cells

DEN

diethylnitrosamine

EMT

epithelial-to-mesenchymal transition

ERβ

estrogen receptor β

FcR

Fc receptors

G-CSF

granulocyte colony-stimulating factor

G-MDSCs

granulocytic myeloid-derived suppressor cells

HA

hyaluronan

HCC

hepatocellular carcinoma

HGF

hepatocyte growth factor

HIF

hypoxia-inducible factor

HMGB1

high mobility group box-1 protein

HNSCC

head and neck squamous cell carcinoma

H2O2

hydrogen peroxide

HOCl

oxidanthypochlorous acid

ICAM-1

intercellular adhesion molecule-1

IFN

interferon

IL

interleukin

IRS-1

insulin receptor substrate-1

LDNs

low density neutrophils

LTB4

leukotriene B4

MDSCs

myeloid derived suppressor cells

MIF

macrophage migration inhibitory factor

MM

multiple myeloma

MMP-9

matrix metalloproteinase 9

MPO

myeloperoxidase

MSCs

mesenchymal stem cells

NE

neutrophil elastase

NETs

neutrophil extracellular traps

NLR

neutrophil/lymphocyte ratio

NK

natural killer

NO

nitric oxide

NSCLC

non-small cell lung cancer

OSCC

oral squamous cell carcinoma

OSM

oncostatin M

PDA

pancreatic ductal adenocarcinoma

PD-L1

programmed death-ligand 1

PGE2

prostaglandin E2

RAGE

receptor for advanced glycation end products

RCC

renal cell carcinoma

ROS

reactive oxygen species

TAN

tumor-associated neutrophils

TGF

transforming growth factor

TLR

Toll-like receptor

Tsp-1

thrombospondin-1

VEGF

vascular endothelial growth factor

References

1 

Piccard H, Muschel RJ and Opdenakker G: On the dual roles and polarized phenotypes of neutrophils in tumor development and progression. Crit Rev Oncol Hematol. 82:296–309. 2012. View Article : Google Scholar

2 

Brandau S, Dumitru CA and Lang S: Protumor and antitumor functions of neutrophil granulocytes. Semin Immunopathol. 35:163–176. 2013. View Article : Google Scholar

3 

Dumitru CA, Lang S and Brandau S: Modulation of neutrophil granulocytes in the tumor microenvironment: Mechanisms and consequences for tumor progression. Semin Cancer Biol. 23:141–148. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Yan J, Kloecker G, Fleming C, Bousamra M II, Hansen R, Hu X, Ding C, Cai Y, Xiang D, Donninger H, et al: Human polymorphonuclear neutrophils specifically recognize and kill cancerous cells. OncoImmunology. 3:e9501632014. View Article : Google Scholar

5 

Jaganjac M, Poljak-Blazi M, Kirac I, Borovic S, Joerg Schaur R and Zarkovic N: Granulocytes as effective anticancer agent in experimental solid tumor models. Immunobiology. 215:1015–1020. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Dissemond J, Weimann TK, Schneider LA, Schneeberger A, Scharffetter-Kochanek K, Goos M and Wagner SN: Activated neutrophils exert antitumor activity against human melanoma cells: Reactive oxygen species-induced mechanisms and their modulation by granulocyte-macrophage-colony-stimulating factor. J Invest Dermatol. 121:936–938. 2003. View Article : Google Scholar : PubMed/NCBI

7 

Granot Z, Henke E, Comen EA, King TA, Norton L and Benezra R: Tumor entrained neutrophils inhibit seeding in the premetastatic lung. Cancer Cell. 20:300–314. 2011. View Article : Google Scholar : PubMed/NCBI

8 

López-Lago MA, Posner S, Thodima VJ, Molina AM, Motzer RJ and Chaganti RS: Neutrophil chemokines secreted by tumor cells mount a lung antimetastatic response during renal cell carcinoma progression. Oncogene. 32:1752–1760. 2013. View Article : Google Scholar

9 

Stockmeyer B, Beyer T, Neuhuber W, Repp R, Kalden JR, Valerius T and Herrmann M: Polymorphonuclear granulocytes induce antibody-dependent apoptosis in human breast cancer cells. J Immunol. 171:5124–5129. 2003. View Article : Google Scholar : PubMed/NCBI

10 

Albanesi M, Mancardi DA, Jönsson F, Iannascoli B, Fiette L, Di Santo JP, Lowell CA and Bruhns P: Neutrophils mediate antibody-induced antitumor effects in mice. Blood. 122:3160–3164. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Mayadas TN, Cullere X and Lowell CA: The multifaceted functions of neutrophils. Annu Rev Pathol. 9:181–218. 2014. View Article : Google Scholar :

12 

Amulic B, Cazalet C, Hayes GL, Metzler KD and Zychlinsky A: Neutrophil function: From mechanisms to disease. Annu Rev Immunol. 30:459–489. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Kolaczkowska E and Kubes P: Neutrophil recruitment and function in health and inflammation. Nat Rev Immunol. 13:159–175. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Scapini P and Cassatella MA: Social networking of human neutrophils within the immune system. Blood. 124:710–719. 2014. View Article : Google Scholar : PubMed/NCBI

15 

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

16 

Eruslanov EB, Bhojnagarwala PS, Quatromoni JG, Stephen TL, Ranganathan A, Deshpande C, Akimova T, Vachani A, Litzky L, Hancock WW, et al: Tumor-associated neutrophils stimulate T cell responses in early-stage human lung cancer. J Clin Invest. 124:5466–5480. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Riise RE, Bernson E, Aurelius J, Martner A, Pesce S, Della Chiesa M, Marcenaro E, Bylund J, Hellstrand K, Moretta L, et al: TLR-stimulated neutrophils instruct NK cells to trigger dendritic cell maturation and promote adaptive T cell responses. J Immunol. 195:1121–1128. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Powell DR and Huttenlocher A: Neutrophils in the tumor micro-environment. Trends Immunol. 37:41–52. 2016. View Article : Google Scholar

19 

Haqqani AS, Sandhu JK and Birnboim HC: Expression of inter-leukin-8 promotes neutrophil infiltration and genetic instability in mutatect tumors. Neoplasia. 2:561–568. 2000. View Article : Google Scholar

20 

Sandhu JK, Privora HF, Wenckebach G and Birnboim HC: Neutrophils, nitric oxide synthase, and mutations in the mutatect murine tumor model. Am J Pathol. 156:509–518. 2000. View Article : Google Scholar : PubMed/NCBI

21 

Knaapen AM, Güngör N, Schins RP, Borm PJ and Van Schooten FJ: Neutrophils and respiratory tract DNA damage and mutagenesis: A review. Mutagenesis. 21:225–236. 2006. View Article : Google Scholar : PubMed/NCBI

22 

Güngör N, Knaapen AM, Munnia A, Peluso M, Haenen GR, Chiu RK, Godschalk RW and van Schooten FJ: Genotoxic effects of neutrophils and hypochlorous acid. Mutagenesis. 25:149–154. 2010. View Article : Google Scholar

23 

Campregher C, Luciani MG and Gasche C: Activated neutrophils induce an hMSH2-dependent G2/M checkpoint arrest and replication errors at a (CA)13-repeat in colon epithelial cells. Gut. 57:780–787. 2008. View Article : Google Scholar : PubMed/NCBI

24 

Shang K, Bai YP, Wang C, Wang Z, Gu HY, Du X, Zhou XY, Zheng CL, Chi YY, Mukaida N, et al: Crucial involvement of tumor-associated neutrophils in the regulation of chronic colitis-associated carcinogenesis in mice. PLoS One. 7:e518482012. View Article : Google Scholar : PubMed/NCBI

25 

Wang Y, Wang K, Han GC, Wang RX, Xiao H, Hou CM, Guo RF, Dou Y, Shen BF, Li Y, et al: Neutrophil infiltration favors colitis-associated tumorigenesis by activating the interleukin-1 (IL-1)/ IL-6 axis. Mucosal Immunol. 7:1106–1115. 2014. View Article : Google Scholar : PubMed/NCBI

26 

Ning C, Li YY, Wang Y, Han GC, Wang RX, Xiao H, Li XY, Hou CM, Ma YF, Sheng DS, et al: Complement activation promotes colitis-associated carcinogenesis through activating intestinal IL-1β/IL-17A axis. Mucosal Immunol. 8:1275–1284. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Lakritz JR, Poutahidis T, Mirabal S, Varian BJ, Levkovich T, Ibrahim YM, Ward JM, Teng EC, Fisher B, Parry N, et al: Gut bacteria require neutrophils to promote mammary tumorigenesis. Oncotarget. 6:9387–9396. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Wilson CL, Jurk D, Fullard N, Banks P, Page A, Luli S, Elsharkawy AM, Gieling RG, Chakraborty JB, Fox C, et al: NFκB1 is a suppressor of neutrophil-driven hepatocellular carcinoma. Nat Commun. 6:68182015. View Article : Google Scholar

29 

Yan C, Huo X, Wang S, Feng Y and Gong Z: Stimulation of hepatocarcinogenesis by neutrophils upon induction of oncogenic kras expression in transgenic zebrafish. J Hepatol. 63:420–428. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Satpathy SR, Jala VR, Bodduluri SR, Krishnan E, Hegde B, Hoyle GW, Fraig M, Luster AD and Haribabu B: Crystalline silica-induced leukotriene B4-dependent inflammation promotes lung tumour growth. Nat Commun. 6:70642015. View Article : Google Scholar : PubMed/NCBI

31 

Houghton AM, Rzymkiewicz DM, Ji H, Gregory AD, Egea EE, Metz HE, Stolz DB, Land SR, Marconcini LA, Kliment CR, et al: Neutrophil elastase-mediated degradation of IRS-1 accelerates lung tumor growth. Nat Med. 16:219–223. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Gong L, Cumpian AM, Caetano MS, Ochoa CE, De la Garza MM, Lapid DJ, Mirabolfathinejad SG, Dickey BF, Zhou Q and Moghaddam SJ: Promoting effect of neutrophils on lung tumorigenesis is mediated by CXCR2 and neutrophil elastase. Mol Cancer. 12:1542013. View Article : Google Scholar : PubMed/NCBI

33 

Hattar K, Franz K, Ludwig M, Sibelius U, Wilhelm J, Lohmeyer J, Savai R, Subtil FS, Dahlem G, Eul B, et al: Interactions between neutrophils and non-small cell lung cancer cells: Enhancement of tumor proliferation and inflammatory mediator synthesis. Cancer Immunol Immunother. 63:1297–1306. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Ma X, Aoki T, Tsuruyama T and Narumiya S: Definition of prostaglandin E2-EP2 signals in the colon tumor microenvironment that amplify inflammation and tumor growth. Cancer Res. 75:2822–2832. 2015. View Article : Google Scholar : PubMed/NCBI

35 

Antonio N, Bønnelykke-Behrndtz ML, Ward LC, Collin J, Christensen IJ, Steiniche T, Schmidt H, Feng Y and Martin P: The wound inflammatory response exacerbates growth of pre-neoplastic cells and progression to cancer. EMBO J. 34:2219–2236. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Liang J, Piao Y, Holmes L, Fuller GN, Henry V, Tiao N and de Groot JF: Neutrophils promote the malignant glioma phenotype through S100A4. Clin Cancer Res. 20:187–198. 2014. View Article : Google Scholar

37 

Song W, Li L, He D, Xie H, Chen J, Yeh CR, Chang LS, Yeh S and Chang C: Infiltrating neutrophils promote renal cell carcinoma (RCC) proliferation via modulating androgen receptor (AR) → c-Myc signals. Cancer Lett. 368:71–78. 2015. View Article : Google Scholar : PubMed/NCBI

38 

Grégoire M, Guilloton F, Pangault C, Mourcin F, Sok P, Latour M, Amé-Thomas P, Flecher E, Fest T and Tarte K: Neutrophils trigger a NF-κB dependent polarization of tumor-supportive stromal cells in germinal center B-cell lymphomas. Oncotarget. 6:16471–16487. 2015. View Article : Google Scholar

39 

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

40 

Liang W and Ferrara N: The complex role of neutrophils in tumor angiogenesis and metastasis. Cancer Immunol Res. 4:83–91. 2016. View Article : Google Scholar : PubMed/NCBI

41 

Psaila B and Lyden D: The metastatic niche: Adapting the foreign soil. Nat Rev Cancer. 9:285–293. 2009. View Article : Google Scholar : PubMed/NCBI

42 

Tazawa H, Okada F, Kobayashi T, Tada M, Mori Y, Une Y, Sendo F, Kobayashi M and Hosokawa M: Infiltration of neutrophils is required for acquisition of metastatic phenotype of benign murine fibrosarcoma cells: Implication of inflammation-associated carcinogenesis and tumor progression. Am J Pathol. 163:2221–2232. 2003. View Article : Google Scholar : PubMed/NCBI

43 

Welch DR, Schissel DJ, Howrey RP and Aeed PA: Tumor-elicited polymorphonuclear cells, in contrast to ‘normal’ circulating poly-morphonuclear cells, stimulate invasive and metastatic potentials of rat mammary adenocarcinoma cells. Proc Natl Acad Sci USA. 86:5859–5863. 1989. View Article : Google Scholar

44 

El Rayes T, Catena R, Lee S, Stawowczyk M, Joshi N, Fischbach C, Powell CA, Dannenberg AJ, Altorki NK, Gao D, et al: Lung inflammation promotes metastasis through neutrophil protease-mediated degradation of Tsp-1. Proc Natl Acad Sci USA. 112:16000–16005. 2015. View Article : Google Scholar : PubMed/NCBI

45 

Queen MM, Ryan RE, Holzer RG, Keller-Peck CR and Jorcyk CL: Breast cancer cells stimulate neutrophils to produce oncostatin M: Potential implications for tumor progression. Cancer Res. 65:8896–8904. 2005. View Article : Google Scholar : PubMed/NCBI

46 

Wu Y, Zhao Q, Peng C, Sun L, Li XF and Kuang DM: Neutrophils promote motility of cancer cells via a hyaluronan-mediated TLR4/PI3K activation loop. J Pathol. 225:438–447. 2011. View Article : Google Scholar : PubMed/NCBI

47 

Dumitru CA, Gholaman H, Trellakis S, Bruderek K, Dominas N, Gu X, Bankfalvi A, Whiteside TL, Lang S and Brandau S: Tumor-derived macrophage migration inhibitory factor modulates the biology of head and neck cancer cells via neutrophil activation. Int J Cancer. 129:859–869. 2011. View Article : Google Scholar : PubMed/NCBI

48 

Kowanetz M, Wu X, Lee J, Tan M, Hagenbeek T, Qu X, Yu L, Ross J, Korsisaari N, Cao T, et al: Granulocyte-colony stimulating factor promotes lung metastasis through mobilization of Ly6G+Ly6C+ granulocytes. Proc Natl Acad Sci USA. 107:21248–21255. 2010. View Article : Google Scholar : PubMed/NCBI

49 

Gaida MM, Steffen TG, Günther F, Tschaharganeh DF, Felix K, Bergmann F, Schirmacher P and Hänsch GM: Polymorphonuclear neutrophils promote dyshesion of tumor cells and elastase-mediated degradation of E-cadherin in pancreatic tumors. Eur J Immunol. 42:3369–3380. 2012. View Article : Google Scholar : PubMed/NCBI

50 

Grosse-Steffen T, Giese T, Giese N, Longerich T, Schirmacher P, Hänsch GM and Gaida MM: Epithelial-to-mesenchymal transition in pancreatic ductal adenocarcinoma and pancreatic tumor cell lines: The role of neutrophils and neutrophil-derived elastase. Clin Dev Immunol. 2012:7207682012. View Article : Google Scholar : PubMed/NCBI

51 

Freisinger CM and Huttenlocher A: Live imaging and gene expression analysis in zebrafish identifies a link between neutrophils and epithelial to mesenchymal transition. PLoS One. 9:e1121832014. View Article : Google Scholar : PubMed/NCBI

52 

Hu P, Shen M, Zhang P, Zheng C, Pang Z, Zhu L and Du J: Intratumoral neutrophil granulocytes contribute to epithelial-mesenchymal transition in lung adenocarcinoma cells. Tumour Biol. 36:7789–7796. 2015. View Article : Google Scholar : PubMed/NCBI

53 

Lin C, Lin W, Yeh S, Li L and Chang C: Infiltrating neutrophils increase bladder cancer cell invasion via modulation of androgen receptor (AR)/MMP13 signals. Oncotarget. 6:43081–43089. 2015.PubMed/NCBI

54 

Song W, Yeh CR, He D, Wang Y, Xie H, Pang ST, Chang LS, Li L and Yeh S: Infiltrating neutrophils promote renal cell carcinoma progression via VEGFa/HIF2α and estrogen receptor β signals. Oncotarget. 6:19290–19304. 2015. View Article : Google Scholar : PubMed/NCBI

55 

Coffelt SB, Kersten K, Doornebal CW, Weiden J, Vrijland K, Hau CS, Verstegen NJ, Ciampricotti M, Hawinkels LJ, Jonkers J, et al: IL-17-producing γδ T cells and neutrophils conspire to promote breast cancer metastasis. Nature. 522:345–348. 2015. View Article : Google Scholar : PubMed/NCBI

56 

Wu QD, Wang JH, Condron C, Bouchier-Hayes D and Redmond HP: Human neutrophils facilitate tumor cell transendothelial migration. Am J Physiol Cell Physiol. 280:C814–C822. 2001.PubMed/NCBI

57 

Strell C, Lang K, Niggemann B, Zaenker KS and Entschladen F: Neutrophil granulocytes promote the migratory activity of MDA-MB-468 human breast carcinoma cells via ICAM-1. Exp Cell Res. 316:138–148. 2010. View Article : Google Scholar

58 

Huh SJ, Liang S, Sharma A, Dong C and Robertson GP: Transiently entrapped circulating tumor cells interact with neutrophils to facilitate lung metastasis development. Cancer Res. 70:6071–6082. 2010. View Article : Google Scholar : PubMed/NCBI

59 

Spicer JD, McDonald B, Cools-Lartigue JJ, Chow SC, Giannias B, Kubes P and Ferri LE: Neutrophils promote liver metastasis via Mac-1-mediated interactions with circulating tumor cells. Cancer Res. 72:3919–3927. 2012. View Article : Google Scholar : PubMed/NCBI

60 

Tabariès S, Ouellet V, Hsu BE, Annis MG, Rose AA, Meunier L, Carmona E, Tam CE, Mes-Masson AM and Siegel PM: Granulocytic immune infiltrates are essential for the efficient formation of breast cancer liver metastases. Breast Cancer Res. 17:452015. View Article : Google Scholar : PubMed/NCBI

61 

Tazzyman S, Lewis CE and Murdoch C: Neutrophils: Key mediators of tumour angiogenesis. Int J Exp Pathol. 90:222–231. 2009. View Article : Google Scholar : PubMed/NCBI

62 

Tazzyman S, Niaz H and Murdoch C: Neutrophil-mediated tumour angiogenesis: Subversion of immune responses to promote tumour growth. Semin Cancer Biol. 23:149–158. 2013. View Article : Google Scholar : PubMed/NCBI

63 

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

64 

Nozawa H, Chiu C and Hanahan D: Infiltrating neutrophils mediate the initial angiogenic switch in a mouse model of multistage carcinogenesis. Proc Natl Acad Sci USA. 103:12493–12498. 2006. View Article : Google Scholar : PubMed/NCBI

65 

Deryugina EI, Zajac E, Juncker-Jensen A, Kupriyanova TA, Welter L and Quigley JP: Tissue-infiltrating neutrophils constitute the major in vivo source of angiogenesis-inducing MMP-9 in the tumor microenvironment. Neoplasia. 16:771–788. 2014. View Article : Google Scholar : PubMed/NCBI

66 

Kuang DM, Zhao Q, Wu Y, Peng C, Wang J, Xu Z, Yin XY and Zheng L: Peritumoral neutrophils link inflammatory response to disease progression by fostering angiogenesis in hepatocellular carcinoma. J Hepatol. 54:948–955. 2011. View Article : Google Scholar

67 

Ardi VC, Kupriyanova TA, Deryugina EI and Quigley JP: Human neutrophils uniquely release TIMP-free MMP-9 to provide a potent catalytic stimulator of angiogenesis. Proc Natl Acad Sci USA. 104:20262–20267. 2007. View Article : Google Scholar : PubMed/NCBI

68 

Bekes EM, Schweighofer B, Kupriyanova TA, Zajac E, Ardi VC, Quigley JP and Deryugina EI: Tumor-recruited neutrophils and neutrophil TIMP-free MMP-9 regulate coordinately the levels of tumor angiogenesis and efficiency of malignant cell intravasation. Am J Pathol. 179:1455–1470. 2011. View Article : Google Scholar : PubMed/NCBI

69 

Rodriguez PC, Ernstoff MS, Hernandez C, Atkins M, Zabaleta J, Sierra R and Ochoa AC: Arginase I-producing myeloid-derived suppressor cells in renal cell carcinoma are a subpopulation of activated granulocytes. Cancer Res. 69:1553–1560. 2009. View Article : Google Scholar : PubMed/NCBI

70 

Rotondo R, Barisione G, Mastracci L, Grossi F, Orengo AM, Costa R, Truini M, Fabbi M, Ferrini S and Barbieri O: IL-8 induces exocytosis of arginase 1 by neutrophil polymorphonuclears in nonsmall cell lung cancer. Int J Cancer. 125:887–893. 2009. View Article : Google Scholar : PubMed/NCBI

71 

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

72 

Koyama S, Akbay EA, Li YY, Aref AR, Skoulidis F, Herter-Sprie GS, Buczkowski KA, Liu Y, Awad MM, Denning WL, et al: STK11/LKB1 deficiency promotes neutrophil recruitment and proinflammatory cytokine production to suppress T-cell activity in the lung tumor microenvironment. Cancer Res. 76:999–1008. 2016. View Article : Google Scholar : PubMed/NCBI

73 

Zhang J, Qiao X, Shi H, Han X, Liu W, Tian X and Zeng X: Circulating tumor-associated neutrophils (cTAN) contribute to circulating tumor cell survival by suppressing peripheral leukocyte activation. Tumour Biol. 37:5397–5404. 2016. View Article : Google Scholar

74 

Brandau S, Trellakis S, Bruderek K, Schmaltz D, Steller G, Elian M, Suttmann H, Schenck M, Welling J, Zabel P, et al: Myeloid-derived suppressor cells in the peripheral blood of cancer patients contain a subset of immature neutrophils with impaired migratory properties. J Leukoc Biol. 89:311–317. 2011. View Article : Google Scholar

75 

Fridlender ZG, Sun J, Mishalian I, Singhal S, Cheng G, Kapoor V, Horng W, Fridlender G, Bayuh R, Worthen GS, et al: Transcriptomic analysis comparing tumor-associated neutrophils with granulocytic myeloid-derived suppressor cells and normal neutrophils. PLoS One. 7:e315242012. View Article : Google Scholar : PubMed/NCBI

76 

Sceneay J, Chow MT, Chen A, Halse HM, Wong CS, Andrews DM, Sloan EK, Parker BS, Bowtell DD, Smyth MJ, et al: Primary tumor hypoxia recruits CD11b+/Ly6Cmed/Ly6G+ immune suppressor cells and compromises NK cell cytotoxicity in the premetastatic niche. Cancer Res. 72:3906–3911. 2012. View Article : Google Scholar : PubMed/NCBI

77 

Spiegel A, Brooks MW, Houshyar S, Reinhardt F, Ardolino M, Fessler E, Chen MB, Krall JA, DeCock J, Zervantonakis IK, et al: Neutrophils suppress intraluminal NK-cell mediated tumor cell clearance and enhance extravasation of disseminated carcinoma cells. Cancer Discov. 6:630–649. 2016. View Article : Google Scholar : PubMed/NCBI

78 

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

79 

Zhou SL, Zhou ZJ, Hu ZQ, Huang XW, Wang Z, Chen EB, Fan J, Cao Y, Dai Z and Zhou J: Tumor-associated neutrophils recruit macrophages and T-regulatory cells to promote progression of hepatocellular carcinoma and resistance to sorafenib. Gastroenterology. S0016-5085(16)00231-6. 2016.

80 

Casbon AJ, Reynaud D, Park C, Khuc E, Gan DD, Schepers K, Passegué E and Werb Z: Invasive breast cancer reprograms early myeloid differentiation in the bone marrow to generate immunosuppressive neutrophils. Proc Natl Acad Sci USA. 112:E566–E575. 2015. View Article : Google Scholar : PubMed/NCBI

81 

Branzk N and Papayannopoulos V: Molecular mechanisms regulating NETosis in infection and disease. Semin Immunopathol. 35:513–530. 2013. View Article : Google Scholar : PubMed/NCBI

82 

Demers M and Wagner DD: NETosis: A new factor in tumor progression and cancer-associated thrombosis. Semin Thromb Hemost. 40:277–283. 2014. View Article : Google Scholar : PubMed/NCBI

83 

Cools-Lartigue J, Spicer J, Najmeh S and Ferri L: Neutrophil extracellular traps in cancer progression. Cell Mol Life Sci. 71:4179–4194. 2014. View Article : Google Scholar : PubMed/NCBI

84 

Fuchs TA, Brill A, Duerschmied D, Schatzberg D, Monestier M, Myers DD Jr, Wrobleski SK, Wakefield TW, Hartwig JH and Wagner DD: Extracellular DNA traps promote thrombosis. Proc Natl Acad Sci USA. 107:15880–15885. 2010. View Article : Google Scholar : PubMed/NCBI

85 

Demers M, Krause DS, Schatzberg D, Martinod K, Voorhees JR, Fuchs TA, Scadden DT and Wagner DD: Cancers predispose neutrophils to release extracellular DNA traps that contribute to cancer-associated thrombosis. Proc Natl Acad Sci USA. 109:13076–13081. 2012. View Article : Google Scholar : PubMed/NCBI

86 

Demers M and Wagner DD: Neutrophil extracellular traps: A new link to cancer-associated thrombosis and potential implications for tumor progression. OncoImmunology. 2:e229462013. View Article : Google Scholar : PubMed/NCBI

87 

Cools-Lartigue J, Spicer J, McDonald B, Gowing S, Chow S, Giannias B, Bourdeau F, Kubes P and Ferri L: Neutrophil extracellular traps sequester circulating tumor cells and promote metastasis. J Clin Invest. 674842013.PubMed/NCBI

88 

Cedervall J, Zhang Y, Huang H, Zhang L, Femel J, Dimberg A and Olsson AK: Neutrophil extracellular traps accumulate in peripheral blood vessels and compromise organ function in tumor-bearing animals. Cancer Res. 75:2653–2662. 2015. View Article : Google Scholar : PubMed/NCBI

89 

Guglietta S, Chiavelli A, Zagato E, Krieg C, Gandini S, Ravenda PS, Bazolli B, Lu B, Penna G and Rescigno M: Coagulation induced by C3aR-dependent NETosis drives protumorigenic neutrophils during small intestinal tumorigenesis. Nat Commun. 7:110372016. View Article : Google Scholar : PubMed/NCBI

90 

Tohme S, Yazdani HO, Al-Khafaji AB, Chidi AP, Loughran P, Mowen K, Wang Y, Simmons RL, Huang H and Tsung A: Neutrophil extracellular traps promote the development and progression of liver metastases after surgical stress. Cancer Res. 76:1367–1380. 2016. View Article : Google Scholar : PubMed/NCBI

91 

Cortez-Retamozo V, Etzrodt M, Newton A, Rauch PJ, Chudnovskiy A, Berger C, Ryan RJ, Iwamoto Y, Marinelli B, Gorbatov R, et al: Origins of tumor-associated macrophages and neutrophils. Proc Natl Acad Sci USA. 109:2491–2496. 2012. View Article : Google Scholar : PubMed/NCBI

92 

De Larco JE, Wuertz BR and Furcht LT: The potential role of neutrophils in promoting the metastatic phenotype of tumors releasing interleukin-8. Clin Cancer Res. 10:4895–4900. 2004. View Article : Google Scholar : PubMed/NCBI

93 

Wu P, Wu D, Ni C, Ye J, Chen W, Hu G, Wang Z, Wang C, Zhang Z, Xia W, et al: γδT17 cells promote the accumulation and expansion of myeloid-derived suppressor cells in human colorectal cancer. Immunity. 40:785–800. 2014. View Article : Google Scholar : PubMed/NCBI

94 

Raccosta L, Fontana R, Maggioni D, Lanterna C, Villablanca EJ, Paniccia A, Musumeci A, Chiricozzi E, Trincavelli ML, Daniele S, et al: The oxysterol-CXCR2 axis plays a key role in the recruitment of tumor-promoting neutrophils. J Exp Med. 210:1711–1728. 2013. View Article : Google Scholar : PubMed/NCBI

95 

Raccosta L, Fontana R, Traversari C and Russo V: Oxysterols recruit tumor-supporting neutrophils within the tumor micro-environment: The many facets of tumor-derived oxysterols. OncoImmunology. 2:e264692013. View Article : Google Scholar

96 

Zhou SL, Dai Z, Zhou ZJ, Wang XY, Yang GH, Wang Z, Huang XW, Fan J and Zhou J: Overexpression of CXCL5 mediates neutrophil infiltration and indicates poor prognosis for hepatocellular carcinoma. Hepatology. 56:2242–2254. 2012. View Article : Google Scholar : PubMed/NCBI

97 

Zhou SL, Dai Z, Zhou ZJ, Chen Q, Wang Z, Xiao YS, Hu ZQ, Huang XY, Yang GH, Shi YH, et al: CXCL5 contributes to tumor metastasis and recurrence of intrahepatic cholangio-carcinoma by recruiting infiltrative intratumoral neutrophils. Carcinogenesis. 35:597–605. 2014. View Article : Google Scholar

98 

Bald T, Quast T, Landsberg J, Rogava M, Glodde N, Lopez-Ramos D, Kohlmeyer J, Riesenberg S, van den Boorn-Konijnenberg D, Hömig-Hölzel C, et al: Ultraviolet-radiation-induced inflammation promotes angiotropism and metastasis in melanoma. Nature. 507:109–113. 2014. View Article : Google Scholar : PubMed/NCBI

99 

Trellakis S, Farjah H, Bruderek K, Dumitru CA, Hoffmann TK, Lang S and Brandau S: Peripheral blood neutrophil granulocytes from patients with head and neck squamous cell carcinoma functionally differ from their counterparts in healthy donors. Int J Immunopathol Pharmacol. 24:683–693. 2011.PubMed/NCBI

100 

Walmsley SR, Print C, Farahi N, Peyssonnaux C, Johnson RS, Cramer T, Sobolewski A, Condliffe AM, Cowburn AS, Johnson N, et al: Hypoxia-induced neutrophil survival is mediated by HIF-1alpha-dependent NF-kappaB activity. J Exp Med. 201:105–115. 2005. View Article : Google Scholar : PubMed/NCBI

101 

Li XF, Chen DP, Ouyang FZ, Chen MM, Wu Y, Kuang DM and Zheng L: Increased autophagy sustains the survival and pro-tumourigenic effects of neutrophils in human hepatocellular carcinoma. J Hepatol. 62:131–139. 2015. View Article : Google Scholar

102 

Andzinski L, Wu CF, Lienenklaus S, Kröger A, Weiss S and Jablonska J: Delayed apoptosis of tumor associated neutrophils in the absence of endogenous IFN-β. Int J Cancer. 136:572–583. 2015.

103 

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

104 

Finisguerra V, Di Conza G, Di Matteo M, Serneels J, Costa S, Thompson AA, Wauters E, Walmsley S, Prenen H, Granot Z, et al: MET is required for the recruitment of anti-tumoural neutrophils. Nature. 522:349–353. 2015. View Article : Google Scholar : PubMed/NCBI

105 

Galli SJ, Borregaard N and Wynn TA: Phenotypic and functional plasticity of cells of innate immunity: Macrophages, mast cells and neutrophils. Nat Immunol. 12:1035–1044. 2011. View Article : Google Scholar : PubMed/NCBI

106 

Gabrilovich DI, Ostrand-Rosenberg S and Bronte V: Coordinated regulation of myeloid cells by tumours. Nat Rev Immunol. 12:253–268. 2012. View Article : Google Scholar : PubMed/NCBI

107 

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

108 

Sagiv JY, Michaeli J, Assi S, Mishalian I, Kisos H, Levy L, Damti P, Lumbroso D, Polyansky L, Sionov RV, et al: Phenotypic diversity and plasticity in circulating neutrophil subpopulations in cancer. Cell Rep. 10:562–573. 2015. View Article : Google Scholar : PubMed/NCBI

109 

Mishalian I, Bayuh R, Levy L, Zolotarov L, Michaeli J and Fridlender ZG: Tumor-associated neutrophils (TAN) develop pro-tumorigenic properties during tumor progression. Cancer Immunol Immunother. 62:1745–1756. 2013. View Article : Google Scholar : PubMed/NCBI

110 

Yan B, Wei JJ, Yuan Y, Sun R, Li D, Luo J, Liao SJ, Zhou YH, Shu Y, Wang Q, et al: IL-6 cooperates with G-CSF to induce protumor function of neutrophils in bone marrow by enhancing STAT3 activation. J Immunol. 190:5882–5893. 2013. View Article : Google Scholar : PubMed/NCBI

111 

Zhu Q, Zhang X, Zhang L, Li W, Wu H, Yuan X, Mao F, Wang M, Zhu W, Qian H, et al: The IL-6-STAT3 axis mediates a reciprocal crosstalk between cancer-derived mesenchymal stem cells and neutrophils to synergistically prompt gastric cancer progression. Cell Death Dis. 5:e12952014. View Article : Google Scholar : PubMed/NCBI

112 

Hu X, Zhou Y, Dong K, Sun Z, Zhao D, Wang W, Yu G, Liu W, Xu G, Han Z, et al: Programming of the development of tumor-promoting neutrophils by mesenchymal stromal cells. Cell Physiol Biochem. 33:1802–1814. 2014. View Article : Google Scholar : PubMed/NCBI

113 

Jensen HK, Donskov F, Marcussen N, Nordsmark M, Lundbeck F and von der Maase H: Presence of intratumoral neutrophils is an independent prognostic factor in localized renal cell carcinoma. J Clin Oncol. 27:4709–4717. 2009. View Article : Google Scholar : PubMed/NCBI

114 

Trellakis S, Bruderek K, Dumitru CA, Gholaman H, Gu X, Bankfalvi A, Scherag A, Hütte J, Dominas N, Lehnerdt GF, et al: Polymorphonuclear granulocytes in human head and neck cancer: Enhanced inflammatory activity, modulation by cancer cells and expansion in advanced disease. Int J Cancer. 129:2183–2193. 2011. View Article : Google Scholar

115 

Jensen TO, Schmidt H, Møller HJ, Donskov F, Høyer M, Sjoegren P, Christensen IJ and Steiniche T: Intratumoral neutrophils and plasmacytoid dendritic cells indicate poor prognosis and are associated with pSTAT3 expression in AJCC stage I/II melanoma. Cancer. 118:2476–2485. 2012. View Article : Google Scholar

116 

Chen X, Sun J, Song Y, Gao P, Zhao J, Huang X, Liu B, Xu H and Wang Z: The novel long noncoding RNA AC138128.1 may be a predictive biomarker in gastric cancer. Med Oncol. 31:2622014. View Article : Google Scholar : PubMed/NCBI

117 

Yang SZ, Ji WH, Mao WM and Ling ZQ: Elevated levels of preoperative circulating CD44+ lymphocytes and neutrophils predict poor survival for non-small cell lung cancer patients. Clin Chim Acta. 439:172–177. 2015. View Article : Google Scholar

118 

Rao HL, Chen JW, Li M, Xiao YB, Fu J, Zeng YX, Cai MY and Xie D: Increased intratumoral neutrophil in colorectal carcinomas correlates closely with malignant phenotype and predicts patients’ adverse prognosis. PLoS One. 7:e308062012. View Article : Google Scholar

119 

Zhao JJ, Pan K, Wang W, Chen JG, Wu YH, Lv L, Li JJ, Chen YB, Wang DD, Pan QZ, et al: The prognostic value of tumor-infiltrating neutrophils in gastric adenocarcinoma after resection. PLoS One. 7:e336552012. View Article : Google Scholar : PubMed/NCBI

120 

Li YW, Qiu SJ, Fan J, Zhou J, Gao Q, Xiao YS and Xu YF: Intra-tumoral neutrophils: A poor prognostic factor for hepatocellular carcinoma following resection. J Hepatol. 54:497–505. 2011. View Article : Google Scholar

121 

Wang N, Feng Y, Wang Q, Liu S, Xiang L, Sun M, Zhang X, Liu G, Qu X and Wei F: Neutrophils infiltration in the tongue squamous cell carcinoma and its correlation with CEACAM1 expression on tumor cells. PLoS One. 9:e899912014. View Article : Google Scholar : PubMed/NCBI

122 

Hu P, Pang Z, Shen H, Wang G, Sun H and Du J: Tumor-infiltrating neutrophils predict poor outcome in adenocarcinoma of the esophagogastric junction. Tumour Biol. 36:2965–2971. 2015. View Article : Google Scholar

123 

Wang J, Jia Y, Wang N, Zhang X, Tan B, Zhang G and Cheng Y: The clinical significance of tumor-infiltrating neutrophils and neutrophil-to-CD8+ lymphocyte ratio in patients with resectable esophageal squamous cell carcinoma. J Transl Med. 12:72014. View Article : Google Scholar

124 

Shen M, Hu P, Donskov F, Wang G, Liu Q and Du J: Tumor-associated neutrophils as a new prognostic factor in cancer: A systematic review and meta-analysis. PLoS One. 9:e982592014. View Article : Google Scholar : PubMed/NCBI

125 

Templeton AJ, McNamara MG, Šeruga B, Vera-Badillo FE, Aneja P, Ocaña A, Leibowitz-Amit R, Sonpavde G, Knox JJ, Tran B, et al: Prognostic role of neutrophil-to-lymphocyte ratio in solid tumors: A systematic review and meta-analysis. J Natl Cancer Inst. 106:dju1242014. View Article : Google Scholar : PubMed/NCBI

126 

Ferrucci PF, Gandini S, Battaglia A, Alfieri S, Di Giacomo AM, Giannarelli D, Cappellini GC, De Galitiis F, Marchetti P, Amato G, et al: Baseline neutrophil-to-lymphocyte ratio is associated with outcome of ipilimumab-treated metastatic melanoma patients. Br J Cancer. 112:1904–1910. 2015. View Article : Google Scholar : PubMed/NCBI

127 

Gregory AD and Houghton AM: Tumor-associated neutrophils: New targets for cancer therapy. Cancer Res. 71:2411–2416. 2011. View Article : Google Scholar : PubMed/NCBI

128 

Sun R, Luo J, Li D, Shu Y, Luo C, Wang SS, Qin J, Zhang GM and Feng ZH: Neutrophils with protumor potential could efficiently suppress tumor growth after cytokine priming and in presence of normal NK cells. Oncotarget. 5:12621–12634. 2014. View Article : Google Scholar

129 

Pang Y, Gara SK, Achyut BR, Li Z, Yan HH, Day CP, Weiss JM, Trinchieri G, Morris JC and Yang L: TGF-β signaling in myeloid cells is required for tumor metastasis. Cancer Discov. 3:936–951. 2013. View Article : Google Scholar : PubMed/NCBI

130 

Andzinski L, Kasnitz N, Stahnke S, Wu CF, Gereke M, von Köckritz-Blickwede M, Schilling B, Brandau S, Weiss S and Jablonska J: Type I IFNs induce anti-tumor polarization of tumor associated neutrophils in mice and human. Int J Cancer. 138:1982–1993. 2016. View Article : Google Scholar

131 

Jamieson T, Clarke M, Steele CW, Samuel MS, Neumann J, Jung A, Huels D, Olson MF, Das S, Nibbs RJ, et al: Inhibition of CXCR2 profoundly suppresses inflammation-driven and spontaneous tumorigenesis. J Clin Invest. 122:3127–3144. 2012. View Article : Google Scholar : PubMed/NCBI

132 

Tazzyman S, Barry ST, Ashton S, Wood P, Blakey D, Lewis CE and Murdoch C: Inhibition of neutrophil infiltration into A549 lung tumors in vitro and in vivo using a CXCR2-specific antagonist is associated with reduced tumor growth. Int J Cancer. 129:847–858. 2011. View Article : Google Scholar : PubMed/NCBI

133 

Wculek SK and Malanchi I: Neutrophils support lung colonization of metastasis-initiating breast cancer cells. Nature. 528:413–417. 2015. View Article : Google Scholar : PubMed/NCBI

134 

Shrestha S, Noh JM, Kim SY, Ham HY, Kim YJ, Yun YJ, Kim MJ, Kwon MS, Song DK and Hong CW: Angiotensin converting enzyme inhibitors and angiotensin II receptor antagonist attenuate tumor growth via polarization of neutrophils toward an antitumor phenotype. OncoImmunology. 5:e10677442015. View Article : Google Scholar

135 

Hagerling C and Werb Z: Neutrophils: Critical components in experimental animal models of cancer. Semin Immunol. 28:197–204. 2016. View Article : Google Scholar : PubMed/NCBI

136 

Zhou SL, Zhou ZJ, Hu ZQ, Li X, Huang XW, Wang Z, Fan J, Dai Z and Zhou J: CXCR2/CXCL5 axis contributes to epithelial-mesenchymal transition of HCC cells through activating PI3K/ Akt/GSK-3β/Snail signaling. Cancer Lett. 358:124–135. 2015. View Article : Google Scholar

137 

Benevides L, da Fonseca DM, Donate PB, Tiezzi DG, De Carvalho DD, de Andrade JM, Martins GA and Silva JS: IL17 Promotes mammary tumor progression by changing the behavior of tumor cells and eliciting tumorigenic neutrophils recruitment. Cancer Res. 75:3788–3799. 2015. View Article : Google Scholar : PubMed/NCBI

138 

Wislez M, Rabbe N, Marchal J, Milleron B, Crestani B, Mayaud C, Antoine M, Soler P and Cadranel J: Hepatocyte growth factor production by neutrophils infiltrating bronchioloalveolar subtype pulmonary adenocarcinoma: Role in tumor progression and death. Cancer Res. 63:1405–1412. 2003.PubMed/NCBI

139 

Ibrahim SA, Katara GK, Kulshrestha A, Jaiswal MK, Amin MA and Beaman KD: Breast cancer associated a2 isoform vacuolar ATPase immunomodulates neutrophils: Potential role in tumor progression. Oncotarget. 6:33033–33045. 2015.PubMed/NCBI

Related Articles

Journal Cover

September-2016
Volume 49 Issue 3

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang X, Zhang W, Yuan X, Fu M, Qian H and Xu W: Neutrophils in cancer development and progression: Roles, mechanisms, and implications (Review). Int J Oncol 49: 857-867, 2016
APA
Zhang, X., Zhang, W., Yuan, X., Fu, M., Qian, H., & Xu, W. (2016). Neutrophils in cancer development and progression: Roles, mechanisms, and implications (Review). International Journal of Oncology, 49, 857-867. https://doi.org/10.3892/ijo.2016.3616
MLA
Zhang, X., Zhang, W., Yuan, X., Fu, M., Qian, H., Xu, W."Neutrophils in cancer development and progression: Roles, mechanisms, and implications (Review)". International Journal of Oncology 49.3 (2016): 857-867.
Chicago
Zhang, X., Zhang, W., Yuan, X., Fu, M., Qian, H., Xu, W."Neutrophils in cancer development and progression: Roles, mechanisms, and implications (Review)". International Journal of Oncology 49, no. 3 (2016): 857-867. https://doi.org/10.3892/ijo.2016.3616